US20220220221A1 - Extracellular matrix-producing composition using mast4 gene and preparation method therefor - Google Patents

Extracellular matrix-producing composition using mast4 gene and preparation method therefor Download PDF

Info

Publication number
US20220220221A1
US20220220221A1 US17/494,765 US202117494765A US2022220221A1 US 20220220221 A1 US20220220221 A1 US 20220220221A1 US 202117494765 A US202117494765 A US 202117494765A US 2022220221 A1 US2022220221 A1 US 2022220221A1
Authority
US
United States
Prior art keywords
composition
mast4
fragment
protein
eukaryotic cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/494,765
Inventor
Seong Jin Kim
Han Sung JUNG
Satoru Takahashi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kim Yeung Won June
Kim Ypsse
Original Assignee
Individual
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
Priority claimed from PCT/KR2018/002763 external-priority patent/WO2018164507A2/en
Application filed by Individual filed Critical Individual
Priority to US17/494,765 priority Critical patent/US20220220221A1/en
Assigned to KIM, YEUNG WON JUNE, KIM, YPSSE, Kim, Saerom, KIM, SEONG JIN reassignment KIM, YEUNG WON JUNE ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JUNG, HAN SUNG, TAKAHASHI, SATORU, KIM, SEONG JIN
Publication of US20220220221A1 publication Critical patent/US20220220221A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N5/0655Chondrocytes; Cartilage
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • C12N15/1031Mutagenizing nucleic acids mutagenesis by gene assembly, e.g. assembly by oligonucleotide extension PCR
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells 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
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • 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)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11001Non-specific serine/threonine protein kinase (2.7.11.1), i.e. casein kinase or checkpoint kinase
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/65MicroRNA
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/70Enzymes
    • C12N2501/72Transferases [EC 2.]
    • C12N2501/727Kinases (EC 2.7.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites

Definitions

  • the present disclosure relates to a composition for producing an extracellular matrix from eukaryotic cells using Microtubule Associated Serine/Threonine Kinase Family Member 4 (MAST4) gene, a method of producing the extracellular matrix from the eukaryotic cells, and a composition for promoting chondrogenesis, the composition including the above composition.
  • MAST4 Microtubule Associated Serine/Threonine Kinase Family Member 4
  • MAST4 Microtubule associated serine/threonine kinase 4 is known to be expressed in cartilage ( BMC Genomics 2007, 8: 165), but its role has not been clearly elucidated.
  • CN 105636614 discloses that MAST4 may be used for the treatment of cartilage, but this is based only on the stochastic results of MAST4 expression in the cartilage, and does not elucidate specific roles thereof.
  • MAST4 as a novel central regulator that is involved in chondrogenesis, and provide a source technology for the development of substances that modulate the activity of MAST4.
  • An aspect provides a composition for promoting production of an extracellular matrix from eukaryotic cells, the composition including a compound capable of specifically binding to Microtubule Associated Serine/Threonine Kinase Family Member 4 (MAST4) protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • MAST4 Microtubule Associated Serine/Threonine Kinase Family Member 4
  • compositions for promoting chondrogenesis of chondrocytes including a compound capable of specifically binding to MAST4 protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • Still another aspect provides a method of producing an extracellular matrix from eukaryotic cells, the method including contacting the eukaryotic cells with the composition for promoting production of the extracellular matrix from eukaryotic cells.
  • compositions for promoting production of an extracellular matrix from eukaryotic cells including a compound capable of specifically binding to Microtubule Associated Serine/Threonine Kinase Family Member 4 (MAST4) protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • MAST4 Microtubule Associated Serine/Threonine Kinase Family Member 4
  • compositions for promoting chondrogenesis of chondrocytes including a compound capable of specifically binding to MAST4 protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • the compound capable of specifically binding to the MAST4 protein or the fragment thereof, or the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof includes those capable of at least partially binding to the protein or the fragment thereof, or the nucleic acid.
  • the compound may be a chemically synthesized compound, polypeptide, or polynucleotide, or a combination thereof. These compounds may inhibit activity or expression of MAST4 protein.
