WO2015163711A1 - Talen targeting myostatin gene and method for making animal with knockout myostatin gene using same - Google Patents

Talen targeting myostatin gene and method for making animal with knockout myostatin gene using same Download PDF

Info

Publication number
WO2015163711A1
WO2015163711A1 PCT/KR2015/004068 KR2015004068W WO2015163711A1 WO 2015163711 A1 WO2015163711 A1 WO 2015163711A1 KR 2015004068 W KR2015004068 W KR 2015004068W WO 2015163711 A1 WO2015163711 A1 WO 2015163711A1
Authority
WO
WIPO (PCT)
Prior art keywords
myostatin gene
myostatin
gene
sequence
animal
Prior art date
Application number
PCT/KR2015/004068
Other languages
French (fr)
Korean (ko)
Inventor
윤희준
김석중
김진수
강금단
Original Assignee
윤희준
주식회사 툴젠
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 윤희준, 주식회사 툴젠 filed Critical 윤희준
Publication of WO2015163711A1 publication Critical patent/WO2015163711A1/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
    • 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
    • A01K67/0276Knockout animals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0656Adult fibroblasts
    • 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
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • 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

Definitions

  • Myostatin is a protein belonging to the TGF- ⁇ family and is a regulatory factor that inhibits muscle differentiation and growth during muscle differentiation and is also known as growth differentiation factor 8 (GDF-8). .
  • Myostatin is produced primarily from bone cells and circulates through the blood to act on muscle tissue. Therefore, if there is no myostatin gene in animals or inhibits the activity of myostatin, it can be expected to increase muscle.
  • Humans and cattle (Ravi ambadur et al., Genome Res., 7: 910- 915, 2007), sheep and dogs (Dana S. Mosher et al., Plos genet., 3: 779-786, 2007) have identified a myostatin mutation present in nature.
  • muscle phenotype was increased compared to wild-type.
  • mice knocked out the myostatin gene were made, a small amount of muscle was increased when the gene knockout was a heterozygote and a double-muscle when the knockout was a homozygote. Muscle growth was observed (Se-Jin Lee et al. Nature, 16; 98: 9306-9311, 1997).
  • the present inventors have tried to find a method for producing a transgenic pig knocked out of the myostatin gene. As a result, the present inventors have completed the present invention by confirming that, when using TALEN and somatic cell nuclear transfer technology specific to myostatin gene, it is possible to effectively make a transgenic pig knocked out of myostatin gene.
  • Another object of the present invention is to provide a recombinant expression vector for myostatin gene knockout using TALEN.
  • Another object of the present invention to provide a method for producing a cell knocked out myostatin gene using TALEN and to provide a cell knocked out myostatin gene prepared using the same. Another object of the present invention to provide a method for producing a transgenic animal other than a human knocked out myostatin gene using TALEN and a transgenic animal other than a human knocked out myostatin gene prepared using the same There is.
  • a fusion protein having the activity of binding to and cleaving a specific sequence of a myostatin (MSTN) gene:
  • statin Maio in the present invention is known as GDF8 (. Growth differentiation factor 8), is known as a gene that suppresses muscle growth and differentiation ol.
  • Myostatin is a regulatory ligand belonging to the TGF- ⁇ superfamily and its gene structure is similar to other genes belonging to the TGF- ⁇ superfamily.
  • Myostatin gene sequence and the like can be obtained from a known database (GenBank et al.).
  • Myostatin acts as a negative regulator involved in the development, growth, and regulation of muscle homeostasis in the musculoskeletal structure, so if the animal lacks the myostatin gene or inhibits the activity of myostatin, muscle can be expected to increase.
  • Myostatin mutations in nature have been identified in cattle, sheep and dogs, and the deletion of the myostatin gene showed an increased phenotype compared to the wild-type.
  • mice knocked out the myostatin gene were made, a small amount of muscle was increased when the gene knockout was a heterozygote, and a double-muscle when the knockout was a homozygote. It was observed that the muscles are greatly increased.
  • the myostatin gene can be effectively knocked out in an animal that supplies meat to humans, the production of an animal with increased muscles can be expected. For example, to produce pigs with increased muscle mass, they tried to knock out the myostatin gene in pigs.
  • the term "having the activity of binding to and cleaving a specific sequence of myostatin gene” means that the specific nucleic acid sequence of the myostatin gene is specifically recognized and cleaved. Recognizing a specific nucleic acid sequence of the ostatin specifically, it means that the cleavage is generated in the myostatin gene by the nucleotide cleavage domain connected by the TALE domain and the peptide bond.
  • sequence means a nucleotide sequence regardless of its length, which may be linear, circular or branched DNA or RNA, and may be single helical or double helical.
  • TALE domains can be designed for all specific sequences, but experimentally, the length of specific sequences to which TALE domains bind, the length of the spacer between two half-digits forming one target region, etc. This is known to affect the effective action of TALENs.
  • the half-digits are 15-20 bp in length
  • the spacers are 12-14 bp in length
  • the total sequence length is 42-54 bp
  • the starting sequence of the specific sequence is thymine (T) and the last sequence.
  • A adenine
  • specific positions should be selected so that TALENs can cleave myostatin genes specifically and effectively.
  • four specific sequences of the sequence of the pig myostatin gene using the database of TALEN cleavage ability for various specific sequences possessed by Bioinformatics Co., Ltd. SEQ ID NO: TALEN for 1 to 4 sequences
  • TALE Transcription Activator Like Effector
  • TALE domains of the invention refer to protein domains that bind to nucleotides in a sequence-specific manner by a combination of one or more TALE-repeat parents.
  • the TALE domain includes, but is not limited to, at least one TALE-repeats moiety, specifically 1-30 TALE-repeats moieties.
  • TALE-repeat parents may have the following general amino acid sequence as a site that recognizes a particular nucleotide sequence in the TALE domain:
  • XX refers to the super-modified amino acids at positions 12 and 13 to determine the specificity of base recognition. Specifically, the twelfth and thirteenth amino acids of the TALE-repeat parent recognize one specific nucleic acid.
  • the TALE-repeat modalities recognize C, T when XX is NG, A when XX is NI, and G when XX is NN, respectively, thus combining various TALE-repeat mods
  • a TALE domain capable of recognizing a specific position in the target gene can be prepared.
  • a method for preparing a specific TALE domain is described in detail in the Korean patent application 'Genome Engineering 1 through Designed TAL Effector Nuclease (Application No. 10-2013-7018743).
  • TALEN generally acts as a dimer, it is necessary to design two TALEN monomers to cut a specific position in the target gene (FIG. 1).
  • the specific position on the left to which the TALE domain binds is named as a first half-site and the specific position on the right as a second half-site based on the cleavage site.
  • Two TALEN monomers are each made up of two half-sites in different target genes.
  • One recognizes and the two half-digits are designed to be separated from each other by spacers of 9 to 14-bp. According to a specific example of the present invention, it is separated from each other by a 12-bp spacer and recognizes 21 bp (first half-digit, sequence listing first sequence) and 20 bp (second half-site, sequence listing second sequence), respectively.
  • the TALEN monomer was designed and used to effectively induce knockout of porcine myostatin gene.
  • cutting refers to unlinking the covalently bonded backbone of the myostatin nucleotide molecule, and the "nucleotide cleavage domain" 'is a polypeptide having enzymatic activity for nucleotide cutting of such myostatin. Refers to the sequence.
  • Nucleotide cleavage domains can be obtained from endonucleases or exonucleases. Examples of endonucleases from which nucleotide cleavage domains can be obtained include, but are not limited to, endogenous nucleases, recurrent endonucleases, and the like. It is not limited. Such enzymes can be used as the origin of the nucleotide cleavage domain. In addition, the nucleotide cleavage domain can cleave a single nucleotide sequence, as well as cleave a nucleotide sequence of a double bond, depending on the origin of the cleavage domain.
  • the restriction endonuclease is capable of sequence specific binding with DNA (recognition site), and can cleave DNA in the vicinity of the binding site.
  • Fokl a type lis enzyme, promotes double helix cleavage of DNA at 9 nucleotides from the recognition site in one helix and 13 nucleotides from the recognition site in the other helix.
  • Nucleotide cleavage domains of the invention may be derived from type lis restriction endonucleases, wherein the type lis restriction endonucleases are described, for example, in Fokl, Aarl, Acelll, Acil, AloI, ' BaeI, Bbr7I, Cdil, CjePI, Ecil, Esp3I, Finl, Mbol, sa I or SspD51 may be, but is not limited thereto.
  • the nucleotide cleavage domain of the present invention may be Fokl.
  • the fusion protein of the present invention may further comprise a nuclear localization signal sequence.
  • the nuclear position signal of the present invention TALENs targeting myostatin can be used as a means to move into the nucleus to knock out the myostatin gene.
  • a nucleotide sequence encoding the fusion protein and (b) provides a recombinant expression vector for knockout myostatin gene comprising a transcriptional regulatory sequence operably linked to the nucleotide sequence (operat i ve lyli nked).
  • the expression vector may be introduced into a cell to express a fusion protein (TALEN) of the present invention, and a known expression vector such as a plasmid vector, a cosmid vector, and a bacteriophage vector may be used. It can be easily prepared by those skilled in the art according to any known method.
  • TALEN fusion protein
  • opert i ve ly li nked refers to a functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and other nucleic acid sequences. Whereby the regulatory sequence controls transcription and / or translation of the other nucleic acid sequence.
  • the transcriptional regulatory sequence bound to the nucleotide sequence encoding the fusion protein is capable of controlling transcription of the nucleotide encoding the fusion protein by operating in an animal cell (eg, a mammalian cell), which is derived from a mammalian virus.
  • Promoters and promoters derived from genomes of mammalian cells such as, for example, cyt omega lo virus (CMV) promoter, adenovirus late promoter, vaccinia virus 7. 5K promoter, SV40 promoter, tk promoter of HSV, RSV Promoter, EF1 alpha promoter, metallothionine promoter, beta-actin promoter, promoter of human IL-2 gene, promoter of human IFN gene, promoter of human IL-4 gene, promoter of human lymphotoxin gene and human GM-CSF Including but not limited to promoters of genes.
  • the combinatorial expression vector of the present invention may comprise a polyaninated sequence.
  • a method of producing a cell knocked out myostatin gene comprising the following steps.
  • any method known in the art may be used to introduce a recombinant expression vector for myostatin gene knockout into a cell.
  • a recombinant expression vector for myostatin gene knockout was introduced into porcine fetal fibroblasts using electroporation.
  • the double-strand damage on the myostatin gene can be induced.
  • the cell repairs the damaged area using its own repair mechanism.
  • the cell repairs the damaged region by non-homologous end j oini ng (NHEJ) method, and insertion (i nser ti on) or deletion (de l et i on) at the end of the broken DNA strand is performed. Repairs are made in the direction of error-prone. Thus, mutations are induced in the myostatin gene, which results in knockout of the myostatin gene.
  • the mutation may be a mutation by substitution, deletion, insertion, and a combination thereof.
  • various mutations such as insertion and deletion were induced by the TALEN fusion protein, thereby successfully knocking out the myostatin gene.
  • a cell in which myostatin gene is knocked out by the above method is provided.
  • the cell may be a variety of animal cells, but for the purposes of the present invention, since the myostatin gene may be used in the process of preparing a knocked out transgenic animal, it may specifically mean a cell of an animal to be transformed.
  • a pig fetus for preparing a transformed pig in which the myostatin gene is knocked out Fibroblasts were used to prepare cells knocked out of the myostatin gene.
  • a method for producing a transgenic animal other than a human whose knocked out myostatin gene comprises the following steps:
  • step (c) transplanting the nucleus of the transformed cell of step (a) into a somatic cell nuclear transfer step of transplanting the nucleus of step (b);
  • step (d) implanting the embryo cells in which the somatic cells of step (c) have been nuclear-transplanted into the oviduct of the surrogate mother of the animal to be transformed and pregnant.
  • each step is as follows:
  • the follicles are extracted from the ovary of a female pig and matured in oocyte in vitro mature culture medium (in vi t ro maturat i on medi a, IVM med ia), and then, the oocytes are collected using an egg collecting needle The nucleus of the egg was then removed using chemical and physical methods.
  • step (c) the nucleus of the transformed cell of step (a) is replaced by the nucleus of step (b) Somatic cell nuclear transfer to transplant the removed ovum
  • Somat i c-cel l nuc lear transfer refers to a technique for transplanting a nucleus of a cell into an egg which has already had its nucleus removed. After activating cytin to progress, an individual born from a surrogate mother becomes a genetically identical clone because the genetic material of the donor cell is transferred to the nuclear donor cytoplasm.
  • somatic cell nuclear transfer method cell membrane fusion, cytoplasmic microinjection, or the like can be used.
  • Cell membrane fusion has the advantage of being simple and suitable for large-scale fertilized egg production, and intracellular microinjection has the advantage of maximizing contact between the nucleus and egg material.
  • Fusion of somatic cells and nuclei from which the nucleus has been removed is achieved through a method of fusion by changing the viscosity of the cell membrane through electrical stimulation.
  • a cell membrane fusion method was used in which oocytes from which nuclei were removed and donor cells to be transplanted with the somatic cell nucleus were paralleled with electrodes, and then fusion was performed by applying a DC current pill to the fusion device.
  • step (d) implanting embryos in which the somatic cells of step (C) have been nuclear-transplanted and implanted into a fallopian tube of a surrogate mother of an animal to be transformed
  • Implantation of embryonic cells in the surrogate mother's oviduct can be carried out using a conventional method known in the art, and is not particularly limited.
  • the method for preparing a transgenic animal other than a human in which the myostatin gene is knocked out may further include the following steps.
  • step (e) separating the fetus from the surrogate mother whose pregnancy in step (d) is confirmed and confirming whether the myostatin gene is knocked out;
  • step (g) implanting embryonic cells cultured with the cells transplanted with the somatic cells of step (f) into the oviduct of the surrogate mother to be pregnant.
  • the additional steps above are for more effectively producing a transgenic animal knocked out of the myostatin gene, and the second somatic cell nuclear transfer using the nucleus of the fetal cell whose knockout of the myostatin gene is confirmed. And the embryo is to proceed. Since the specific somatic cell nuclear transfer and embryo transfer method is the same as the above method, the description is omitted to avoid excessive duplication. According to a specific example of the present invention, secondary somatic cell nuclear transfer and embryo transfer could effectively produce a transformed pig in which myostatin gene was knocked out.
  • transgenic animals other than human may be mammalian fish, birds, and the like, and specifically, may be an edible animal which may provide meat useful to humans for the purposes of the present invention.
  • transgenic animals other than human may be mammalian fish, birds, and the like, and specifically, may be an edible animal which may provide meat useful to humans for the purposes of the present invention.
  • a specific example of the invention provides a transformed pig knocked out myostatin gene by the above method.
  • the present invention provides a TALEN having the activity of binding to and cleaving a specific position of the myostatin gene.
  • the present invention provides a recombinant expression vector for myostatin gene knockout using TALEN.
  • the present invention provides a method for producing a transgenic animal other than a human whose myostatin gene is knocked out using TALEN.
  • FIG. 1 is a view showing a general principle of the working TALEN of the present invention.
  • Figure 2 is a chart analyzing and analyzing the activity of TALEN produced by targeting a specific sequence of porcine MSTN gene.
  • A is SEQ ID NO: 1
  • B is SEQ ID NO: 2
  • C is SEQ ID NO: 3
  • D is TALEN for SEQ ID NO: 4 The activity was measured and analyzed.
  • FIG. 3 is a view showing a recombinant expression vector for myostatin gene knockout using TALEN of the present invention.
  • Figure 4 shows the results of sequencing analysis to determine whether the mutation of the MSTN gene in male pig embryos produced through secondary somatic cell nuclear transfer and embryo transfer of the present invention.
  • Bold G of F1 and 18-2, 164bp of 19-3 indicate that the nucleotide sequence is inserted compared to the native form, and "- ⁇ " indicates that the nucleotide sequence is deleted compared to the native form (de l et i on).
  • FIG. 5 is a photograph comparing MSTN ⁇ / ⁇ male pigs and natural pigs produced through secondary somatic cell nuclear transfer and embryo transfer.
  • Figure 6 is a photograph comparing the analysis of the expression of MSTN protein in MSTN-/-male pigs and natural pigs produced through the secondary somatic cell nuclear transfer and embryo transfer of the present invention by Western blot.
  • Figure 7 is a photograph of the morphological comparison of muscle tissue through the immunohistochemical method in MSTN-/-male pigs and natural pigs produced through secondary somatic cell nuclear transfer and embryo transfer of the present invention.
  • FIG. 8 analyzes 400 muscle fibers by measuring the diameter of muscle fibers using Sc i on Image software in MSTN-/-male pigs and natural pigs produced through secondary somatic cell nuclear transfer and embryo transfer according to the present invention, and their average values This is a chart that calculates and compares.
  • MSTN by mutating pig MSTN gene using TALEN Recombinant expression vectors were constructed to knock out genes.
  • the specific sequence of the MSTN gene to which the TALE domain will bind was selected.
  • the first half-site sequence and the second half-site sequence to which the TALE domain binds are selected. It was. Sequence analysis of pig MSTN gene by bioinformatics method and DNA cleavage ability analysis of TALEN in specific sequence that Tlzen has in database have a length of 42-54 bp in pig MSTN gene and the starting sequence is thymine (T), a specific sequence was selected for the 4 'sequences (SEQ ID NOS: 1 to 4), the last sequence being adenine (A).
  • TALENs were generated for four sequences to measure the activity of TALENs (FIG. 2).
  • the sequence of the first half-site to which the left TALE domain is bound is 5 ⁇ -ttcaaatcctcagtaaaactt-3 ⁇
  • the sequence of the second half-site to which the right TALE domain is bound is 5,-gctcctaacattagcaaaga-.
  • the sequence to be cleaved by the Fokl cleavage domain linked to the TALE domain was cgcctggaaaca.
  • TALENs that will act on specific sequences of selected porcine MSTN genes
  • the method disclosed in the Korean patent application 'Genome engineering through designed TAL effector nuclease' was used.
  • TALE-repeats that specifically bind to each base in the selected particular sequence
  • a TALE domain capable of specifically binding to the entire specific sequence was designed, and the cleavage domain of nucleic acid endonuclease ( Fokl) were allowed to connect.
  • the entire amino acid sequence of TALEN to bind to the first half is shown in SEQ ID NO: 5
  • the entire amino acid sequence of TALEN to bind to the second half is shown in SEQ ID NO: 6.
  • the Pig myostatin TALEN for knockout of the myostatin gene contains 135 ampicillin drug resistance genes, CMV promoter, HA epitope tag, nuclear localization signal, and N-terminal end of AvrBs3. Amino acids, TALE domains that bind to specific sequences of porcine myostatin, and Fokl cleavage domains were arranged in order (FIG. 3). To join in the first half digit. The sequence of the recombinant expression vector for expressing TALEN is shown in SEQ ID NO: 7, the sequence of the recombinant expression vector for expressing TALEN to bind to the second half-digit is shown in SEQ ID NO: 8.
  • SEQ ID NO: 7 The sequence of the recombinant expression vector for expressing TALEN to bind to the second half-digit is shown in SEQ ID NO: 8.
  • Porcine fetal fibroblasts were constructed by subcultured primary fibroblasts obtained from fetuses isolated from adult female pigs 40 days old, of which the cells from the male fetus were taken as donor cells. The conventional method was applied to the process of establishing the cell line. The following related reagents were all used by Gibco Corporation (USA). The sterile membrane was cut under sterile conditions, the pig embryo was placed in PBS, repeatedly marched 3-5 times until the wash solution became clear, then transferred to Petridish, and the head and organs were cut with ophthalmic scissors. Sufficiently washed and sterilized with alcohol, it was again cut into a 0.5 lmm 3 tissue mass.
  • the cut tissue mass was placed in DMEM medium containing 0.25% trypsin and left for 4 minutes in a C02 incubator at 38.5 ° C., followed by addition of DMEM medium containing fetal bovine serum to stop the trypsin activity, and then 15 mfi. Transfer to the test tube was centrifuged for 10 minutes at a rate of 1,500 pi ⁇ . Discard the centrifuged supernatant, add 3 m £ of 20% FBS / DMEM preheated to 38.5 ° C to loosen the mass, spread evenly into a sterile cell culture dish and place in a 38.5 ° C C02 incubator Put incubated. Afterwards, the cells were passaged 3-5 times while observing the morphology and growth of the cells every day, and then used for cell transformation experiments.
  • the pig fetal fibroblasts were 15% fetal bovine blood serum, 1% non-essential amino acid,
  • the ovaries were obtained from female pigs before puberty, and then follicles of 3-8 mm diameter were sucked out of the ovaries with a follicle suction pump. Cultivated, dense, and homogeneous cumulus cell-oocyte complexes (Cu ⁇ Lus-oocyte-complexes, COCs) were selected and washed three times with TALP medium, followed by oocyte in vitro maturation (in vitro maturation media, IVM media). The components of the IVM culture were as follows: 10% pig follicle (pEE) with TCM-199, 0.6 ⁇ l iolLl L-cysteine, 10 IU / m.
  • oocytes selected as receptors were treated for 1 hour in a culture solution containing 0.4 ng / mi demecolcine and 0.05 mol / L sucrose, followed by 5 mg / cytokalacin BCcytochalasin. B) and 0.4 mg / demecolcine were transferred to the solution containing.
  • the micromanipulator (1 ⁇ 71, Olympus, Japan) was used to remove M II oocytes, first pole cells, and progenitor cell nuclei with a nucleus removal needle, and then used for somatic cell nuclear transfer.
  • Example 6 Primary Somatic Cell Nuclear Transplantation by Fusion
  • the oocytes from which the nucleus of Example 5 was removed were equilibrated in a fusion solution (0.28 ⁇ L ol / L mannitol, 0.1 ⁇ L ol / L MgC12), and then placed in an fusion bath to which the fusion solution was applied. .
  • the donor cells of Example 3 were also placed in the fusion tank, and then the donor cell-receptor oocyte membrane contact surface was parallel to the electrode using a glass needle, and then 2 kV / cm, using a fusion device (LF301, BEX, Japan). Fusion was performed by adding a DC current field of 20 mu 5 .
  • the reconstituted egg was incubated for 1 hour in a culture solution containing 0.4 mg / ⁇ demecolcine. Activated by treatment with 1.5 kV / cm, 100 us DC current field in a solution containing activator (0.28 ⁇ ol / L mannitol, 0.1 ⁇ ol / L MgC12) and 0.1 ⁇ ol / L Ca2 +, followed by 2 ⁇ ol / Incubated for 4 hours in a culture medium containing L 6-DMAP.
  • Example 7 Embryo Transfer and Pregnancy Diagnosis of Surrogate Sows
  • the embryo reconstituted by fusion in the above manner was placed in NCSU-37 culture medium,
  • R-18 205 25 3 18-1, 18-2, 18-3
  • Fetal gDNA was extracted from the fetus using the G—DEX II cDNA Extraction Kit (Intronbio, South Korea). The extracted gDNA was used as a template for nested PCR, and the primer was designed to amplify the target site of TALEN for myostatin knockout: forward C-ctggtcccgtggatctgaatg-S '), reverse direction (5'-gatcgtttccgtcgtagcgtg-3) ')
  • the primers were used to obtain a 302 bp PCR product, which was Nested PCR was performed as a template and using the following primers: 7 ⁇ 3 ⁇ 4 " 3 ⁇ 4 (5'—gaatgagaacaacacgcgagcaaagg_3 '), reverse (5' ⁇ catcttccaaggagccatcac-3 ')
  • Second somatic cell nuclear transfer and embryonic transplantation were performed using a cell line established from F2 mutated to two MSTN alleles in the mutant fetus of Example 8 as donor cells. Somatic cell nuclear transfer and embryonic transplantation were carried out in the same manner as the primary somatic cell nuclear transfer and embryo transplantation method of Examples 4 to 7. Transplantation of 8 embryos into 4 surrogate sows yielded 20 piglets (Table 2). The MSTN + cloned pigs produced by this method were pigs of the same age in the control group (2 Month age) showed distinct double muscle characteristics (FIG. 5).
  • biceps brachii were extracted from MSTN— / clone pigs and native pigs, 25 days after birth, respectively.
  • the muscle tissue was fixed in formaldehyde for 10 hours, then dehydrated using an alcohol gradient, then clarified with chloroform, embedded with paraffin, and then sliced with a microtome to a thickness of 3-5 ⁇ a day. Cut.
  • the sliced tissue slices were quickly immersed in a 30% aqueous alcohol solution, and then transferred to a 40 ° C. constant water cooler to completely spread the tissue slices and adhered to the slides.
  • MSTN— ⁇ cloned pigs were found to have an increased diameter of muscle fibers compared to native piglets (FIG. 8).