  • the composition for promoting production of extracellular matrix from eukaryotic cells may be a composition for promoting chondrogenesis from eukaryotic cells.
  • the activity inhibitor of MAST4 protein or the expression inhibitor of MAST4 protein includes any one, as long as it is able to inhibit expression of MAST4 gene or activity of MAST4 protein.
  • the activity inhibitor or the expression inhibitor may be a polynucleotide complementary to the entire or a part of the MAST4 gene.
  • the polynucleotide sequence may be RNA, DNA, or a hybrid thereof.
  • the activity inhibition of the MAST4 protein may be kinase activity inhibition of the MAST4 protein.
  • MAST4 is a kinase capable of phosphorylating Ser or Thr of a target substrate, and the kinase activity inhibition of the MAST4 protein means blocking of phosphorylation of a target substrate of MAST4, specifically, blocking of phosphorylation of Ser or Thr.
  • the polypeptide specifically binding to MAST4 protein or the fragment thereof, or the polypeptide specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof may be an antibody or an antigen-binding fragment thereof.
  • antibody means a specific immunoglobulin directed against an antigenic site.
  • MAST4 gene is cloned into an expression vector to obtain the MAST4 protein encoded by the gene, and the antibody may be prepared from the protein according to a common method in the art.
  • a type of the antibody includes a polyclonal antibody or a monoclonal antibody, and includes all immunoglobulin antibodies.
  • the antibody includes not only complete forms having two full-length light chains and two full-length heavy chains but also functional fragments of antibody molecules which have a specific antigen binding site (binding domain) directed against an antigenic site to retain an antigen-binding function, although they do not have the intact complete antibody structure having two light chains and two heavy chains.
  • polynucleotide may be used in the same meaning as a nucleotide or a nucleic acid, unless otherwise mentioned, and refers to a deoxyribonucleotide or a ribonucleotide.
  • the polynucleotide may include an analog of a natural nucleotide and an analog having a modified sugar or base moiety, unless otherwise mentioned.
  • the polynucleotide may be modified by various methods known in the art, as needed.
  • modification may include methylation, capping, substitution of a natural nucleotide with one or more homologues, and modification between nucleotides, for example, modification to uncharged linkages (e.g., methylphosphonate, phosphotriester, phosphoroamidate, carbamate, etc.) or charged linkages (e.g., phosphorothioate, phosphorodithioate, etc.).
  • uncharged linkages e.g., methylphosphonate, phosphotriester, phosphoroamidate, carbamate, etc.
  • charged linkages e.g., phosphorothioate, phosphorodithioate, etc.
  • the polynucleotide capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof may be microRNA (miRNA), small interfering RNA (siRNA), short hairpin RNA (shRNA), Piwi-interacting RNA (piRNA), small nuclear RNA (snRNA), or antisense oligonucleotide, each specific to the nucleic acid encoding the MAST4 protein or the fragment thereof, or a combination thereof.
  • miRNA microRNA
  • siRNA small interfering RNA
  • shRNA short hairpin RNA
  • piRNA Piwi-interacting RNA
  • snRNA small nuclear RNA
  • antisense oligonucleotide each specific to the nucleic acid encoding the MAST4 protein or the fragment thereof, or a combination thereof.
  • the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof may include the polynucleotide capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof, and may be CRISPR-Cas including guide RNA specific to the nucleic acid encoding the MAST4 protein or the fragment thereof.
  • the Cas may be Cas9.
  • CRISPRs Clustered Regularly Interspaced Short Palindromic Repeats
  • Cas9 an essential protein element which forms a complex with guide RNA (specifically, two RNAs, called CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA), included in guide RNA), and it serves as an active endonuclease.
  • the guide RNA may have a form of a dual RNA including CRISPR RNA (crRNA) and transactivating crRNA (tracrRNA) specific to the nucleic acid encoding the MAST4 protein, or a single strand guide RNA including parts of the crRNA and the tracrRNA and hybridizing with the nucleic acid encoding the MAST4 protein.