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Environmental Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Rheumatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to a transcription activator like effector-nuclease (TALEN) targeting a myostatin (MSTN) gene and a method for making an animal with a knockout myostatin gene. The present invention has confirmed that, in the case of using TALEN specific to the myostatin gene and a somatic cell nuclear transplantation technique, a transgenic pig with a knockout myostatin gene can be effectively made. Therefore, when the method for making a transgenic animal with a knockout myostatin gene of the present invention is employed, a pig with an increased muscle amount can be effectively produced, and this method can also be used for other animals that provide meat to humans.

Description

【명세서】  【Specification】
【발명의 명칭】 마이오스타틴 유전자를 표적으로 하는 TALEN 및 이를 이용한 마이오스타틴 유전자가 녹아웃된 동물을 제조하는 방법 【기술 분야】 본 특허출원은 2014 년 4 월 23 일에 중국 특허청에 제출된 중국 특허출원 제 201410168615.5 호에 대하여 우선권을 주장하며, 상기 특허출원의 개시 사항은 본 명세서에 참조로서 삽입된다. 본 발명은 마이오스타틴 (myostatin: MSTN) 유전자를 표적으로 하는 TALEN(Transcription Activator Like Effector Nuclease) 및 이를 이용한 마이오스타틴 유전자가 녹아웃된 동물을 제조하는 방법에 관한 것이다. [Name of Invention] TALEN Targeting Myostatin Gene and Method for Manufacturing Animal with Myostatin Gene Knocked Out [Technical Field] This patent application is filed with the Chinese Patent Office on April 23, 2014 in China. Priority is claimed on patent application 201410168615.5, the disclosure of which is incorporated herein by reference. The present invention relates to a Transcription Activator Like Effector Nuclease (TALEN) that targets a myostatin (MSTN) gene and a method of producing an animal knocked out of the myostatin gene.
【배경 기술】 [Background technology]
최근 돼지고기 섭취로 인한 영양문제뿐 아니라 건강문제에 대한 관심이 점점 높아지면서, 지방량이 적은 살코기에 대한 수요가 증가하고 있다. 한편, 세계에서 가장 큰 돼지고기 생산, 소비국가인 중국이 성장하면서 일인당 돼지고기 소비량은 증가하는 추세인 반면, 사육량은 이러한 증가추세에 미치지 못하고 있는 실정이다. 이와 같은 문제를 해결하는 방법 중의 하나로 근육량이 증가한 돼지를 연구, 개발하려는 노력이 있어왔다.  Recently, as the interest in health problems as well as nutritional problems due to pork intake is increasing, the demand for lean meat less fat is increasing. Meanwhile, as China, the world's largest producer and producer of pork, grows, per capita pork consumption is on the rise, while breeding is not up to this trend. One way to solve this problem has been to research and develop pigs with increased muscle mass.
마이오스타틴 (Myostatin, MSTN)은 TGF-β 패밀리에 속하는 단백질로서 근육분화과정에서 근육의 분화 및 성장을 억제하는 조절인자이며, 성장분화인자 8(growth differentiation factor 8, GDF- 8)로도 알려져 있다. 마이오스타틴은 주로 골근 세포에서 생산되어 혈액을 통해 순환하여 근육 조직에 작용을 한다. 따라서 동물에서 마이오스타틴 유전자가 없거나 마이오스타틴의 활성을 억제하는 경우 근육이 증가할 것을 기대할 수 있다. 인간과 소 (Ravi ambadur et al. , Genome Res. , 7: 910- 915, 2007), 양과 개 (Dana S. Mosher et al . , Plos genet. , 3: 779—786, 2007)에서 자연에 존재하는 마이오스타틴 돌연변이가 확인되었는더 1, 마이오스타틴 유전자에 결실이 있는 경우 천연형 (wild-type)에 비해 근육이 증가한 표현형을 나타내었다. 또한, 마이오스타틴 유전자를 녹아웃 (knock out)한 마우스를 만드는 경우, 유전자 녹아웃이 이형접합체 (heterozygote)일 때 근육이 소량 증가하였고 녹아웃이 동형접합체 (homozygote)일 때 이중 근육 (double-muscle)이 되어 근육이 크게 증가하는 것을 관찰하였다 (Se-Jin Lee et al .ᅳ Nature, 16; 98: 9306- 9311, 1997) . Myostatin (MSTN) is a protein belonging to the TGF-β family and is a regulatory factor that inhibits muscle differentiation and growth during muscle differentiation and is also known as growth differentiation factor 8 (GDF-8). . Myostatin is produced primarily from bone cells and circulates through the blood to act on muscle tissue. Therefore, if there is no myostatin gene in animals or inhibits the activity of myostatin, it can be expected to increase muscle. Humans and cattle (Ravi ambadur et al., Genome Res., 7: 910- 915, 2007), sheep and dogs (Dana S. Mosher et al., Plos genet., 3: 779-786, 2007) have identified a myostatin mutation present in nature. If present, muscle phenotype was increased compared to wild-type. In addition, when mice knocked out the myostatin gene were made, a small amount of muscle was increased when the gene knockout was a heterozygote and a double-muscle when the knockout was a homozygote. Muscle growth was observed (Se-Jin Lee et al. Nature, 16; 98: 9306-9311, 1997).
따라서 돼지에서 마이오스타틴 유전자를 효과적으로 녹아웃시킬 수 있다면, 근육이 증가한 돼지의 생산을 기대할 수 있다. 최근 출원한 중국특허 (출원번호 CN101892264A)의 경우, TALEN(Trans Activation Like Effector-Nuclease)이 아닌 다른 기술을 사용하여 마이오스타틴 유전자가 녹아웃된 형질전환돼지를 생산하였는데 마이오스타틴 단측 유전자가 녹아웃된 이형접합체이었으며, 아직까지 돼지에서 마이오스타틴 양측 유전자가 녹아웃된 동형접합체가 보고된 바는 없다.  Therefore, if the myostatin gene can be knocked out effectively in pigs, production of pigs with increased muscle can be expected. In the case of the recently filed Chinese patent (application number CN101892264A), a transgenic pig that knocked out myostatin gene was produced using a technique other than TALEN (Trans Activation Like Effector-Nuclease). Heterozygotes have not been reported in pigs with homozygotes knocked out of both myostatin genes.
【발명의 내용】 [Content of invention]
【해결하려는 과제】  [Problem to solve]
본 발명자들은 마이오스타틴 유전자가 녹아웃된 형질전환 돼지를 생산하는 방법을 발굴하고자 연구 노력하였다. 그 결과, 본 발명자들은 마이오스타틴 유전자에 특이적인 TALEN 과 체세포 핵이식 기술을 이용하는 경우, 효과적으로 마이오스타틴 유전자가 녹아웃된 형질전환 돼지를 만들 수 있다는 것을 확인함으로써, 본 발명을 완성하게 되었다.  The present inventors have tried to find a method for producing a transgenic pig knocked out of the myostatin gene. As a result, the present inventors have completed the present invention by confirming that, when using TALEN and somatic cell nuclear transfer technology specific to myostatin gene, it is possible to effectively make a transgenic pig knocked out of myostatin gene.
본 발명의 목적은 마이오스타틴 유전자의 특정 서열에 결합하여 절단하는 활성을 가지는 TALEN을 제공하는 데 있다.  It is an object of the present invention to provide a TALEN having the activity of binding to and cleaving a specific sequence of myostatin gene.
본 발명의 다른 목적은 TALEN 을 이용하는 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 제공하는 데 있다.  Another object of the present invention is to provide a recombinant expression vector for myostatin gene knockout using TALEN.
본 발명의 또 다른 목적은 TALEN 을 이용하여 마이오스타틴 유전자가 녹아웃된 세포를 제조하는 방법 및 이를 이용하여 제조된 마이오스타틴 유전자가 녹아웃된 세포를 제공하는 데 있다. 본 발명의 또 다른 목적은 TALEN 을 이용하여 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물을 제조하는 방법 및 이를 이용하여 제조된 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물을 제공하는 데 있다. Another object of the present invention to provide a method for producing a cell knocked out myostatin gene using TALEN and to provide a cell knocked out myostatin gene prepared using the same. Another object of the present invention to provide a method for producing a transgenic animal other than a human knocked out myostatin gene using TALEN and a transgenic animal other than a human knocked out myostatin gene prepared using the same There is.
본 발명의 다른 목적 및 이점은 하기의 발명의 상세한 설명, 청구범위 및 도면에 의해 보다 명확하게 된다.  Other objects and advantages of the present invention will become apparent from the following detailed description, claims and drawings.
【과제의 해결 수단】 [Measures of problem]
본 발명의 일 양태에 따르면, 마이오스타틴 (myostatin: MSTN) 유전자의 특정 서열에 결합하여 절단하는 활성을 가지는 다음을 포함하는 융합단백질을 제공한다:  According to one aspect of the present invention, there is provided a fusion protein having the activity of binding to and cleaving a specific sequence of a myostatin (MSTN) gene:
(a) TALEdranscription Activator Like Effector) 도메인; 및  (a) a TALEdranscription Activator Like Effector) domain; And
(b) 뉴클레오티드 절단 도메인.  (b) nucleotide cleavage domains.
본 발명에서 용어, "마이오스타틴 (myostatin: MSTN)"은 GDF8 (growth differentiation factor .8)로도 알려져 있으며, 근육 분화 및 성장올 억제하는 유전자로 알려져 있다. 마이오스타틴은 TGF-β 슈퍼패밀리에 속하는 조절 리간드로서 그 유전자 구조는 TGF-β 슈퍼패밀리에 속하는 다른 유전자들과 유사하다. 마이오스타틴 유전자 서열 등은 공지된 데이터 베이스 (GenBank 등)에서 얻을 수 있다. The term "statin Maio (myostatin: MSTN)" in the present invention is known as GDF8 (. Growth differentiation factor 8), is known as a gene that suppresses muscle growth and differentiation ol. Myostatin is a regulatory ligand belonging to the TGF-β superfamily and its gene structure is similar to other genes belonging to the TGF-β superfamily. Myostatin gene sequence and the like can be obtained from a known database (GenBank et al.).
마이오스타틴은 근골격 구조의 발달, 성장 및 체내의 근육 항상성의 조절에 관여하는 음성 조절자로 작용하므로, 동물에서 마이오스타틴 유전자가 없거나 마이오스타틴의 활성을 억제하는 경우 근육이 증가할 것을 기대할 수 있다. 소, 양 및 개에서 자연에 존재하는 마이오스타틴 돌연변이가 확인되었으며, 마이오스타틴 유전자에 결실이 있는 경우 천연형 (wild-type)에 비해 근육이 증가한 표현형을 나타내었다. 또한, 마이오스타틴 유전자를 녹아웃 (knock out)한 마우스를 만드는 경우, 유전자 녹아웃이 이형접합체 (heterozygote)일 때 근육이 소량 증가하였고, 녹아웃이 동형접합체 (homozygote)일 때 이중 근육 (double-muscle)이 되어 근육이 크게 증가하는 것을 관찰하였다. 따라서 인간에게 고기를 공급하는 식용동물에서 마이오스타틴 유전자를 효과적으로 녹아웃시킬 수 있다면, 근육이 증가한 식용동물의 생산을 기대할 수 있다ᅳ 본 발명의 구체적인 예에 따르면, 근육량이 증가한 돼지를 생산하기 위해, 돼지의 마이오스타틴 유전자를 녹아웃하고자 하였다. Myostatin acts as a negative regulator involved in the development, growth, and regulation of muscle homeostasis in the musculoskeletal structure, so if the animal lacks the myostatin gene or inhibits the activity of myostatin, muscle can be expected to increase. have. Myostatin mutations in nature have been identified in cattle, sheep and dogs, and the deletion of the myostatin gene showed an increased phenotype compared to the wild-type. In addition, when mice knocked out the myostatin gene were made, a small amount of muscle was increased when the gene knockout was a heterozygote, and a double-muscle when the knockout was a homozygote. It was observed that the muscles are greatly increased. Therefore, if the myostatin gene can be effectively knocked out in an animal that supplies meat to humans, the production of an animal with increased muscles can be expected. For example, to produce pigs with increased muscle mass, they tried to knock out the myostatin gene in pigs.
본 발명에서 용어, "마이오스타틴 유전자의 특정 서열에 결합하여 절단하는 활성을 가지는" 은 마이오스타틴 유전자의 특정 핵산 서열을 특이적으로 인식하여 절단하는 것을 의미하며, 본 발명에서는 TALE 도메인이 마이오스타틴의 특정 핵산 서열을 특이적으로 인식하고, 상기 TALE 도메인과 펩티드 결합으로 연결된 뉴클레오티드 절단 도메인에 의해 마이오스타틴 유전자에 절단이 발생되는 것을 의미한다. 상기 "서열''이란 그 길이와 관계없이 뉴클레오티드 서열을 의미하며, 이는 직선형, 원형 또는 가지형의 DNA 또는 RNA가 될 수도 있고, 단일 나선형이거나 이중 나선형일 수 있다.  In the present invention, the term "having the activity of binding to and cleaving a specific sequence of myostatin gene" means that the specific nucleic acid sequence of the myostatin gene is specifically recognized and cleaved. Recognizing a specific nucleic acid sequence of the ostatin specifically, it means that the cleavage is generated in the myostatin gene by the nucleotide cleavage domain connected by the TALE domain and the peptide bond. The term “sequence” means a nucleotide sequence regardless of its length, which may be linear, circular or branched DNA or RNA, and may be single helical or double helical.
TALEN이 결합하여 절단하는 특정서열을 선정하기 위해서는 여러 가지 요소를 고려할 수 있다. 우선, 타겟으로 하는 마이오스타틴 유전자만을 특이적으로 인식하고 절단하며 다른 유전자서열을 절단하지 않도록 하여야 한다 . 이를 위해서는 이미 공지되어 있는 다양한 생물정보학의 방법을 이용할 수 있다. 또한, 본 발명의 TALEN이 특정위치의 서열에 효과적으로 결합하고 절단할 수 있어야 한다. 이론적으로 모든 특정서열에 대한 TALE 도메인을 설계할 수 있지만, 실험적으로 TALE 도메인이 결합하는 특정서열의 길이 , 하나의 타겟 영역을 구성하는 2개의 절반 -자리 (hal f s i te) 사이의 스페이서의 길이 등이 TALEN의 효과적인 작용에 영향을 미치는 것으로 알려져 있다. 실험에 의하면, 절반-자리의 길이가 각각 15- 20 bp , 스페이서의 길이가 12-14 bp로서 총 서열의 길이가 42-54 bp이며, 특정서열의 시작하는 서열이 티민 (T) , 마지막 서열이 아데닌 (A)인 경우 TALEN이 효과적으로 작용한다. 따라서 이러한 요소들을 고려하여 TALEN이 특이적이고 효과적으로 마이오스타틴 유전자를 절단할 수 있도록 특정위치를 선정하여야 한다. 본 발명의 구체적인 예에 따르면, 생물정보학과 (주)를젠이 보유하고 있는 다양한 특정서열에 대한 TALEN의 절단능력에 대한 데이터베이스를 이용하여 돼지 마이오스타틴 유전자의 서열 중 4개의 특정서열 (서열목록 제 1 내지 4서열)에 대한 TALEN을 제작하였으며 최종적으로 그 중에서. 엑손 1 내에 위치하는 서열목록 저 U서열을 TALEN이 결합하여 절단하는 특정서열로 선정하였다. 본 발명의 "TALE(Transcription Activator Like Effector) 도메인 "에 대해서는 대한민국 출원특허인 '디자인된 TAL 이펙터 뉴클레아제를 통한 게놈 엔지니어링' (출원번호 10-2013-7018743)에 그 상세한 내용이 기재되어 있다. TALE은 크산토모나스 박테리아가 다양한 식물 종에 감염될 때 이들의 타입 m 분비 시스템을 통해 분비되는 단백질로서 숙주 식물 내의 프로모터 서열과 결합하여 박테리아 감염을 돕는 식물 유전자의 발현을 활성화시킬 수 있다. TALE 단백질은 34개 이하의 다양한 수의 아미노산 반복으로 구성된 중심 반복 도메인을 통해 식물의 특정한 DNA 서열을 인식하므로 이러한 특징을 이용하면 TALE을 게놈 엔지니어링의 도구를 위한 신규 폴랫품으로 사용할 수 있다. Several factors can be considered to select specific sequences to which TALENs bind and cut. First, only target myostatin genes must be specifically recognized and cleaved, and other gene sequences not to be cleaved. To this end, various known bioinformatics methods can be used. In addition, the TALENs of the present invention should be able to effectively bind and cleave sequences at specific positions. Theoretically, TALE domains can be designed for all specific sequences, but experimentally, the length of specific sequences to which TALE domains bind, the length of the spacer between two half-digits forming one target region, etc. This is known to affect the effective action of TALENs. According to the experiment, the half-digits are 15-20 bp in length, the spacers are 12-14 bp in length, the total sequence length is 42-54 bp, and the starting sequence of the specific sequence is thymine (T) and the last sequence. In the case of this adenine (A) TALEN works effectively. Therefore, in consideration of these factors, specific positions should be selected so that TALENs can cleave myostatin genes specifically and effectively. According to a specific example of the present invention, four specific sequences of the sequence of the pig myostatin gene using the database of TALEN cleavage ability for various specific sequences possessed by Bioinformatics Co., Ltd. (SEQ ID NO: TALEN for 1 to 4 sequences) . The U sequence located in exon 1 was selected as a specific sequence to be cleaved by TALEN. The "Transcription Activator Like Effector (TALE) domain" of the present invention is described in detail in the Korean patent application 'Genome engineering through designed TAL effector nuclease' (Application No. 10-2013-7018743). TALE is a protein that is secreted through their type m secretion system when xanthonomonas bacteria are infected with various plant species, and can bind to promoter sequences in the host plant to activate expression of plant genes that aid bacterial infection. The TALE protein recognizes plant specific DNA sequences through a central repeat domain consisting of up to 34 different numbers of amino acid repeats, allowing these features to be used as a novel platform for genome engineering tools.