  • crRNA CRISPR RNA
  • tracrRNA transactivating crRNA
  • the dual RNA and the single strand guide RNA may at least partially hybridize with the polynucleotide encoding the MAST4 protein, and specifically, may hybridize with a region corresponding to “5′-TACCCTGCCGCTGCCGCACC-3′ (SEQ ID NO: 17)” in the polynucleotide sequence encoding the amino acid sequence of MAST4 protein.
  • the guide RNA may be a dual RNA including crRNA and tracrRNA that hybridize with a target sequence selected from the nucleotide sequence encoding the MAST4 protein, or a single strand guide RNA including parts of the crRNA and the tracrRNA and hybridizing with the nucleotide encoding the MAST4 protein.
  • the MAST4 gene which is the target sequence includes a polynucleotide sequence at least partially complementary to the crRNA or sgRNA, and a sequence including a protospacer-adjacent motif (PAM).
  • the PAM may be a sequence well-known in the art, which may have a sequence suitable to be recognized by a nuclease protein.
  • the MAST4 gene targeted by the CRISPR-Cas system may be endogenous DNA or artificial DNA.
  • the nucleotide encoding the MAST4 protein may be specifically endogenous DNA of a eukaryotic cell, and more specifically, endogenous DNA of a chondrocyte.
  • the crRNA or sgRNA may include twenty consecutive polynucleotides complementary to the target DNA.
  • the target DNA of the complementary twenty consecutive polynucleotides may be 5′-TACCCTGCCGCTGCCGCACC-3′ (SEQ ID NO: 17), and may be selected from the sequences marked in bold in SEQ ID NOS: 74, 76, and 77 of Table 6.
  • a nucleic acid encoding the Cas9 protein or the Cas9 protein may be derived from a microorganism of the genus Streptococcus .
  • the microorganism of the genus Streptococcus may be Streptococcus pyogenes .
  • the PAM may mean 5′-NGG-3′ trinucledotide, and the Cas9 protein may further include a nuclear localization signal (NLS) at the C-terminus or N-terminus to enhance the efficiency.
  • NLS nuclear localization signal
  • the eukaryotic cells may be yeast cells, fungal cells, protozoa cells, plant cells, higher plant cells, insect cells, amphibian cells, or mammalian cells.
  • the mammal may vary such as humans, monkeys, cows, horses, pigs, etc.
  • the eukaryotic cells may include cultured cells (in vitro) isolated from an individual, graft cells, in vivo cells, or recombinant cells, but are not limited thereto.
  • the eukaryotic cells isolated from an individual may be eukaryotic cells isolated from an individual the same as an individual into which the product including extracellular matrix produced from the eukaryotic cells is injected. In this case, it is advantageous in that side effects such as unnecessary hyperimmune reactions or rejection reactions including graft-versus-host reaction generated by injecting a product produced from a different individual may be prevented.
  • the eukaryotic cells may be fibroblasts or chondrocytes.
  • the composition for promoting the production of extracellular matrix from the eukaryotic cells and/or the composition for promoting chondrogenesis of chondrocytes may further include TGF- ⁇ 1.
  • TGF- ⁇ 1 The present inventors confirmed that MAST4 expression in human chondrocytes is reduced by TGF- ⁇ 1, and as a result, production of extracellular matrix is promoted. Therefore, to more effectively and easily promote extracellular matrix in MAST4 knockout cells of eukaryotic cells (or chondrocytes), combination treatment with TGF- ⁇ 1 may be advantageous.
  • the MAST4 is a protein derived from a human ( Homo sapiens ) or a mouse ( Musmusculus ), but the same protein may also be expressed in other mammals such as monkeys, cows, horses, etc.
  • the human-derived MAST4 may include all of seven isoforms present in human cells.
  • the seven isoforms may include amino acid sequences of NP_055998.1 (SEQ ID NO: 1), NP_942123.1 (SEQ ID NO: 2), NP_001158136.1 (SEQ ID NO: 3), NP_001277155.1 (SEQ ID NO: 4), NP_001277156.1 (SEQ ID NO: 5), NP_001277157.1 (SEQ ID NO: 6), or NP_001284580.1 (SEQ ID NO: 7), based on NCBI reference sequence, and a protein or a polypeptide having each of the amino acid sequences may be translated from mRNA including polynucleotide sequences of SEQ ID NOS: 8 to 14 each encoding the amino acid sequences of SEQ ID NOS: 1 to in the sequence of NM_015183.2, NM_198828.2, NM_001164664.1, NM_001290226.1, NM
  • the mouse-derived MAST4 may include an amino acid sequence of NP_780380.2 (SEQ ID NO: 15), based on NCBI reference sequence, and a protein or a polypeptide having the amino acid sequence may be translated from mRNA including a polynucleotide sequence of SEQ ID NO: 16 encoding the amino acid sequence of SEQ ID NO: 15 in the sequence of NM_175171.3.
  • amino acid sequence or a polynucleotide sequence having biologically equivalent activity may also be regarded as the MAST4 protein or mRNA thereof.
  • the MAST4 protein may include any one sequence of SEQ ID NOS: 1 to 7 and 15, and the nucleotide sequence encoding the MAST4 protein may include any one sequence of SEQ ID NOS: 8 to 14 and 16.
  • the MAST4 protein or polypeptide may include an amino acid sequence having 60% or more, for example, 70% or more, 80% or more, 90% or more, 95% or more, 99% or more, or 100% sequence identity to SEQ ID NOS: 1 to 7 and 15. Further, the MAST4 protein may have an amino acid sequence having modification of 1 or more amino acids, 2 or more amino acids, 3 or more amino acids, 4 or more amino acids, 5 or more amino acids, 6 or more amino acids, or 7 or more amino acids in the amino acid sequences of SEQ ID NOS: 1 to 7 and 15.
  • Each polynucleotide encoding MAST4 may have a sequence having 60% or more, for example, 70% or more, 80% or more, 90% or more, 95% or more, 99% or more, or 100% sequence identity to SEQ ID NOS: 8 to 14 and 16. Further, the polynucleotide encoding MAST4 may be a polynucleotide having a different sequence of 1 or more nucleotides, 2 or more nucleotides, 3 or more nucleotides, 4 or more nucleotides, 5 or more nucleotides, 6 or more nucleotides, or 7 or more nucleotides in the sequences of SEQ ID NOS: 8 to 14 and 16.
  • the present inventors first demonstrated that production of extracellular matrix is increased and chondrogenesis is promoted by inhibiting MAST4 gene expression in chondrocytes.
  • the composition for promoting production of an extracellular matrix from eukaryotic cells or the composition for promoting chondrogenesis of chondrocytes of the present disclosure may prevent or treat a joint disease, or improve symptoms thereof.
  • the composition for promoting the production of extracellular matrix from the eukaryotic cells and/or the composition for promoting chondrogenesis of chondrocytes of the present disclosure may be to induce chondrogenesis.
  • the composition for promoting the production of extracellular matrix from the eukaryotic cells may be used for tissue regeneration or anti-aging.
  • the tissue regeneration refers to regeneration of the skin damaged or deformed by wounds, burns, injury, aging, chronic inflammation, diseases, genetic factors, etc., and includes all those used for medical or skin cosmetic purposes.
  • the damage or deformation is caused by the loss or reduced production of extracellular matrix in a tissue, or impossibility of recovery of the extracellular matrix in the tissue by the above factors, and the damage or deformation means symptoms improved, alleviated, recovered, or cured by promoting the production of extracellular matrix by the composition of the present disclosure.
  • the composition of the present disclosure may promote the production of extracellular matrix, thereby preventing or recovering reduced elasticity, deformation, or damage of tissues caused by aging.
  • composition for tissue regeneration or anti-aging may be used as a component of fillers or collagen supplement cosmetics.
  • composition for tissue regeneration or anti-aging may be used as a component of functional cosmetics to block the adsorption of fine dust or minerals.