본 발명의 TALE 도메인은 하나 이상의 TALE-반복 모들의 조합에 의해 서열-특이적 방식으로 뉴클레오티드에 결합하는 단백질 도메인을 가리킨다. 상기 TALE 도메인은 적어도 하나 이상의 TALE-반복 모들, 구체적으로 1 내지 30개의 TALE-반복 모들을 포함하나 이에 한정되지 않는다. TALE-반복 모들은 TALE 도메인 내의 특정 뉴클레오티드 서열을 인식하는 부위로서 다음의 일반적인 아미노산 서열을 가질 수 있다:  TALE domains of the invention refer to protein domains that bind to nucleotides in a sequence-specific manner by a combination of one or more TALE-repeat parents. The TALE domain includes, but is not limited to, at least one TALE-repeats moiety, specifically 1-30 TALE-repeats moieties. TALE-repeat parents may have the following general amino acid sequence as a site that recognizes a particular nucleotide sequence in the TALE domain:
H2N-LTPEQWA I ASXXGGKQALETVQRLLPVLCQAHG-COOH . XX는 위치 12 및 13에서의 초 -변성 아미노산을 가리키는 것으로서 염기 인식의 특이성을 결정한다. 상세하게는, 상기 TALE-반복 모들의 제 12 및 제 13 아미노산은 1개의 특정 핵산을 인식한다. XX가 HD일 때 , 상기 TALE-반복 모들은 C를, XX가 NG일 때 T를, XX가 NI일 때 A를, XX가 NN일 때 G를 각각 인식한다, 따라서 다양한 TALE-반복 모들을 조합하는 경우, 타겟유전자 내의 특정위치를 인식할 수 있는 TALE 도메인을 제조할 수 있다. 구체적인 TALE 도메인의 제조방법은 상기 대한민국 출원특허인 '디자인된 TAL 이펙터 뉴클레아제를 통한 게놈 엔지니어링 1 (출원번호 10-2013-7018743)에 상세하게 기재되어 있다. 한편, TALEN은 일반적으로 이량체 (dimer)로 작용하기 때문에, 타켓유전자 내의 특정위치를 절단하기 위해서는 2개의 TALEN 단량체 (monomer)를 설계할 필요가 있다 (도 1). 본 발명에서는 절단 부위를 기준으로 TALE 도메인이 결합하는 왼쪽의 특정위치를 제 1 절반- 자리 (half-site), 오른쪽의 특정위치를 제 2 절반-자리로 명명하였다. 2개의 TALEN 단량체는 각각 상이한 타켓유전자 내의 2개의 절반 -자리 중 하나를 인식하며 , 2개의 절반-자리는 9 내지 14-bp의 스페이서에 의해 서로 분리되도록 설계한다. 본 발명의 구체적인 예에 따르면 12-bp의 스페이서에 의해 서로 분리되며 각각 21bp (제 1 절반-자리, 서열목록 제 1서열), 20bp (제 2 절반-자리, 서열목록 제 2서열)를 인식하도록 TALEN 단량체를 설계하였으며, 이를 이용하여 효과적으로 돼지 마이오스타틴 유전자의 녹아웃을 유도할 수 있었다. H2N-LTPEQWA I ASXXGGKQALETVQRLLPVLCQAHG-COOH. XX refers to the super-modified amino acids at positions 12 and 13 to determine the specificity of base recognition. Specifically, the twelfth and thirteenth amino acids of the TALE-repeat parent recognize one specific nucleic acid. When XX is HD, the TALE-repeat modalities recognize C, T when XX is NG, A when XX is NI, and G when XX is NN, respectively, thus combining various TALE-repeat mods In this case, a TALE domain capable of recognizing a specific position in the target gene can be prepared. A method for preparing a specific TALE domain is described in detail in the Korean patent application 'Genome Engineering 1 through Designed TAL Effector Nuclease (Application No. 10-2013-7018743). On the other hand, since TALEN generally acts as a dimer, it is necessary to design two TALEN monomers to cut a specific position in the target gene (FIG. 1). In the present invention, the specific position on the left to which the TALE domain binds is named as a first half-site and the specific position on the right as a second half-site based on the cleavage site. Two TALEN monomers are each made up of two half-sites in different target genes. One recognizes and the two half-digits are designed to be separated from each other by spacers of 9 to 14-bp. According to a specific example of the present invention, it is separated from each other by a 12-bp spacer and recognizes 21 bp (first half-digit, sequence listing first sequence) and 20 bp (second half-site, sequence listing second sequence), respectively. The TALEN monomer was designed and used to effectively induce knockout of porcine myostatin gene.
본 발명에서 용어, "절단 "은 마이오스타틴 뉴클레오티드 분자의 공유결합된 백본의 연결을 해제하는 것을 의미하며, "뉴클레오티드 절단 도메인' '은 이러한 마이오스타틴의 뉴클레오티드 절단을 위한 효소적 활성을 갖는 폴리펩티드 서열을 의미한다.  As used herein, the term "cutting" refers to unlinking the covalently bonded backbone of the myostatin nucleotide molecule, and the "nucleotide cleavage domain" 'is a polypeptide having enzymatic activity for nucleotide cutting of such myostatin. Refers to the sequence.
상기 . 뉴클레오티드 절단 도메인은 엔도뉴클레아제 또는 엑소뉴클레아제로부터 얻을 수 있다.. 뉴클레오티드 절단 도메인을 얻어낼 수 있는 엔도뉴클레아제의 예로는 제한성 엔도뉴클레아제, 회귀성 엔도뉴클레아제 등이 있으나, 이에 한정되는 것은 아니다. 이러한 효소들은 뉴클레오티드 절단 도메인의 기원으로 사용할 수 있다. 또한 , 상기 뉴클레오티드 절단 도메인은 단일의 뉴클레오티드 서열을 절단할 수 있을 뿐만 아니라, 그 절단 도메인의 기원에 따라 이중 결합의 뉴클레오티드 서열을 절단할 수 있다.  remind . Nucleotide cleavage domains can be obtained from endonucleases or exonucleases. Examples of endonucleases from which nucleotide cleavage domains can be obtained include, but are not limited to, endogenous nucleases, recurrent endonucleases, and the like. It is not limited. Such enzymes can be used as the origin of the nucleotide cleavage domain. In addition, the nucleotide cleavage domain can cleave a single nucleotide sequence, as well as cleave a nucleotide sequence of a double bond, depending on the origin of the cleavage domain.
상기 제한성 엔도뉴클레아제는 DNA (인식 부위)와 서열 특이적 결합이 가능하며, 결합 부위의 근처에서 DNA를 절단할 수 있다. 예를 들어, 타입 lis 효소인 Fokl는 하나의 나선에서 인식 부위와 9 뉴클레오티드 떨어진 장소 및 다른 하나의 나선에서 인식 부위로부터 13 뉴클레오티드 떨어진 장소에서 DNA의 이중 나선 절단을 촉진한다.  The restriction endonuclease is capable of sequence specific binding with DNA (recognition site), and can cleave DNA in the vicinity of the binding site. For example, Fokl, a type lis enzyme, promotes double helix cleavage of DNA at 9 nucleotides from the recognition site in one helix and 13 nucleotides from the recognition site in the other helix.
본 발명의 뉴클레오티드 절단 도메인은 타입 lis 제한 엔도뉴클레아제 유래일 수 있으며, 타입 lis 제한 엔도뉴클레아제는 예를 들어, Fokl, Aarl, Acelll, Acil, AloI,'BaeI, Bbr7I , Cdil, CjePI, Ecil, Esp3I, Finl, Mbol, sa I 또는 SspD51 일 수 있으나 이에 한정되는 것은 아니다. 구체적으로 본 발명의 뉴클레오티드 절단 도메인은 Fokl 일 수 있다. Nucleotide cleavage domains of the invention may be derived from type lis restriction endonucleases, wherein the type lis restriction endonucleases are described, for example, in Fokl, Aarl, Acelll, Acil, AloI, ' BaeI, Bbr7I, Cdil, CjePI, Ecil, Esp3I, Finl, Mbol, sa I or SspD51 may be, but is not limited thereto. Specifically, the nucleotide cleavage domain of the present invention may be Fokl.
본 발명의 융합단백질은 핵 위치 신호 (nuclear localization signal) 서열을 추가로 포함할 수 있다. 상기 핵 위치 신호는 본 발명의 마이오스타틴을 표적으로 하는 TALEN을 마이오스타틴 유전자를 녹아웃시키기 위하여 핵 내로 이동시키기 위한 수단으로 사용될 수 있다. 본 발명의 다른 양태에 따르면, 상기 융합 단백질을 코딩하는 뉴클레오티드 서열; 및 (b) 상기 뉴클레오티드 서열에 작동적으로 연결되어 있는 (operat i ve l y l i nked) 전사조절서열을 포함하는 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 제공한다. 상기 발현 백터는 세포 내에 도입하여 본 발명의 융합단백질 (TALEN)을 발현시키기 위한 수단으로서, 플라스미드 백터, 코즈미드 백터, 박테리오파아지 백터 등 공지의 발현백터를 사용할 수 있으며 , 백터는 DNA 재조합 기술을 이용한 임의의 공지된 방법에 따라 당업자가 용이하게 제조할 수 있다. The fusion protein of the present invention may further comprise a nuclear localization signal sequence. The nuclear position signal of the present invention TALENs targeting myostatin can be used as a means to move into the nucleus to knock out the myostatin gene. According to another aspect of the present invention, a nucleotide sequence encoding the fusion protein; And (b) provides a recombinant expression vector for knockout myostatin gene comprising a transcriptional regulatory sequence operably linked to the nucleotide sequence (operat i ve lyli nked). The expression vector may be introduced into a cell to express a fusion protein (TALEN) of the present invention, and a known expression vector such as a plasmid vector, a cosmid vector, and a bacteriophage vector may be used. It can be easily prepared by those skilled in the art according to any known method.
상기 "작동적으로 연결되어 있는 (operat i ve ly l i nked) " 은 핵산 발현 조절서열 (예: 프로모터, 시그널 서열, 또는 전사조절인자 결합 위치의 어레이 )과 다른 핵산 서열사이의 기능적인 결합을 의미하며 , 이에 의해 상기 조절서열은 상기 다른 핵산 서열의 전사 및 /또는 해독을 조절하게 된다. 본 발명에 있어서, 융합 단백질을 코딩하는 뉴클레오티드 서열에 결합된 전사조절서열은 동물세포 (예컨대, 포유동물 세포)에서 작동하여 융합 단백질을 코딩하는 뉴클레오티드의 전사를 조절할 수 있는 것으로서, 포유동물 바이러스로부터 유래된 프로모터 및 포유동물 세포의 지놈으로부터 유래된 프로모터를 포함하며 , 예컨대, CMV (cyt omega l o vi rus ) 프로모터, 아데노바이러스 후기 프로모터, 백시니아 바이러스 7. 5K 프로모터, SV40 프로모터, HSV의 tk 프로모터, RSV 프로모터, EF1 알파 프로모터, 메탈로티오닌 프로모터, 베타 -액틴 프로모터, 인간 IL-2 유전자의 프로모터, 인간 IFN 유전자의 프로모터, 인간 IL-4 유전자의 프로모터, 인간 림포톡신 유전자의 프로모터 및 인간 GM-CSF 유전자의 프로모터를 포함하나, 이에 한정되는 것은 아니다. 또한, 본 발명의 조합 발현 백터는 폴리 아네닐화 서열을 포함할 수 있다.  The term “operat i ve ly li nked” refers to a functional binding between a nucleic acid expression control sequence (eg, an array of promoters, signal sequences, or transcriptional regulator binding sites) and other nucleic acid sequences. Whereby the regulatory sequence controls transcription and / or translation of the other nucleic acid sequence. In the present invention, the transcriptional regulatory sequence bound to the nucleotide sequence encoding the fusion protein is capable of controlling transcription of the nucleotide encoding the fusion protein by operating in an animal cell (eg, a mammalian cell), which is derived from a mammalian virus. Promoters and promoters derived from genomes of mammalian cells, such as, for example, cyt omega lo virus (CMV) promoter, adenovirus late promoter, vaccinia virus 7. 5K promoter, SV40 promoter, tk promoter of HSV, RSV Promoter, EF1 alpha promoter, metallothionine promoter, beta-actin promoter, promoter of human IL-2 gene, promoter of human IFN gene, promoter of human IL-4 gene, promoter of human lymphotoxin gene and human GM-CSF Including but not limited to promoters of genes. In addition, the combinatorial expression vector of the present invention may comprise a polyaninated sequence.
본 발명의 또 다른 양태에 따르면, 다음의 단계를 포함하는 마이오스타틴 유전자가 녹아웃된 세포를 제조하는 방법을 제공한다.  According to another aspect of the invention, there is provided a method of producing a cell knocked out myostatin gene comprising the following steps.
(a) TALEN을 이용하는 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 세포 내로 도입하는 단계;  (a) introducing into the cell a recombinant expression vector for myostatin gene knockout using TALEN;
(b) 상기 세포 내에 도입된 발현 백터로부터 발현된 TALEN에 의해 마이오스타틴 유전자가 절단되는 단계; 및 ᅳ (b) by TALEN expressed from an expression vector introduced into said cell Cleaving the myostatin gene; And ᅳ
( C ) 상기 절단에 의해 마이오스타틴 유전자에 돌연변이가 유발되는 단계.  (C) mutagenesis of the myostatin gene by the cleavage.
마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 세포 내로 도입하기 위해서 당업계에 공지된 어떠한 방법도 사용할 수 있다. 예를 들면, 상기 재조합 발현 백터를 세포 내로 도입하기 위하여 칼슘 포스페이트 -DNA 공침전법, DEAE-덱스트란 -매개 형질감염법, 폴리브렌 -매개 형질감염법 , 전기천공법, 미세주사법, 리포좀 융합법, 리포펙타민 및 원형질체 융합법 등의 당분야에 공지된 다양한 방법을 사용할 수 있다. 본 발명의 구체적인 예에 따르면 전기천공법을 이용하여 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 돼지의 태아섬유아 세포 내에 도입하였다. 상기 마이오스타틴 유전자 녹아웃용 재조합 발현 백터가 세포 내로 도입되어 이로부터 TALEN 융합단백질이 발현되면 마이오스타틴 유전자상의 원하는 부분에 이중 가닥 손상을 유도할 수 있으며, 마이오스타틴 유전자상에 이중 가닥 손상이 발생하면 세포는 자체적으로 가지고 있는 수리 기작을 이용하여 손상된 부위를 고치게 된다. 이때, 세포는 비상동 말단 결합 (non— homol ogous end j oini ng , NHEJ) 방식으로 손상된 부위를 고치게 되고, 끊어진 DNA 가닥 말단에서 삽입 ( i nser t i on)이나 결실 (de l et i on)이 일어나는 오류가 발생하기 쉬운 (error-prone ) 방향으로 수리가 진행된다. 따라서 마이오스타틴 유전자에 돌연변이가 유발되고, 결과적으로 마이오스타틴 유전자를 녹아웃 시킬 수 있다. 구체적으로 상기 돌연변이는 치환, 결실, 삽입 및 이들의 조합에 의한 돌연변이일 수 있다. 본 발명의 구체적인 예에 따르면, TALEN 융합단백질에 의해 삽입, 결실 등의 다양한 돌연변이가 유발되었으며, 이로 인해 마이오스타틴 유전자가 성공적으로 녹아웃되었다.  Any method known in the art may be used to introduce a recombinant expression vector for myostatin gene knockout into a cell. For example, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, liposome fusion to introduce the recombinant expression vector into cells. Various methods known in the art, such as lipofectamine and protoplast fusion methods, can be used. According to a specific example of the present invention, a recombinant expression vector for myostatin gene knockout was introduced into porcine fetal fibroblasts using electroporation. When the recombinant expression vector for knockout of the myostatin gene is introduced into the cell and the TALEN fusion protein is expressed therefrom, the double-strand damage on the myostatin gene can be induced. When they occur, the cell repairs the damaged area using its own repair mechanism. At this time, the cell repairs the damaged region by non-homologous end j oini ng (NHEJ) method, and insertion (i nser ti on) or deletion (de l et i on) at the end of the broken DNA strand is performed. Repairs are made in the direction of error-prone. Thus, mutations are induced in the myostatin gene, which results in knockout of the myostatin gene. Specifically, the mutation may be a mutation by substitution, deletion, insertion, and a combination thereof. According to a specific example of the present invention, various mutations such as insertion and deletion were induced by the TALEN fusion protein, thereby successfully knocking out the myostatin gene.
본 발명의 또 다른 양태에 따르면, 상기 방법에 의해 마이오스타틴 유전자가 녹아웃된 세포를 제공한다. 상기 세포는 다양한 동물 세포일 수 있으나, 본 발명의 목적상 마이오스타틴 유전자가 녹아웃된 형질전환동물을 제조하기 위한 과정에서 이용될 수 있으므로 , 구체적으로 형질전환하고자 하는 동물의 세포를 의미할 수 있다. 본 발명의 구체적인 예에 따르면, 마이오스타틴 유전자가 녹아웃된 형질전환돼지를 제조하기 위해 돼지 태아 섬유아세포를 이용하여 마이오스타틴 유전자가 녹아웃된 세포를 제조하였다. 본 발명의 또 다른 양태에 따르면, 다음의 단계를 포함하는 마이오스타틴 유전자가 녹아웃된 인간올 제외한 형질전환동물을 제조하는 방법을 제공한다: According to another aspect of the present invention, a cell in which myostatin gene is knocked out by the above method is provided. The cell may be a variety of animal cells, but for the purposes of the present invention, since the myostatin gene may be used in the process of preparing a knocked out transgenic animal, it may specifically mean a cell of an animal to be transformed. . According to a specific example of the present invention, a pig fetus for preparing a transformed pig in which the myostatin gene is knocked out Fibroblasts were used to prepare cells knocked out of the myostatin gene. According to another aspect of the present invention, there is provided a method for producing a transgenic animal other than a human whose knocked out myostatin gene comprises the following steps:
(a) TALEN을 이용하는 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 형질전환을 하고자 하는 동물의 세포 내로 도입하여 형질전환세포를 제조하는 단계 ;  (a) introducing a recombinant expression vector for myostatin gene knockout using TALEN into cells of an animal to be transformed to prepare a transformed cell;
(b) 형질전환을 하고자 하는 동물의 난소에서 난자를 채취한 후, 난자에서 핵을 제거하는 단계 ;  (b) harvesting the egg from the ovary of the animal to be transformed, and then removing the nucleus from the egg;
( c) 상기 단계 (a)의 형질전환세포의 핵을 상기 단계 (b)의 핵이 제거된 난자에 이식하는 체세포 핵이식 단계; 및  (c) transplanting the nucleus of the transformed cell of step (a) into a somatic cell nuclear transfer step of transplanting the nucleus of step (b); And
(d) 상기 단계 ( c)의 체세포가 핵이식된 세포를 배양한 배아세포를 형질전환을 하고자 하는 동물의 대리모의 수란관에 이식하여 임신시키는 단계.  (d) implanting the embryo cells in which the somatic cells of step (c) have been nuclear-transplanted into the oviduct of the surrogate mother of the animal to be transformed and pregnant.
각 단계를 보다 구체적으로 설명하면 다음과 같다:  More specifically, each step is as follows:
( a) TALEN을 이용하는 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 형질전환을 하고자 하는 동물의 세포 내로 도입하여 형질전환세포를 제조하는 단계  (A) preparing a transformed cell by introducing a recombinant expression vector for myostatin gene knockout using TALEN into the cells of the animal to be transformed
상기 단계는 마이오스타틴 유전자가 녹아웃된 세포를 제조하는 방법에서 상술하였으므로, 과도한 중복을 피하기 위해 그 기재를 생략한다.  Since the above steps are detailed in the method for preparing cells knocked out of the myostatin gene, the description thereof is omitted to avoid excessive duplication.
(b) 형질전환을 하고자 하는 동물의 난소에서 난자를 채취한 후, 난자에서 핵을 제거하는 단계  (b) collecting eggs from the ovaries of the animals to be transformed and removing the nuclei from the eggs;
동물의 난소에서 난자를 채취하기 위해서는 당업계에 공지된 통상의 방법을 이용할 수 있다ᅳ 난자에서 핵을 제거하기 위해서는 물리적인 방법, 화학적인 방법 또는 Cytocha l as i n B를 사용한 원심분리법 등을 사용할 수 있다. 본 발명의 구체적인 예에 따르면, 암컷 돼지의 난소로부터 난포를 추출하여 난모세포 체외 성숙 배양액 ( i n vi t ro maturat i on medi a , IVM med i a )에서 성숙시킨 다음, 난자 채취 바늘을 이용하여 난구세포를 제거하였으며, 이어서 화학적인 방법과 물리적인 방법을 이용하여 난자의 핵을 제거하였다.  To harvest eggs from animal ovaries, conventional methods known in the art can be used. To remove nuclei from eggs, physical methods, chemical methods or centrifugation using Cytocha l as in B can be used. have. According to a specific example of the present invention, the follicles are extracted from the ovary of a female pig and matured in oocyte in vitro mature culture medium (in vi t ro maturat i on medi a, IVM med ia), and then, the oocytes are collected using an egg collecting needle The nucleus of the egg was then removed using chemical and physical methods.
(c) 상기 단계 (a)의 형질전환세포의 핵을 상기 단계 (b)의 핵이 제거된 난자에 이식하는 체세포 핵이식 단계 (c) the nucleus of the transformed cell of step (a) is replaced by the nucleus of step (b) Somatic cell nuclear transfer to transplant the removed ovum
본 발명에서 용어, "체세포 핵 이식 (Somat i c-cel l nuc lear transfer , SCNT) "이란 세포의 핵을 이미 핵을 제거한 난자에 이식시키는 기술을 의미하며, 이와 같이 핵이식된 수정란을 발생이 진행되도록 활성화시틴 후, 대리모에 착상시켜서 태어난 개체는 핵공여 세포의 유전적 물질이 핵수여 세포질로 그대로 전달되었기 때문에 유전적으로 완전히 동일한 복제 개체가 된다. 체세포 핵 이식 방법에는 세포막융합법, 세포질내미세주입법 등을 이용할 수 있다. 세포막융합법은 간단하며 대규모 수정란 생산에 적합하다는 장점이 있으며, 세포질내 미세주입법은 핵과 난자내 물질들과의 접촉을 극대화시킨다는 장점이 있다. 체세포와 핵이 제거된 난자와의 융합은 전기자극을 통하여 세포막의 점도를 변화시켜 융합시키는 방법을 통하여 이루어진다. 본 발명의 구체적인 예에 따르면 , 핵 등이 제거된 난모세포와 체세포 핵을 이식할 공여세포를 전극과 평행하게 한 다음, 융합기기로 직류 전류 필스를 가하여 융합을 진행하는 세포막융합법을 이용하였다.  As used herein, the term “Somat i c-cel l nuc lear transfer (SCNT)” refers to a technique for transplanting a nucleus of a cell into an egg which has already had its nucleus removed. After activating cytin to progress, an individual born from a surrogate mother becomes a genetically identical clone because the genetic material of the donor cell is transferred to the nuclear donor cytoplasm. As the somatic cell nuclear transfer method, cell membrane fusion, cytoplasmic microinjection, or the like can be used. Cell membrane fusion has the advantage of being simple and suitable for large-scale fertilized egg production, and intracellular microinjection has the advantage of maximizing contact between the nucleus and egg material. Fusion of somatic cells and nuclei from which the nucleus has been removed is achieved through a method of fusion by changing the viscosity of the cell membrane through electrical stimulation. According to a specific example of the present invention, a cell membrane fusion method was used in which oocytes from which nuclei were removed and donor cells to be transplanted with the somatic cell nucleus were paralleled with electrodes, and then fusion was performed by applying a DC current pill to the fusion device.
(d ) 상기 단계 (C )의 체세포가 핵이식된 세포를 배양한 배아세포를 형질전환을 하고자 하는 동물의 대리모의 수란관에 이식하여 임신시키는 단계  (d) implanting embryos in which the somatic cells of step (C) have been nuclear-transplanted and implanted into a fallopian tube of a surrogate mother of an animal to be transformed
배아세포를 대리모의 수란관에 이식하여 임신시키는 방법은 당업계에 공지된 통상적인 방법을 이용할 수 있으며 , 특별히 한정되는 것은 아니다. 상기 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물을 제조하는 방법은 다음의 단계를 더 포함할 수 있다.  Implantation of embryonic cells in the surrogate mother's oviduct can be carried out using a conventional method known in the art, and is not particularly limited. The method for preparing a transgenic animal other than a human in which the myostatin gene is knocked out may further include the following steps.
(e) 상기 단계 (d)의 임신이 확정된 대리모에서 태아를 분리하고 마이오스타틴 유전자의 녹아웃 여부를 확인하는 단계;  (e) separating the fetus from the surrogate mother whose pregnancy in step (d) is confirmed and confirming whether the myostatin gene is knocked out;
( f ) 마이오스타틴 유전자가 녹아웃된 태아의 세포의 핵을 상기 단계 (f) the nuclei of fetal cells in which myostatin gene is knocked out
(b)의 핵이 제거된 난자에 이식하는 체세포 핵이식 단계; 및 a somatic cell nuclear transfer step of implanting an egg from which the nucleus of (b) was removed; And
(g) 상기 단계 ( f )의 체세포가 핵이식된 세포를 배양한 배아세포를 대리모 동물의 수란관에 이식하여 임신시키는 단계.  (g) implanting embryonic cells cultured with the cells transplanted with the somatic cells of step (f) into the oviduct of the surrogate mother to be pregnant.
상기의 추가적인 단계들은 마이오스타틴 유전자가 녹아웃된 형질전환동물을 더 효과적으로 제조하기 위한 것으로서 , 마이오스타틴 유전자의 녹아웃이 확인된 태아의 세포의 핵을 이용하여 2차 체세포 핵이식 및 배아이식을 진행하는 것이다. 구체적인 체세포 핵이식 및 배아이식 방법은 상기 방법과 동일하므로 , 과도한 중복을 피하기 위해 그 기재를 생략한다. 본 발명의 구체적인 예에 따르면, 2차 체세포 핵이식 및 배아이식을 통해 효과적으로 마이오스타틴 유전자가 녹아웃된 형질전환돼지를 생산할 수 있었다.. The additional steps above are for more effectively producing a transgenic animal knocked out of the myostatin gene, and the second somatic cell nuclear transfer using the nucleus of the fetal cell whose knockout of the myostatin gene is confirmed. And the embryo is to proceed. Since the specific somatic cell nuclear transfer and embryo transfer method is the same as the above method, the description is omitted to avoid excessive duplication. According to a specific example of the present invention, secondary somatic cell nuclear transfer and embryo transfer could effectively produce a transformed pig in which myostatin gene was knocked out.
본 발명의 또 다른 양태에 따르면, 상기 방법에 의해 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물을 제공한다. 본 발명에서 용어, "인간을 제외한 형질전환동물 "은 포유동물 어류, 조류 등일 수 있으며 , 구체적으로 본 발명의 목적상 인간에게 유용한 고기를 제공할 수 있는 식용동물일 수 있다. 본 발명의 구체적인 예에 따르면 상기 방법에 의해 마이오스타틴 유전자가 녹아웃된 형질전환돼지를 제공한다 .  According to another aspect of the present invention, a transgenic animal other than a human whose myostatin gene is knocked out by the above method is provided. In the present invention, the term "transgenic animals other than human" may be mammalian fish, birds, and the like, and specifically, may be an edible animal which may provide meat useful to humans for the purposes of the present invention. According to a specific example of the invention provides a transformed pig knocked out myostatin gene by the above method.
【발명의 효과】 【Effects of the Invention】
본 발명의 특징 및 이점을 요약하면 다음과 같다:  The features and advantages of the present invention are summarized as follows:
( a ) 본 발명은 마이오스타틴 유전자의 특정 위치에 결합하여 절단하는 활성을 가지는 TALEN을 제공한다.  (a) The present invention provides a TALEN having the activity of binding to and cleaving a specific position of the myostatin gene.
(b) 본 발명은 TALEN을 이용하는 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 제공한다.  (b) The present invention provides a recombinant expression vector for myostatin gene knockout using TALEN.
( c ) 본 발명은 TALEN을 이용하여 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물을 제조하는 방법을 제공한다.  (c) The present invention provides a method for producing a transgenic animal other than a human whose myostatin gene is knocked out using TALEN.
( d) 본 발명의 방법을 이용하면, 마이오스타틴 유전자가 녹아웃된 형질전환돼지를 효과적으로 생산할 수 있으므로, 근육량이 증가된 돼지를 얻을 수 있으며, 이러한 방법은 인간에게 고기를 공급하는 다른 동물에도 이용될 수 있다.  (d) By using the method of the present invention, it is possible to effectively produce a transformed pig in which the myostatin gene is knocked out, so that a pig having an increased muscle mass can be obtained, and this method is also used for other animals supplying meat to humans Can be.
【도면의 간단한 설명】 [Brief Description of Drawings]
도 1은 본 발명의 TALEN이 작용하는 일반적인 원리를 나타내는 도면이다.  1 is a view showing a general principle of the working TALEN of the present invention.
도 2는 돼지 MSTN 유전자의 특정서열을 타켓팅하여 제작한 TALEN의 활성을 측정하여 분석한 도표이다. A는 서열목록 제 1서열, B는 서열목록 게 2서열, C는 서열목록 제 3서열, D는 서열목록 제 4서열에 대한 TALEN의 활성을 측정하여 분석한 것이다. Figure 2 is a chart analyzing and analyzing the activity of TALEN produced by targeting a specific sequence of porcine MSTN gene. A is SEQ ID NO: 1, B is SEQ ID NO: 2, C is SEQ ID NO: 3, D is TALEN for SEQ ID NO: 4 The activity was measured and analyzed.
도 3은 본 발명의 TALEN을 이용한 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 나타내는 도면이다.  3 is a view showing a recombinant expression vector for myostatin gene knockout using TALEN of the present invention.
도 4는 본 발명의 2차 체세포 핵이식 및 배아이식을 통해 생산된 수컷 돼지 태아에서 MSTN 유전자의 돌연변이 여부를 확인하기 위해 시퀀싱 분석을 한 결과이다. F1 및 18-2의 볼드체 G , 19-3의 164bp는 천연형에 비해 염기서열이 삽입 ( i nser t i on)된 것을 나타내며, " -ᅳ " 는 천연형에 비해 염기서열이 결실 (de l et i on)된 것을 나타낸다.  Figure 4 shows the results of sequencing analysis to determine whether the mutation of the MSTN gene in male pig embryos produced through secondary somatic cell nuclear transfer and embryo transfer of the present invention. Bold G of F1 and 18-2, 164bp of 19-3 indicate that the nucleotide sequence is inserted compared to the native form, and "-ᅳ" indicates that the nucleotide sequence is deleted compared to the native form (de l et i on).
도 5는 본 발명의 2차 체세포 핵이식 및 배아이식을 통해 생산된 MSTN-/- 수컷 돼지와 천연형 돼지를 비교한 사진이다.  FIG. 5 is a photograph comparing MSTN − / − male pigs and natural pigs produced through secondary somatic cell nuclear transfer and embryo transfer.
도 6는 본 발명의 2차 체세포 핵이식 및 배아이식을 통해 생산된 MSTN-/- 수컷 돼지와 천연형 돼지에서 MSTN 단백질의 발현여부를 웨스턴 블롯으로 비교분석한 사진이다.  Figure 6 is a photograph comparing the analysis of the expression of MSTN protein in MSTN-/-male pigs and natural pigs produced through the secondary somatic cell nuclear transfer and embryo transfer of the present invention by Western blot.
도 7는 본 발명의 2차 체세포 핵이식 및 배아이식을 통해 생산된 MSTN-/- 수컷 돼지와 천연형 돼지에서 면역조직화학분석 방법을 통해 근육조직을 형태학적으로 비교분석한 사진이다.  Figure 7 is a photograph of the morphological comparison of muscle tissue through the immunohistochemical method in MSTN-/-male pigs and natural pigs produced through secondary somatic cell nuclear transfer and embryo transfer of the present invention.