  • composition for promoting the production of extracellular matrix from the eukaryotic cells or the composition for promoting chondrogenesis of chondrocytes of the present disclosure may further include a pharmaceutically acceptable salt or carrier.
  • pharmaceutically acceptable salt means any organic or inorganic addition salt of the compound in the composition of the present disclosure, whose concentration has effective action because it is relatively non-toxic and harmless to patients and whose side effects do not degrade the beneficial efficacy of the composition of the present disclosure. These salt may be selected from any one known to those skilled in the art.
  • composition of the present disclosure may further include a pharmaceutically acceptable carrier.
  • the composition including the pharmaceutically acceptable carrier may have various formulations for oral or parenteral administration.
  • the composition may be prepared using commonly used diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrating agents, surfactants, etc.
  • Solid formulations for oral administration may include tablets, pills, powders, granules, capsules, troches, etc., and these solid formulations may be prepared by mixing one or more compounds of the present disclosure with at least one excipient such as starch, calcium carbonate, sucrose, lactose, or gelatin.
  • lubricants such as magnesium stearate, talc, etc.
  • Liquid formulations for oral administration may include suspensions, liquids for internal use, emulsions, syrups, etc.
  • excipients such as wetting agents, sweeteners, flavoring agents, preservatives, etc. may be included, in addition to commonly used simple diluents such as water, liquid paraffin, etc.
  • Formulations for parenteral administration may include sterilized aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried preparations, suppositories, etc.
  • the non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, injectable esters such as ethyl oleate, etc.
  • As a base of a suppository witepsol, macrogol, Tween 61, cocoa butter, laurin butter, glycerol, gelatin, etc. may be used.
  • An aspect provides a method of preventing, treating, or improving a joint disease, the method including administering the composition to a subject.
  • Another aspect provides a method of producing an extracellular matrix, the method including contacting eukaryotic cells with the composition for producing the extracellular matrix from eukaryotic cells of the present disclosure.
  • the eukaryotic cells may be isolated from a subject.
  • the eukaryotic cells may be chondrocytes.
  • the contacting with the eukaryotic cells may include co-transfecting or serial-transfecting the composition into the eukaryotic cells.
  • various methods known in the art such as microinjection, electroporation, DEAE-dextran treatment, lipofection, nanoparticle-mediated transfection, protein transduction domain-mediated transduction, virus-mediated gene delivery, and PEG-mediated transfection in protoplast, etc. may be used, but are not limited thereto.
  • the contacting with the eukaryotic cells may include culturing the eukaryotic cells in the presence of the composition.
  • the culturing includes culturing in the presence of a chondrogenic inducer.
  • the method of producing the extracellular matrix of the present disclosure may further include isolating the extracellular matrix from the contacting product.
  • the method of producing the extracellular matrix may include contacting chondrocytes with the composition for promoting chondrogenesis of the present disclosure.
  • Still another aspect provides a method of forming a cartilage, the method including contacting chondrocytes with the composition for promoting chondrogenesis of the present disclosure.
  • the chondrocytes may be isolated from a subject.
  • the chondrocytes may be derived from a subject to be transplanted with the produced cartilage.
  • Still another aspect provides a method of producing ECM, the method including culturing eukaryotic cells having increased extracellular matrix productivity of the present disclosure to produce ECM; and isolating ECM from the culture.
  • the culturing may be culturing in the presence of a chondrogenic inducer.
  • the chondrogenic inducer may be BMP.
  • a composition for promoting production of an extracellular matrix according to an aspect may be injected into a subject who requires supply of the extracellular matrix, thereby preventing or treating diseases including a joint disease, and improving symptoms thereof, and the composition may be applied to a method of efficiently producing the extracellular matrix from eukaryotic cells.
  • a composition for promoting chondrogenesis of chondrocytes may be injected into a subject, thereby preventing or treating diseases including a joint disease, and improving symptoms thereof.
  • the composition may promote chondrogenesis of chondrocytes isolated from the subject, and thus it may be applied to a method of efficiently producing various components including extracellular matrice which are produced by chondrogenesis.
  • the extracellular matrix may be efficiently produced from eukaryotic cells.
  • FIG. 1 illustrates a method of preparing MAST4 knockout mice using a CRISPR/Cas9 system
  • FIG. 2A shows RT-PCR results of examining changes in expression levels of respective genes in MAST4 knockout mouse type A and B, and FIG. 2B shows protein expression patterns in MAST4 knockout mice;
  • FIG. 3 shows identification of MAST4 knockout in C3H10T1/2 cells in which MAST4 was knocked out using the CRISPR/Cas9 system
  • FIG. 4 shows RT-PCR results of examining changes in expression levels of respective genes in C3H10T1/2 cells in which MAST4 was knocked out using the CRISPR/Cas9 system;
  • FIG. 5 shows RT-PCR results of examining changes in expression levels of respective genes in a micromass culture to confirm chondrogenesis
  • FIG. 6 shows alcian blue staining results of examining a difference in cartilage differentiation in C3H10T1/2 cells in which MAST4 was knocked out using the CRISPR/Cas9 system;
  • FIG. 7 shows sequence information of target genes used to knockout MAST4 of human cells
  • FIG. 8A shows human chondrocytes in which MAST4 was knocked out using siRNA
  • FIG. 8B shows expression levels of extracellular matrix factors in human chondrocytes in which MAST4 was knocked out using the CRISPR/Cas9 system
  • FIG. 9A shows changes in the expression level of MAST4 after treatment of human primary chondrocytes with TGF- ⁇ 1, and expression levels of extracellular matrix factors thereby
  • FIG. 9B shows changes in the expression level of MAST4 after treatment of human primary chondrocytes with TGF- ⁇ 1, and expression levels of extracellular matrix factors thereby;
  • FIG. 10 shows chondrogenesis and regeneration effects in the tibia of the MAST4 knockout mouse.
  • MAST4 knockout mice were prepared using a CRISPR/Cas9 system.
  • pX330-U6-Chimeric_BB-CBh-hSpCas9 (Addgene, #42230), donated by Dr. Feng Zhang (Cong et al., 2013), was used as a plasmid capable of expressing Cas9 mRNA and guide RNA.
  • MAST4 is a large protein of 7 kb or more, it was designed such that the gene editing was allowed to target two parts, exon 1 and exon 15.
  • a guide RNA sequence targeting exon 1 of MAST4 is 5′-GGAAACTCTGTCGGAGGAAGGGG-3′ and a sequence targeting exon 15 is 5′-GGCACAAAGAGTCCCGCCAGAGG-3′.
  • the guide RNA sequence was used to prepare oligomers as in MAST4 CRISPR oligomers of the following Table in accordance with the manufacturer's protocol (http://crispr.mit.edu/, Zhang Feng Lab), and each oligomer was inserted into a px330 plasmid to clone two plasmids targeting exon 1 and exon 15, respectively.
  • mice 5 IU of pregnant mare serum gonadotrophin (PMSG; Prospec, cat. No. HOR-272) was administered to a C57BL/6J female mouse 2 days before mating, and after 47 hours, 5 IU of humanchorionic gonadotrophin (hCG, Prospec, cat. HOR-250) was administered thereto . . . . Thereafter, the mouse was mated with C57BL/6J male mouse, and embryos were obtained from fallopian tubes. A microinjection mixture including 5 ng/ ⁇ l of the prepared plasmid and 10 ng of ssDNA donor was injected into the pronuclei of the embryos at a one-cell-stage with reference to an existing standard protocol (Gordon and Ruddle, 1981). The injected one-cell-embryos were transferred to pseudopregnant ICR mice.
  • PMSG pregnant mare serum gonadotrophin
  • hCG humanchorionic gonadotrophin
  • Phenotypic analysis of born mice was performed for exon 1 and exon 15. Finally, two types of MAST4 knockout mice were obtained. Information about the two types of MAST4 knockout mice, type A and type B are shown as in FIG. 1 and the following Table 2 (5′ ⁇ 3′).
  • RNA-sequencing was performed for respective genes.