도 8는 본 발명의 2차 체세포 핵이식 및 배아이식을 통해 생산된 MSTN-/- 수컷 돼지와 천연형 돼지에서 Sc i on Image 소프트웨어로 근섬유 직경을 측정하여 400개의 근섬유를 분석하고, 이들의 평균치를 계산하여 비교분석한 도표이다.  FIG. 8 analyzes 400 muscle fibers by measuring the diameter of muscle fibers using Sc i on Image software in MSTN-/-male pigs and natural pigs produced through secondary somatic cell nuclear transfer and embryo transfer according to the present invention, and their average values This is a chart that calculates and compares.
【발명을 실시하기 위한 구체적인 내용】 [Specific contents to carry out invention]
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시예에 의해 제한되지 않는다는 것은 당업계에서 통상의 지식을 가진 자에 있어서 자명할 것이다. 실시예 실시예 1 : 마이오스타틴 유전자녹아웃용 재조합 발현 백터의 제작  Hereinafter, the present invention will be described in more detail with reference to Examples. These examples are only for illustrating the present invention more specifically, it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples in accordance with the gist of the present invention. . EXAMPLES Example 1 Preparation of a Recombinant Expression Vector for Myostatin Gene Knockout
TALEN을 이용하여 돼지 MSTN 유전자에 돌연변이를 유발하여 MSTN 유전자를 녹아웃하기 위한 재조합 발현 백터를 제작하였다. 이를 위해 우선MSTN by mutating pig MSTN gene using TALEN Recombinant expression vectors were constructed to knock out genes. First of all for this
TALE 도메인이 결합할 MSTN 유전자의 특정서열을 선정하였다. TALE 도메인에 연결된 Fokl 절단 도메인이 동형이량체 (homdimer)를 이루어 작용할 수 있도록 TALE 도메인이 각각 결합하는 제 1 절반 자리 (half- site)의 서열과 제 2 절반 자리 (half-site)의 서열을 선정하였다. 생물 정보학 방법에 의한 돼지 MSTN 유전자의 서열분석과 (주)틀젠이 데이터베이스화하여 가지고 있는 특정서열에서의 TALEN의 DNA절단 능력 분석을 통해 돼지 MSTN 유전자 중 42-54 bp의 길이를 가지며 시작서열이 티민 (T), 마지막 서열이 아데닌 (A)인 4개의' 서열 (서열목톡 제 1 내지 4서열)을 특정서열을 선택하였다. 4개의 서열에 대해 TALEN을 제작하여 TALEN의 활성을 측정하였으며 (도 2), 분석결과, TALEN이 활성을 나타내며 MSTN 유전자의 게 1 액손 내에 위치하고 있는 서열목록 제 1서열을 최종적으로 선택하였다. 왼쪽 TALE 도메인이 결합할 제 1 절반 자리 (half- site)의 서열은 5\-ttcaaatcctcagtaaaactt-3\이었으며, 오른쪽 TALE 도메인이 결합할 제 2 절반 자리 (half-site)의 서열은 5、- gctcctaacattagcaaaga- 이었으며, TALE 도메인에 연결된 Fokl 절단 도메인에 의해 절단될 서열은 cgcctggaaaca이었다. The specific sequence of the MSTN gene to which the TALE domain will bind was selected. In order for the Fokl cleavage domain linked to the TALE domain to act as a homodimer, the first half-site sequence and the second half-site sequence to which the TALE domain binds are selected. It was. Sequence analysis of pig MSTN gene by bioinformatics method and DNA cleavage ability analysis of TALEN in specific sequence that Tlzen has in database have a length of 42-54 bp in pig MSTN gene and the starting sequence is thymine (T), a specific sequence was selected for the 4 'sequences (SEQ ID NOS: 1 to 4), the last sequence being adenine (A). TALENs were generated for four sequences to measure the activity of TALENs (FIG. 2). As a result of analysis, the first sequence of the sequence list, in which TALENs are active and located within the crab 1 axon of the MSTN gene, was finally selected. The sequence of the first half-site to which the left TALE domain is bound is 5 \ -ttcaaatcctcagtaaaactt-3 \, and the sequence of the second half-site to which the right TALE domain is bound is 5,-gctcctaacattagcaaaga-. The sequence to be cleaved by the Fokl cleavage domain linked to the TALE domain was cgcctggaaaca.
선정된 돼지 MSTN 유전자의 특정서열에 작용할 TALEN을 제조하기 위하여 대한민국 출원특허인 '디자인된 TAL 이펙터 뉴클레아제를 통한 게놈 엔지니어링 ' (출원번호 10-2013-7018743)에 개시된 방법을 이용하였다. 선정된 특정서열 내의 각각의 염기에 특이적으로 결합하는 TALE-반복 모들을 조합하여 결과적으로 전체 특정서열에 특이적으로 결합할 수 있는 TALE 도메인을 설계하였으며 여기에 핵산 엔도 뉴클레아제의 절단 도메인 (Fokl )이 연결되도록 하였다. 제 1 절반자리에 결합할 TALEN의 전체 아미노산 서열은 서열목록 제 5서열과 , 제 2 절반자리에 결합할 TALEN의 전체 아미노산 서열은 서열목록 제 6서열과 같다. 결과적으로, 마이오스타틴 유전자 녹아웃용 재조합 발현 백터 (Pig myostatin TALEN)에는 암피실린 (Ampici llin) 약물 내성 유전자, CMV 프로모터, HA 에피토프 태그, 핵 위치 신호 (nuclear localization signal), AvrBs3의 N-말단 135개 아미노산, 돼지 마이오스타틴의 특정서열에 결합하는 TALE 도메인, Fokl 절단 도메인이 순서대로 배열되도록 하였다 (도 3). 제 1 절반자리에 결합할 TALEN을 발현하기 위한 재조합 발현백터의 서열은 서열목록 제 7서열, 제 2 절반자리에 결합할 TALEN을 발현하기 위한 재조합 발현백터의 서열은 서열목록 제 8서열과 같다 . 실시예 2: 돼지 태아섬유아세포주 (fibroblast cell line)의 구축 In order to produce TALENs that will act on specific sequences of selected porcine MSTN genes, the method disclosed in the Korean patent application 'Genome engineering through designed TAL effector nuclease' (Application No. 10-2013-7018743) was used. By combining TALE-repeats that specifically bind to each base in the selected particular sequence, a TALE domain capable of specifically binding to the entire specific sequence was designed, and the cleavage domain of nucleic acid endonuclease ( Fokl) were allowed to connect. The entire amino acid sequence of TALEN to bind to the first half is shown in SEQ ID NO: 5, and the entire amino acid sequence of TALEN to bind to the second half is shown in SEQ ID NO: 6. Consequently, the Pig myostatin TALEN for knockout of the myostatin gene contains 135 ampicillin drug resistance genes, CMV promoter, HA epitope tag, nuclear localization signal, and N-terminal end of AvrBs3. Amino acids, TALE domains that bind to specific sequences of porcine myostatin, and Fokl cleavage domains were arranged in order (FIG. 3). To join in the first half digit. The sequence of the recombinant expression vector for expressing TALEN is shown in SEQ ID NO: 7, the sequence of the recombinant expression vector for expressing TALEN to bind to the second half-digit is shown in SEQ ID NO: 8. Example 2 Construction of Porcine Febroblast Cell Line
돼지 태아 섬유아세포주는 임신한지 40일된 성체 암컷 돼지에서 분리한 태아로부터 얻어진 초대 섬유아세포 (primary fibroblast)를 계대배양하여 구축하였으며, 그 중에서 수컷 태아로부터 얻어진 세포를 공여세포로 삼았다. 세포주를 확립하는 과정에는 통상적인 방법을 적용하였다. 이하 관련된 시약은 모두 Gibco사 (USA)의 것을 사용하였다. 무균 상태에서 태막을 절취하여, 돼지태아를 PBS에 넣고 세척액이 투명해질 때까지 반복적으로 3-5번 행군 다음, 페트리디시로 이동시킨 후, 안과용 가위로 머리 부위와 장기를 자르고, 75%의 알코올로 충분이 세척하여 소독하고, 다시 이를 0.5- lmm3 크기의 조직덩어리로 절단하였다. 상기 절단한 조직덩어리를 0.25% 트립신을 포함한 DMEM 배지에 넣고 38.5°C의 C02 인큐베이터에서 4분 동안 방치한 후, 소 태아 혈청을 함유한 DMEM 배지를 첨가하여 트립신의 활성을 정지시킨 다음, 15 mfi 실험관에 옮계 1,500 π叩의 속도로 10분 동안 원심 분리를 하였다. 원심분리한 상층액을 버리고 , 38.5°C로 예열된 3 m£의 20% FBS/DMEM를 첨가하여 덩어리를 풀어준 후, 무균 처리된 세포배양 접시에 균일하게 뿌린 다음, 38.5°C C02 인큐베이터에 넣어 배양하였다. 이후 매일 세포의 형태와 성장 상황을 관찰하면서 3-5번 계대한 후, 세포 형질전환 실험에 사용하였다. 실시예 3: 전기천공법 (electroporation)에 의한 돼지 태아섬유아세포의 형질전환 Porcine fetal fibroblasts were constructed by subcultured primary fibroblasts obtained from fetuses isolated from adult female pigs 40 days old, of which the cells from the male fetus were taken as donor cells. The conventional method was applied to the process of establishing the cell line. The following related reagents were all used by Gibco Corporation (USA). The sterile membrane was cut under sterile conditions, the pig embryo was placed in PBS, repeatedly marched 3-5 times until the wash solution became clear, then transferred to Petridish, and the head and organs were cut with ophthalmic scissors. Sufficiently washed and sterilized with alcohol, it was again cut into a 0.5 lmm 3 tissue mass. The cut tissue mass was placed in DMEM medium containing 0.25% trypsin and left for 4 minutes in a C02 incubator at 38.5 ° C., followed by addition of DMEM medium containing fetal bovine serum to stop the trypsin activity, and then 15 mfi. Transfer to the test tube was centrifuged for 10 minutes at a rate of 1,500 pi 叩. Discard the centrifuged supernatant, add 3 m £ of 20% FBS / DMEM preheated to 38.5 ° C to loosen the mass, spread evenly into a sterile cell culture dish and place in a 38.5 ° C C02 incubator Put incubated. Afterwards, the cells were passaged 3-5 times while observing the morphology and growth of the cells every day, and then used for cell transformation experiments. Example 3 Transformation of Porcine Fetal Fibroblasts by Electroporation
전기천공법으로 세포를 형질전환하기 위하여 ½axa4D(Lonza, 스위스)를 이용하였으며, 형질전환된 세포를 선별하기 위해 H-2Kk 마그네틱 리포터를 이용하는 방법 (Kim H. et al . , PLoS ONE 8(2):e54676 (2013))을 이용하였다. 구체적인 방법은 다음과 같다: ½axa4D (Lonza, Switzerland) was used to transform the cells by electroporation, and H-2Kk magnetic reporter was used to select the transformed cells (Kim H. et al., PLoS ONE 8 (2)). e 54676 (2013). The specific method is as follows:
상기 돼지 태아 섬유아세포를 15% 소 태아 혈정, 1% 비필수아미노산, The pig fetal fibroblasts were 15% fetal bovine blood serum, 1% non-essential amino acid,
100 mg/m£ 페니실린과 100 mg/m 스트렙토마이신이 함유된 DMEM 배지에서 In DMEM medium containing 100 mg / m £ penicillin and 100 mg / m streptomycin
1.4 배양하였다. 배양한 세포를 트립신을 처리하여 떼어낸 다음, 세포들을 HBSS (Wei Gen, 대한민국)로 세척하여 원심분리를 하였다. 세포를. 카운팅하여 최종적으로 약 1 X 106개의 세포를 세포에 도입하고자 하는 DNA 플라스미드가 포함되어 있는 완층용액에 현탁시킨 다음, 전기천공법 (electroporation)을 수행하였다. 사용한 DNA의 비율은 45:45:10(TALEN monomer를 암호화하는 플라스미드 (DAS): TALEN monomer를 암호화하는 플라스미드 (RR): H-2Kk 마그네틱 리포터 유전자 폴라스미드)로 하였다. 전기천공법을 거친 세포를 배양액에 풀어 배양접시에 뿌린 후 5% C02, 37°C의 인큐베이터에서 2일 동안 배양하였다. 형질전환된 세포를 선별하기 위하여 Magnetic Separation and Antibiotics Select ion(Kim H. et al . , PLoS ONE 8(2):e54676 (2013))을 수행하였다. 이와 같이 선별된 형질전환 세포들을 15% FBS/DMEM에서 배양하여 4-8세대 계대 배양한 다음, 체세포 핵 이식의 공여세포로 활용하였다. 실시예 4: 난모세포 체외 성숙 배양 1.4 Incubated. The cultured cells were removed by treatment with trypsin, and the cells were washed with HBSS (Wei Gen, South Korea) and centrifuged. Cells. After counting, the cells were finally suspended in the complete solution containing the DNA plasmid to be introduced into the cells, followed by electroporation. The ratio of DNA used was 45:45:10 (plasmid encoding TALEN monomer (DAS): plasmid encoding TALEN monomer (RR): H-2Kk magnetic reporter gene polamide). After electroporation, cells were cultured, sprinkled on culture plates, and incubated in an incubator at 5% C02, 37 ° C for 2 days. Magnetic Separation and Antibiotics Select ion (Kim H. et al., PLoS ONE 8 (2): e54676 (2013)) was performed to select transformed cells. The transformed cells thus selected were cultured in 15% FBS / DMEM, passaged 4-8 generation, and used as donor cells for somatic cell nuclear transfer. Example 4: Oocyte In Vitro Maturation Culture
사춘기 전의 암컷 돼지로부터 난소를 얻은 다음, 난포흡입펌프로 난소로부터 직경이 3-8 隱인 난포를 빨아내었다. 난구가 3충 이상으로 감싸져 있고 치밀하며 세포질이 균일한 난구 세포—난모 세포 복합체 (Cu隠 Lus-oocyte-complexes, COCs)를 선택하였고, TALP 배지로 세 번 세척한 다음, 난모세포 체외 성숙 배양액 (in vitro maturation media, IVM media)에 넣었다. IVM 배양액의 성분은 다음과 같다: TCM-199 첨가 10% 돼지 난포액 (pEE), 0.6 謂 iol · L-l L-시스테인, 10 IU/m.£ 인간 융모성 생식선 자극호르몬 (hCG), 10 IU/mf, 임신된 말에서 얻어진 혈청성 성선 자극 호르몬 (PMSG), 10 ng/niC 표피성장인자 (EGF). 22 시간 후 호르몬이 없는 IVM 배양액으로 교체하였다. 다시 38.5°C, 5% C02 인큐베이터에서 38시간 배양한 다음, 0.1% 히알루로니다아제 (hyaluronisase)에 넣고, 직경이 170- 180μηΊ인 난자 채취 바늘을 이용하여 반복적으로 난모세포를 흡입하여 난구세포를 제거하였다. 난구세포가 제거된 난모세포 중에서 난황주위간극 (Per ivi tell ine space)이 선명하고, 난세포가 완전하며, 거 U극 세포가 배출된 난모세포를 선택하여 이후 체세포 핵이식에 사용하였다. 실시예 5: 화학 보조 핵 제거 방법 (chemically assisted enucleation)을 이용한 핵의 제거 The ovaries were obtained from female pigs before puberty, and then follicles of 3-8 mm diameter were sucked out of the ovaries with a follicle suction pump. Cultivated, dense, and homogeneous cumulus cell-oocyte complexes (Cu 隠 Lus-oocyte-complexes, COCs) were selected and washed three times with TALP medium, followed by oocyte in vitro maturation (in vitro maturation media, IVM media). The components of the IVM culture were as follows: 10% pig follicle (pEE) with TCM-199, 0.6 μl iolLl L-cysteine, 10 IU / m. £ Human chorionic gonadotropin (hCG), 10 IU / mf Serum Gonadotropin (PMSG), 10 ng / niC Epidermal Growth Factor (EGF), obtained from pregnant horses. After 22 hours it was replaced with hormone-free IVM culture. Incubate for 38 hours in a 38.5 ° C, 5% C02 incubator, and then put it in 0.1% hyaluronidase and repeatedly inhale oocytes using an oocyte harvesting needle with a diameter of 170-180 μηΊ. Removed. Among the oocytes from which the oocytes were removed, the oocytes with clear viable perivian infiltrate (Per ivi tell ine space), oocytes intact, and giant U-pole cells were selected and used for somatic cell nuclear transfer. Example 5 Removal of Nuclei Using a Chemically Assisted Enucleation
상기 난모세포에서 핵을 제거하기 위하여, 수용체로 선택된 난모세포를 0.4 ng/mi 디메콜신 (Demecolcine)과 0.05 mol/L 수크로즈를 포함한 배양액에서 1 시간 동안 처리한 후, 5 mg/ 시토칼라신 BCcytochalasin B)와 0.4 mg/ 디메콜신 (demecolcine)이 포함된 용액으로 옮기었다. 현미경 조작 시스템 (micromanipulator, 1 X 71, Olympus, Japan)을 사용하여 핵 제거 바늘로 M II 난모세포, 제 1극세포 및 돌기측 세포핵을 제거한 뒤, 체세포 핵이식에 이용하였다. 실시예 6: 융합에 의한 1차 체세포 핵이식  In order to remove nuclei from the oocytes, oocytes selected as receptors were treated for 1 hour in a culture solution containing 0.4 ng / mi demecolcine and 0.05 mol / L sucrose, followed by 5 mg / cytokalacin BCcytochalasin. B) and 0.4 mg / demecolcine were transferred to the solution containing. The micromanipulator (1 × 71, Olympus, Japan) was used to remove M II oocytes, first pole cells, and progenitor cell nuclei with a nucleus removal needle, and then used for somatic cell nuclear transfer. Example 6: Primary Somatic Cell Nuclear Transplantation by Fusion