  • RNA-sequencing was performed by Theragen Etex.
  • mRNA was isolated from 2 ⁇ g of total RNA extracted from the mouse of each group using oligo(dT). After fragmentation of the mRNA, single-stranded cDNA was synthesized through random hexamer priming. This single-strand cDNA was used as a template to synthesize a second strand, thereby synthesizing a double-stranded cDNA. To prepare blunt-ends, end repair was performed, and to ligate an adapter, A-tailing and adapter ligation were performed. Thereafter, cDNA library was amplified by polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • a concentration and size of the final product were examined using 2100 BioAnalyzer.
  • the produced library was finally quantified using a KAPA library quantification kit, and then sequence interpretation was performed using Hiseq2500.
  • sequence interpretation was performed using Hiseq2500.
  • filtering was performed such that reads containing 10% or more of bases marked as ‘N’s in the sequence information or reads containing 40% or more of bases less than Q20 were removed, and reads whose average quality is Q20 or less were also removed.
  • the whole filtering process was performed using the in-house program.
  • the filtered sequences were aligned to a reference genome sequence (hg19) of the corresponding species using STAR v2.4.0b (Dobin et al, 2013).
  • Expression level was measured using Cufflinks v2.1.1 (Trapnell C. et al, 2010), and the calculated expression values were expressed as fragments read per kilobase of exon per million fragments mapped (FPKM).
  • Ensemble 72 was used as a genetic information database, and a non-coding gene region was removed with expression-mask option. To increase measurement accuracy of the expression levels, multi-read-correction and frag-bias-correct options were additionally used, and all other options were set to default values.
  • Example 1-1 To more specifically examine changes in the extracellular matrix as a cartilage component in MAST4 knockout mice prepared in Example 1-1, a part of genes showing changes in the expression in the RNA sequencing results of Example 1-2 was selected and subjected to RT-PCR.
  • RT-PCR was performed using a set of primers of the following Table 4 and AccuPower PCR premix (BIONEER, Korea) according to the manufacturer's instructions.
  • Col2a1 which is known as a chondrocyte marker was stained with fluorescence in the mouse tibia.
  • the tibia tissue was obtained from the mouse model of Example 1-1, and fixed with 4% paraformaldehyde (PFA, Wako, Osaka, JAPAN) in 0.01 M phosphate buffer saline (PBS, pH 7.4) at 4° C. overnight.
  • the tissue was decalcified with 10% EDTA, and embedded in paraffin (Leica Biosystems, MO, USA), and sectioned 6 mm in thickness.
  • the sample slide was stained with hematoxylin and eosin, and the tissue section was incubated with a primary antibody at 4° C. overnight.
  • the primary antibody targets ColI2a1 (Abcam, Cambridge, UK).
  • the tissue section was sequentially incubated with AlexaFluor 488 (Invitrogen, CA, USA) at room temperature for 2 hours. Each image was obtained using a confocal microscope LSM700 (Carl Zeiss, Oberkochen, Germany), and a representative sample section was stained with freshly prepared Russell-Movatmodified pentachrome (American MasterTech, CA, USA).
  • FIG. 10 is an enlargement of a specific area of the observed sample, where Col2a1 (fluorescent green zone/grey background zone) was significantly increased in the tibia of the MAST4 knockout mouse model.
  • TOPRO-3 areas marked by red dots/gray dots shows staining of the nuclei of chondrocytes. Therefore, it was confirmed that chondrogenesis and cartilage regeneration may be promoted by MAST4 knockout.
  • MAST4 knockout cells were prepared using the CRISPR/Cas9 system.
  • C3H/10T1/2, Clone 8 which is a mouse-derived fibroblast cell and is able to differentiate into chondrocytes was purchased ((C3H10T1/2 cell) provided by prof. Seon-Yong Jeong's lab, Department of Medical Genetics, School of Medicine, Ajou University).