상기 실시예 5의 핵 등이 제거된 난모세포를 융합액 (0.28 瞧 ol/L mannitol, 0.1 隱 ol/L MgC12)에서 평형 (equil ibrat i on)시킨 후, 융합액을 도포한 융합조 내에 넣었다. 융합조 내에 상기 실시예 3의 공여세포도 넣은 다음 유리 바늘을 이용하여 공여세포-수용체 난모 세포막 접촉면을 전극과 평행하게 한 다음, 융합기기 (LF301, BEX, 일본)를 이용하여 2 kV/cm, 20μ5의 직류 전류 필스를 가하여 융합을 진행하였다. 융합한 후 재구축된 난자를 0.4 mg/ηιί 디메콜신 (demecolcine)이 포함된 배양액에서 1 시간 동안 배양한 다음. 활성화액 (0.28 画 ol/L mannitol, 0.1 隱 ol/L MgC12)과 0.1 瞧 ol/L Ca2+가 포함된 용액에서 1.5 kV/cm, 100 u s 직류 전류 필스를 처리하여 활성화시킨 후, 2 隱 ol/L 6-DMAP가 포함된 배양액에서 4 시간 동안 배양하였다. 실시예 7: 배아 이식 및 대리모돈의 임신 진단 The oocytes from which the nucleus of Example 5 was removed were equilibrated in a fusion solution (0.28 μL ol / L mannitol, 0.1 μL ol / L MgC12), and then placed in an fusion bath to which the fusion solution was applied. . The donor cells of Example 3 were also placed in the fusion tank, and then the donor cell-receptor oocyte membrane contact surface was parallel to the electrode using a glass needle, and then 2 kV / cm, using a fusion device (LF301, BEX, Japan). Fusion was performed by adding a DC current field of 20 mu 5 . After fusion, the reconstituted egg was incubated for 1 hour in a culture solution containing 0.4 mg / ηιί demecolcine. Activated by treatment with 1.5 kV / cm, 100 us DC current field in a solution containing activator (0.28 画 ol / L mannitol, 0.1 隱 ol / L MgC12) and 0.1 瞧 ol / L Ca2 +, followed by 2 隱 ol / Incubated for 4 hours in a culture medium containing L 6-DMAP. Example 7: Embryo Transfer and Pregnancy Diagnosis of Surrogate Sows
상기 방법으로 융합시켜 재구축된 배아를 NCSU-37 배양액에 넣어, The embryo reconstituted by fusion in the above manner was placed in NCSU-37 culture medium,
38°C , 5% C02 인큐베이터에서 배양하였으며, 배아 구축 당일을 0일로 하여 재구축 배아를 7-8일간 배양하였으며, 배양 후 38시간과 7일째에 각각 난분할율과 배낭 발육 상황 (embryo sac development)을 관찰하고 기록하였다. Incubated in 38 ° C, 5% C02 incubator, reconstructed embryos were incubated for 7-8 days with embryonic day 0, and egg split and backpack development at 38 hours and 7 days after incubation, respectively. Was observed and recorded.
배아 이식을 위해 발정 주기가 정상이고, 생식기관에 질병이 없는 요크셔 (Yorkshire) 돼지를 대리모돈으로 선택하였다. 대리모돈에 1000 II] 임신말 혈청성 성선 자극 호르몬 (PMSG)을 주사하고 72 시간 후쎄 근육에 1000 IU 인간 융모성 생식선 자극호르몬 (hCG)을 주사하여 발정을 유도하였다. hCG를 주사하고 48 시간 후, 1 내지 3일 배양한 난분할이 정상인 1 내지 8 세포기의 배아를 수술을 통해 대리모돈의 수란관에 이식하였다. 배아를 이식한 후 24-26일 동안 B형 초음파 진단을 통해 임신 상황을 검사하였다. 3 마리의 대리모돈에 배아를 이식한 결과, 총 17 개의 태 ( 「 : & 丁 으며 , 이를 정리한 표는 다음과 같다 (표 1). For embryo transplantation, Yorkshire pigs with normal estrous cycle and diseased reproductive organs were selected as surrogate sows. 1000 II to surrogate sows] Estrus was induced by injection of serum gonadotropin (PMSG) at the end of gestation and injection of 1000 IU human chorionic gonadotropin (hCG) into the muscle after 72 hours. 48 hours after hCG injection, embryos of 1 to 8 cell stages with normal egg division cultured for 1 to 3 days were surgically implanted into the surrogate tube of surrogate. The embryo was transplanted and examined for pregnancy during 24-26 days after transplantation. After transplantation of the embryos to a surrogate mother three money, a total of 17 states ( ': was &丁, table summarizes them as follows (Table 1).
[표 1] 체세포 핵 이시 τ 배아 이식을 진행하여 얻은 태아  [Table 1] Fetus obtained through somatic cell nuclear transfer τ embryo transfer
대리임 이식 임 태 태아 번호 신 배아 신나 (이 아수 Representative Lim Tae Im transplanted fetal renal number of embryos thinner (This Assou
암¾돼 수량 (일  Female quantity (day
지 번호 ) Support number)
R-29 196 36 9 Fl, F2, F3  R-29 196 36 9 Fl, F2, F3
F4, F5, F6  F4, F5, F6
F7, F8, F9  F7, F8, F9
R-18 205 25 3 18-1, 18-2, 18-3 R-18 205 25 3 18-1, 18-2, 18-3
R-19 232 27 5 19-1, 19-2, 19-3 R-19 232 27 5 19-1, 19-2, 19-3
19-4, 19-5 실시예 8: 태아에서 MSTN 유전자의 돌연변이 확인  19-4, 19-5 Example 8: Mutation Identification of MSTN Gene in Fetuses
체세포 핵이식 및 배아 이식을 통해 얻은 태아에서 MSTN 유전자의 돌연변이 여부를 확인하기 위해 다음의 실험을 수행하였다:  The following experiments were performed to determine whether the MSTN gene was mutated in fetuses obtained through somatic cell nuclear transfer and embryo transfer:
배아 이식 36일 후 Β형 초음파 진단으로 임신이 확정된 대리모돈으로부터 자궁을 적출하고, 넁장 보관 상태로 실험실로 가져온 후 자웅에서 태아를 분리하였다. 태아에서 G— DEX II cDNA 추출 키트 (인트론바이오, 대한민국)를 사용하여 태아의 gDNA를 추출하였다. 추출한 gDNA를 nested PCR의 주형으로 사용하였으며, 프라이머는 마이오스타틴 녹아웃용 TALEN의 타켓 부위를 증폭할 수 있도록 다음과 같이 설계하였다: 정방향 C-ctggtcccgtggatctgaatg-S'), 역방향 (5'- gatcgtttccgtcgtagcgtg-3' )  After 36 days of embryo transplantation, the uterus was extracted from surrogate sows whose pregnancy was confirmed by Β-type ultrasound diagnosis. Fetal gDNA was extracted from the fetus using the G—DEX II cDNA Extraction Kit (Intronbio, South Korea). The extracted gDNA was used as a template for nested PCR, and the primer was designed to amplify the target site of TALEN for myostatin knockout: forward C-ctggtcccgtggatctgaatg-S '), reverse direction (5'-gatcgtttccgtcgtagcgtg-3) ')
상기 프라이머를 사용하여 302 bp의 PCR 생성물을 얻었으며, 이를 주형으로 하고 다음의 프라이머를 사용하여 nested PCR을 수행하였다: 7"¾(5'—gaatgagaacagcgagcaaaagg_3'), 역방향 (5 '一 catcttccaaggagccatcac-3' ) The primers were used to obtain a 302 bp PCR product, which was Nested PCR was performed as a template and using the following primers: 7 ^ ¾ " ¾ (5'—gaatgagaacaacacgcgagcaaaagg_3 '), reverse (5' 一 catcttccaaggagccatcac-3 ')
PCR 수행결과, 257 bp의 PCR 생성물을 얻었으며, 돌연변이 여부확인을 위한 T7E1 미스매치 (mismatch) 분석을 하기 위해 PCR 생성물을 재풀림시킨후 다시 결합시켜 이종이중체가 만들어지도록 하였다. 이종이중체 DNA를 T7E1 효소 ((주 )를젠 , 대한민국)로 처리하여 15분 반응 후, 아가로스 젤에 로딩하여 분석하였다. 분석결과, T7E1 효소에 의해 절단되어 미스매치가 확인된 PCR 생성물들을 T-Blunt PCR Cloning it(SolGent, 대한민국)를 사용하여 클론닝하였다. 클론닝한 폴라스미드를 M13 프라이머를 이용하여 시퀀싱 분석을 하였다 (도 4). 분석결과, 10개 태아에서 MSTN 유전자에 돌연변이가 있었으며, 이 중 3개는 MSTN 두 대립유전자에 돌연변이 (bi-allelic null mutation, MSTN—ᅳ)가 존재하였고 (Fl, F2, 19-3), 7개는 MSTN 하나의 대립유전자에만 돌연변이 (Heterozygous knockout , MSTN+/_)가 있었다 (F3, F5, F9, 18-2, 18- 3, 19-1, 19-4) . 획득한 10개 돌연변이 태아로부터 상기 실시예 2의 방법을 이용하여 세포주를 확립하였다. 실시예 9: 2차 체세포 핵 이식 및 배아 이식 As a result of PCR, a 257 bp PCR product was obtained, and the heterologous product was made by re-unfolding the PCR product again for the analysis of T7E1 mismatch for mutation. The heteroduplex DNA was treated with T7E1 enzyme (Jen, Republic of Korea) for 15 minutes and then loaded onto an agarose gel for analysis. As a result, PCR products cleaved by T7E1 enzyme and confirmed mismatch were cloned using T-Blunt PCR Cloning it (SolGent, South Korea). The cloned pollmids were subjected to sequencing analysis using M13 primer (FIG. 4). As a result, there were mutations in the MSTN gene in 10 fetuses, three of which had a bi-allelic null mutation (MSTN— ᅳ) in two alleles of MSTN (Fl, F2, 19-3), 7 Dogs had mutations (Heterozygous knockout, MSTN + / _ ) in only one allele of MSTN (F3, F5, F9, 18-2, 18-3, 19-1, 19-4). Cell lines were established from the obtained 10 mutant fetuses using the method of Example 2 above. Example 9: Secondary Somatic Cell Nuclear Transplantation and Embryo Transplantation
상기 실시예 8의 돌연변이 태아 중 MSTN 두 대립유전자에 돌연변이를 나타낸 F2로부터 확립한 세포주를 공여세포로 하여 2차 체세포 핵이식 및 배아이식을 진행하였다. 체세포 핵 이식 및 배아 이식은 실시예 4 내지 7의 1차 체세포 핵이식 및 배아 이식방법과 동일한 방법으로 수행하였다. 4마리의 대리모돈에 8기개의 배아를 이식한 결과 20마리의 새끼돼지를 얻을 수 있었다 (표 2) 상기 방법을 통하여 생산된 MSTN+ 클론 돼지 (cloned pig)는, 대조군의 동일한 나이의 돼지 (2개월 나이 )에 비하여 뚜렷한 더블 근육 (double muscle) 특성을 나타내었다 (도 5).  Second somatic cell nuclear transfer and embryonic transplantation were performed using a cell line established from F2 mutated to two MSTN alleles in the mutant fetus of Example 8 as donor cells. Somatic cell nuclear transfer and embryonic transplantation were carried out in the same manner as the primary somatic cell nuclear transfer and embryo transplantation method of Examples 4 to 7. Transplantation of 8 embryos into 4 surrogate sows yielded 20 piglets (Table 2). The MSTN + cloned pigs produced by this method were pigs of the same age in the control group (2 Month age) showed distinct double muscle characteristics (FIG. 5).
[표 2]
Figure imgf000020_0001
R-l 222 6 MAl-1 485
TABLE 2
Figure imgf000020_0001
Rl 222 6 MAl-1 485
MA 1-2 575  MA 1-2 575
MA1-3 545  MA1-3 545
MA1-4 430  MA1-4 430
MA1-5 1060  MA1-5 1060
MA1-6 655  MA1-6 655
R-3 222 4 MA3-1 840  R-3 222 4 MA3-1 840
MA3-2 980  MA3-2 980
MA3-3 515  MA3-3 515
MA3-4 1075  MA3-4 1075
R-4 232 4 MA4-1 780  R-4 232 4 MA4-1 780
MA4-2 700  MA4-2 700
MA4- 580  MA4- 580
3(사태)  3 (the situation)
MA4-4 615  MA4-4 615
R-28 205 9 MA28-1 700  R-28 205 9 MA28-1 700
MA28-2 1295  MA28-2 1295
MA28-3 840  MA28-3 840
MA28-4 915  MA28-4 915
MA28- 500  MA28- 500
5(사태)  5 (the situation)
MA28-6 1095  MA28-6 1095
MA28-7 630  MA28-7 630
MA28-8 540  MA28-8 540
MA28- 1235  MA28-1235
9(사태) 실시예 10 : MSTN Z 클론 돼지에서의 마이오스타틴 발현여부 확인  9 (Situation) Example 10: Confirmation of Myostatin Expression in MSTN Z Clone Pigs
MSTN+ 클론 돼지에서 마이오스타틴의 발현여부를 확인하기 위하여 웨스턴블롯을 수행하였다. MSTN 클론 돼지와 천연형 돼지의 근육조직으로부터 RIPA 분해액 ( 50 niM Tr i s-HCl , ΡΗ8.0 , 150 mM NaCl , 1% NP-40, 0.5 % sodium deoxycholate, 0.1% SDS)을 사용하여 총 단백질을 추출하였다. 단백질을 SDS-폴리아크릴아미드겔 전기영동 (SDS-PAGE)으로 분리한 다음, PVDF 필름 (GE water & Process Technologies. Trevose, PA. USA)에 옮기고, 5% 탈지 분유용액에 넣어 실내 온도에서 1 시간 동안 배양하여 비특이적 결합을 억제한 다음, 여기에 1차 항체 (anti-Myostatin, 영인프론티어, 대한민국)를 첨가하고 4°C에서 하룻밤 배양하였다. 세척 후, HRP가 표지된 2차 항체 (anti-mouse IgG, Santa Cruz, CA, USA)를 1:5000로 희석하여 첨가하고 실온에서 1시간 동안 배양하였다. 세척 후, 향상된 화학발광 기 즉정방법 (Enhanced chemi luminescence assay, Animal Genetics Inc. , 대한민국)을 통해 발생된 신호를 분석하였다. GAPDH(Santa Cruz, CA, USA)를 내재적 컨트롤로 사용하였다. 분석결과, 천연형 돼지에서는 마이오스타틴이 검출되었으나, MSTN+ 클론 돼지에서는 마이오스타틴이 검출되지 않았으며 (도 6), 이를 통해 MSTN— Λ 클론 돼지에서 마이오스타틴이 발현되지 않는다는 것을 확인할 수 있었다. 실시예 11: 면역 조직 화학 및 형태학 관찰을 통한 MSTff/_ 클론 돼지의 더블 근육 특성 (double-muscle trait) 확인 Western blot was performed to confirm the expression of myostatin in MSTN + cloned pigs. RIPS lysis solution (50 niM Tr i s-HCl, Ρ Η8.0, 150 mM NaCl, 1% from MSTN cloned pig and native pig muscle tissue) Total protein was extracted using NP-40, 0.5% sodium deoxycholate, 0.1% SDS). Proteins were separated by SDS-polyacrylamide gel electrophoresis (SDS-PAGE), then transferred to PVDF film (GE water & Process Technologies. Trevose, PA. USA) and placed in 5% skim milk powder solution for 1 hour at room temperature. After incubation to inhibit nonspecific binding, the primary antibody (anti-Myostatin, Youngin Frontier, Korea) was added thereto and incubated overnight at 4 ° C. After washing, HRP labeled secondary antibody (anti-mouse IgG, Santa Cruz, Calif., USA) was added diluted 1: 5000 and incubated for 1 hour at room temperature. After washing, the signals generated by the enhanced chemiluminescence assay (Enhanced chemi luminescence assay, Animal Genetics Inc., Korea) were analyzed. GAPDH (Santa Cruz, CA, USA) was used as an intrinsic control. As a result, myostatin was detected in natural pigs, but myostatin was not detected in MSTN + cloned pigs (FIG. 6), and it was confirmed that myostatin was not expressed in MSTN— Λ cloned pigs. . Example 11 Identification of Double-Muscle traits of MSTff / _Clone Pigs by Immunohistochemical and Morphological Observations
MSTN^- 클론 돼지의 더블 근육 특성을 확인하기 위해, 출생한 후 25일이 된 MSTN— / 클론 돼지와 천연형 돼지에서 각각 위팔두갈래근 (biceps brachii)을 적출하였다. 근육 조직을 포름알데히드에서 10시간 동안 고정시킨 후, 알코올 구배를 이용하여 탈수시킨 다음, 클로로포름으로 투명화시키고, 파라핀으로 포매 (Embedding)한 다음 절편기 (microtome)로 두께가 일 3-5 μπι가 되도록 절단하였다. 절단한 조직 박편들을 신속하게 30% 알코올 수용액에 담근 다음, 40°C 항온의 은수기에 옮겨 조직 박편을 완전히 펴놓은 후, 슬라이드에 부착시켰다. 조직이 부착된 슬라이드를 건조시킨 후 56-60°C에서 30분 동안 가열한 다음, HE(Hematoxyline- Eosin)염색을 하였다 (도 7). 세포핵은 파란색으로 나타났으며, 근육의 세포질은 빨간색으로 나타났다. MSTN"/_ 클론 돼지는 천연형 새끼 돼지에 비해 MSTN 새끼 돼지가 대조군에 비하여 근육 절편 상의 근섬유가 비대하고 치밀한 것으로 나타났다 . 한편 , Scion Image 소프트웨어를 이용하여 근섬유 직경과 개수를 측정하였다. 400개의 근섬유를 분석한 결과, 4068 To confirm the double muscle characteristics of MSTN ^ -clone pigs, biceps brachii were extracted from MSTN— / clone pigs and native pigs, 25 days after birth, respectively. The muscle tissue was fixed in formaldehyde for 10 hours, then dehydrated using an alcohol gradient, then clarified with chloroform, embedded with paraffin, and then sliced with a microtome to a thickness of 3-5 μπι a day. Cut. The sliced tissue slices were quickly immersed in a 30% aqueous alcohol solution, and then transferred to a 40 ° C. constant water cooler to completely spread the tissue slices and adhered to the slides. After the tissues were attached to the slide was dried for 30 minutes at 56-60 ° C, HE (Hematoxyline-Eosin) staining (Fig. 7). The nucleus is blue and the cytoplasm of muscle is red. MSTN "/ _ cloned pigs were found to have larger and denser muscle fibers on muscle sections than native piglets compared to control piglets. Meanwhile, the diameter and number of muscle fibers were measured using Scion Image software. 400 muscle fibers After analyzing 4068
MSTN— Λ 클론 돼지는 천연형 새끼 돼지에 비해 근섬유의 직경이 증가한 것을 확인하였다 (도 8) . MSTN— Λ cloned pigs were found to have an increased diameter of muscle fibers compared to native piglets (FIG. 8).
이상으로 본 발명의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적인 기술은 단지 구체적인 실현예일 뿐이며 , 이에 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 따라서 본 발명의 실질적인 범위는 첨부된 청구항과 그의 등가물에 의하여 정의된다 .  As described above in detail specific parts of the present invention, it is apparent to those skilled in the art that these specific techniques are merely specific embodiments, and the scope of the present invention is not limited thereto. Thus, the substantial scope of the present invention is defined by the appended claims and their equivalents.

Claims

【특허청구범위】 【Patent Claims】
【청구항 1] [Claim 1]
마이오스타틴 (myostatin: MSTN) 유전자의 특정 서열에 결합하여 절단하는 활성을 가지는 다음을 포함하는 융합단백질: A fusion protein containing the following, which has the activity of binding to and cutting a specific sequence of the myostatin (MSTN) gene:
(a) TALECTranscript ion Activator Like Effector) 도메인; 및 (a) TALECTranscript ion Activator Like Effector) domain; and
(b) 뉴클레오티드 절단 도메인. (b) Nucleotide cleavage domain.
【청구항 2】 【Claim 2】
제 1항에 있어서, 상기 특정 서열은 길이가 42 내지 54 bp이며, 시작하는 염기서열이 티민 (T), 마지막 염기서열이 아데닌 (A)인 것을 특징으로 하는 융합단백질. The fusion protein according to claim 1, wherein the specific sequence is 42 to 54 bp in length, and the starting base sequence is thymine (T) and the last base sequence is adenine (A).
【청구항 3】 【Claim 3】
제 2항에 있어서 , 상기 특정 서열은 서열목록 제 1 내지 제 4서열 중 어느 하나인 것을 특징으로 하는 융합단백질. The fusion protein according to claim 2, wherein the specific sequence is any one of sequences 1 to 4 in the sequence list.
【청구항 4] [Claim 4]
제 2항에 있어서, 상기 특정 서열은 서열목록 제 1서열인 것을 특징으로 하는 융합단백질. The fusion protein according to claim 2, wherein the specific sequence is the first sequence of the sequence list.
【청구항 5】 【Claim 5】
제 1항에 있어서, 상기 뉴클레오티드 절단 도메인은 타입 lis 제한 엔도뉴클레아제 유래인 것을 특징으로 하는 융합단백질. The fusion protein according to claim 1, wherein the nucleotide cleavage domain is derived from type lis restriction endonuclease.
【청구항 6】 【Claim 6】
제 5항에 있어서 , 상기 타입 lis 제한 엔도뉴클레아제는 Fokl인 것을 특징으로 하는 융합단백질. The fusion protein according to claim 5, wherein the type lis restriction endonuclease is Fokl.
【청구항 7】 【Claim 7】
(a) 상기 제 1 항 내지 제 6항 중 어느 한 항의 융합 단백질을 코딩하는 뉴클레오티드 서열; 및 (b) 상기 뉴클레오티드 서열에 작동적으로 연결되어 있는 (operat i ve ly l i nked) 전사조절서열을 포함하는 마이오스타틴 유전자 녹아웃용 재조합 발현 백터. (a) a nucleotide sequence encoding the fusion protein of any one of claims 1 to 6; and (b) operably to the nucleotide sequence. A recombinant expression vector for knocking out the myostatin gene containing linked (operativ ely linked) transcriptional regulatory sequences.
【청구항 8] [Claim 8]
다음의 단계를 포함하는 마이오스타틴 유전자가 녹아웃된 세포를 제조하는 방법 : Method for producing cells in which the myostatin gene has been knocked out, comprising the following steps:
(a) 제 7 항의 재조합 발현 백터를 세포 내로 도입하는 단계; (a) introducing the recombinant expression vector of claim 7 into a cell;
(b) 상기 세포 내에 도입된 발현 백터로부터 발현된 TALEN에 의해 마이오스타틴 유전자가 절단되는 단계 ; 및 (b) cleaving the myostatin gene by TALEN expressed from the expression vector introduced into the cell; and
( c) 상기 절단에 의해 마이오스타틴 유전자에 돌연변이가 유발되는 단계. (c) A step in which a mutation is induced in the myostatin gene by the cleavage.
【청구항 9】 【Claim 9】
제 8 항의 방법에 의해 마이오스타틴 유전자가 녹아웃된 세포. Cells in which the myostatin gene has been knocked out by the method of claim 8.
【청구항 10] [Claim 10]
제 9항에 있어서, 상기 세포는 돼지 태아 .섬유아세포인 것을 특징으로 하는 세포. The cell according to claim 9, wherein the cell is a porcine fetal fibroblast.
【청구항 11】 【Claim 11】
다음의 단계를 포함하는 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물을 제조하는 방법 : Method for producing transgenic animals other than humans in which the myostatin gene has been knocked out, comprising the following steps:
(a ) 제 7항의 마이오스타틴 유전자 녹아웃용 재조합 발현 백터를 형질전환을 하고자 하는 동물의 세포 내로 도입하여 형질전환세포를 제조하는 단계; (a) preparing transformed cells by introducing the recombinant expression vector for knocking out the myostatin gene of claim 7 into cells of an animal to be transformed;
(b) 형질전환을 하고자 하는 동물의 난소에서 난자를 채취한 후, 난자에서 핵을 제거하는 단계 ; (b) collecting eggs from the ovaries of the animal to be transformed and then removing the nucleus from the eggs;
(c ) 상기 단계 (a)의 형질전환세포의 핵을 상기 단계 (b)의 핵이 제거된 난자에 이식하는 체세포 핵이식 단계; 및 (c) a somatic cell nuclear transfer step of transplanting the nucleus of the transformed cell in step (a) into the egg from which the nucleus was removed in step (b); and
(d) 상기 단계 ( c )의 체세포가 핵이식된 세포를 배양한 배아세포를 형질전환을 하고자 하는 동물의 대리모의 수란관에 이식하여 임신시키는 단계 (d) Embryonic cells cultured from the somatic cell nuclear transfer cells of step (c) are implanted into the oviduct of the surrogate mother of the animal to be transformed, resulting in pregnancy. step
【청구항 12] [Claim 12]
제 11항에 있어서 상기 방법은 다음의 단계를 더 포함하는 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물을 제조하는 방법: The method of claim 11, further comprising the following steps:
( e ) 제 11항의 상기 단계 (d)의 임신이 확정된 대리모에서 태아를 분리하고, 태아에서 마이오스타틴 유전자의 녹아웃 여부를 확인하는 단계 ; (e) separating the fetus from the surrogate mother whose pregnancy has been confirmed in step (d) of paragraph 11, and checking whether the myostatin gene has been knocked out in the fetus;
( f ) 마이오스타틴 유전자가 녹아웃된 태아의 세포의 핵을 제 11항의 상기 단계 ( b)의 핵이 제거된 난자에 이식하는 체세포 핵이식 단계 ; 및 (f) a somatic cell nuclear transfer step of transplanting the nucleus of a fetal cell in which the myostatin gene has been knocked out into the egg from which the nucleus has been removed in step (b) of item 11; and
(g) 상기 단계 ( f )의 체세포가 핵이식된 세포를 배양한 배아세포를 대리모 동물의 수란관에 이식하여 임신시키는 단계. (g) A step of implanting the embryonic cells cultured from the somatic cell nuclear transfer cells of step (f) into the oviduct of a surrogate animal to become pregnant.
【청구항 13】 【Claim 13】
제 11항 또는 제 12항에 있어서, 상기 형질전환동물은 형질전환돼지인 것을 특징으로 하는 방법 . The method according to claim 11 or 12, wherein the transgenic animal is a transgenic pig.
【청구항 14] [Claim 14]
제 11 항 또는 제 12항의 방법에 의해 마이오스타틴 유전자가 녹아웃된 인간을 제외한 형질전환동물 . Transgenic animals other than humans in which the myostatin gene has been knocked out by the method of claim 11 or 12.
【청구항 15] [Claim 15]
제 14항에 있어서, 상기 형질전환동물은 형질전환돼지인 것을 특징으로 하는 동물. The animal according to claim 14, wherein the transgenic animal is a transgenic pig.
PCT/KR2015/004068 2014-04-23 2015-04-23 Talen targeting myostatin gene and method for making animal with knockout myostatin gene using same WO2015163711A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201410168615.5A CN103952424B (en) 2014-04-23 2014-04-23 Method for producing double-muscular trait somatic cell cloned pig with MSTN (myostatin) bilateral gene knockout
CN201410168615.5 2014-04-23

Publications (1)

Publication Number Publication Date
WO2015163711A1 true WO2015163711A1 (en) 2015-10-29

Family

ID=51329776

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2015/004068 WO2015163711A1 (en) 2014-04-23 2015-04-23 Talen targeting myostatin gene and method for making animal with knockout myostatin gene using same

Country Status (3)

Country Link
KR (2) KR20150123187A (en)
CN (1) CN103952424B (en)
WO (1) WO2015163711A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104946654A (en) * 2015-07-14 2015-09-30 江苏省家禽科学研究所 shRNA sequence for restraining duck MSTN genetic expression and application thereof
CN116855539A (en) * 2023-07-18 2023-10-10 中农种源(深圳)科技有限公司 Pig breeding method capable of simultaneously knocking out CD163, pAPN and MSTN genes and improving disease resistance and quality

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130116306A (en) * 2011-01-03 2013-10-23 주식회사 툴젠 Genome engineering via designed tal effector nucleases
KR20140046408A (en) * 2011-02-25 2014-04-18 리컴비네틱스 인코포레이티드 Genetically modified animals and methods for making the same

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101892264A (en) * 2010-05-28 2010-11-24 吉林大学 Establishment of myostatin (MSTN) gene knock-out pig
CN102021201A (en) * 2010-09-06 2011-04-20 南京农业大学 Preparation method for myostatin knock-out pig
CN102296073B (en) * 2011-08-11 2014-04-23 中国农业科学院北京畜牧兽医研究所 Specific target site for site-directed knockout of gene Myostatin by zinc finger nuclease
CN103088044B (en) * 2011-11-02 2014-09-03 内蒙古大学 Targeting vector for knockout of bovine MSTN gene and application thereof
AR091482A1 (en) 2012-06-21 2015-02-04 Recombinetics Inc GENETICALLY MODIFIED CELLS AND METHODS FOR OBTAINING
PT3494997T (en) 2012-07-25 2019-12-05 Massachusetts Inst Technology Inducible dna binding proteins and genome perturbation tools and applications thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20130116306A (en) * 2011-01-03 2013-10-23 주식회사 툴젠 Genome engineering via designed tal effector nucleases
KR20140046408A (en) * 2011-02-25 2014-04-18 리컴비네틱스 인코포레이티드 Genetically modified animals and methods for making the same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
SUNG, YOUNG HOON ET AL.: "Knockout mice created by TALEN-mediated gene targeting", NATURE BIOTECHNOLOGY, vol. 31, no. 1, January 2013 (2013-01-01), pages 23 - 24, XP055597463, DOI: 10.1038/nbt.2477 *
TESSON: "Knockout rats generated by embryo microinjection of TALENs", NATURE BIOTECHNOLOGY, vol. 29, no. 8, 2011, pages 695 - 696, XP002681970, DOI: 10.1038/NBT.1940 *
XU: "Targeted myostatin gene editing in multiple mammalian species directed by a single pair of TALE nucleases", MOLECULAR THERAPY-NUCLEIC ACIDS, vol. 2, no. 7, 2013, pages 1 - 10, XP055184659, DOI: doi:10.1038/mtna.2013.39 *

Also Published As

Publication number Publication date
CN103952424A (en) 2014-07-30
CN103952424B (en) 2017-01-11
KR20170023043A (en) 2017-03-02
KR20150123187A (en) 2015-11-03
KR101748823B1 (en) 2017-06-19

Similar Documents

Publication Publication Date Title
CN107988256B (en) Recombinant vector for knocking-in human Huntington gene, construction method thereof and application thereof in construction of model pig
CN106191113B (en) Preparation method of MC3R gene knockout pig
JP5774657B2 (en) Method for genetic modification of mammals using electroporation
KR20180091821A (en) How to manipulate humanized CAR T-cells and platelets by genetic complementarity
AU2015404563A1 (en) Pathogen-resistant animals having modified CD163 genes
JP5817955B2 (en) Production of hemophilia A model pig
JP2019507610A (en) Fel d1 knockout and related compositions and methods based on CRISPR-Cas genome editing
US11419320B2 (en) Cold-resistant and lean-type transgenic pig and preparation method therefor
US20210037797A1 (en) Inducible disease models methods of making them and use in tissue complementation
CA2978457A1 (en) Etv2 and uses thereof
US8119785B2 (en) Nucleic acid sequences and homologous recombination vectors for distruption of a Fel D I gene
WO2015163711A1 (en) Talen targeting myostatin gene and method for making animal with knockout myostatin gene using same
AU2016288671A1 (en) Humanized heart muscle
US10626417B2 (en) Method of genetically altering and producing allergy free cats
WO2019141052A1 (en) Method for preparing non-human primate somatic cell cloned animal
CN104388560A (en) Method for marking Y chromosome and application thereof
US9999206B2 (en) Model and method for a transgenic bovidae expressing cardiac fibrosis and associated pathology
KR100479704B1 (en) Method of producing a transgenic pig expressing green fluorescent protein
JP5771240B2 (en) Immunodeficient pig
WO2016074503A1 (en) Y chromosome modification method and use thereof
KR100502480B1 (en) Method of producing porcine clonal somatic cell line expressing green fluorescent protein
JP2023554545A (en) Transgenic animals with modified myostatin genes
Pinkert Genetic engineering of farm mammals
CN117965622A (en) Method for establishing AIDS disease model cat
CN115322993A (en) Safety locus for site-specific integration of exogenous gene in pig genome and method for constructing pig breeding group by using safety locus

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15783456

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1)EPC, FORM 1205A DATED 11.04.2017

122 Ep: pct application non-entry in european phase

Ref document number: 15783456

Country of ref document: EP

Kind code of ref document: A1