  • lentiCRISPR v2 (Plasmid #52961), pVSVg (AddGene 8454), and psPAX2 (AddGene 12260) were purchased from Addgene, and oligomers of the following Table 5 were used to insert guide RNA targeting exon 1 of mouse MAST4 gene (ENSMUSG00000034751) into LentiCRISPR v2 plasmid according to the manufacturer's instructions (lentiCRISPRv2 and lentiGuide oligo cloning protocol), thereby preparing a plasmid expressing guide RNA and Cas9 enzyme at the same time (as a control group, a plasmid having no guideRNA and expressing only Cas9 was used).
  • This method is a lentivirus-based CRISPR knockout method.
  • the three plasmids prepared above (LentiCRISPR v2 (+guide RNA): guide RNA+Cas9 expressing plasmid, pVSVg: Virus envelop plasmid, psPAX2: Virus packaging plasmid) were transfected into 293T cells using a polyethyenimine (PEI) reagent. 18 hours later, the medium was replaced with a fresh medium, and only the medium was collected, and viruses were obtained using a 0.45 ⁇ m filter. The obtained viruses were transfected into a 6-well dish to which C3H10T/12 was seeded.
  • PEI polyethyenimine
  • PCR was performed using primers specifically amplifying exon 1 (F: 5′->3′ CTGTGGTCCAACCTCTGTCA, R: 5′->3′ ATCGGCTCAGTGACACTTCC).
  • the amplified PCR products were analyzed by the sequencing company.
  • cells in which gene editing by frameshift was identified were used in the experiment, together with control cells.
  • the sequences targeted by the prepared guide RNA were are in bold in Table 5.
  • deletion of two nucleotides occurred in mouse MAST4 exon 1, indicating frameshift induction.
  • Example 2-2 To evaluate chondrogenic ability of the MAST4 knockout cells of Example 2-2, micromass culture was performed.
  • MAST4 knockout cells were prepared as in Example 2-1, and micromass culture was performed with reference to a known method (Differentiation and Mineralization of Murine Mesenchymal C3H10T1/2 Cells in Micromass Culture, 2010, Rani Roy).
  • 10 ⁇ l of a medium containing total 10 5 fibroblast cells were put in the center of each well of a 12-well plate, and incubated for 2 hours.
  • 1 ml of DMEM containing 10% FBS was added to each well.
  • 100 ng/ml, 500 ng/ml, or 1000 ng/ml of BMP2 was added to each culture depending on the purpose of cartilage induction, respectively. Thereafter, the medium was replaced with a fresh medium every three days.
  • Example 3-2 With regard to the overexpression of the respective extracellular matrix-associated genes observed in Example 3-2, to examine whether or not the expression was actually increased at the level of isolated proteins, not at the gene expression level, alcian blue staining was performed.
  • plates of cells corresponding to each date were washed twice with PBS and fixed for 15 minutes by adding 1 ml of 4% paraformaldehyde. Then, 1 ml of 1% alcian blue 8-GX (Sigma-Aldrich, A5268) dissolved in 0.1 N HCl (pH 1.0) was added and stained overnight. After washing twice with 500 ⁇ l of 0.1 N HCl, images were obtained.
  • MAST4 siRNA(h) sc-106201; Santa Cruz biotechnology
  • FIG. 8A MAST4 expression was knocked-out by the CRISPR/Cas9 system.
  • MAST4 siRNA was transfected using a Lipofectamine RNAiMAX transfection reagent of ThermoFisher SCIENTIFIC, and information of primers used herein is as described in the following Table 6.
  • siRNA transfection was performed by a reverse transfection technique in which cell planting and transfection are performed at the same time, and a transfection reagent was Lipofectamine RNAiMAX transfection reagent of ThermoFisher SCIENTIFIC.
  • a transfection reagent was Lipofectamine RNAiMAX transfection reagent of ThermoFisher SCIENTIFIC.
  • 15 nM of MAST4 siRNA and 4.5 ⁇ l of Lipofectamine RNAiMax were mixed in 40 ⁇ l of GibcoTM Opti-MEMTM, and incubated for 15 minutes. Thereafter, human primary chondrocytes of 1.5 ⁇ 10 5 cell/well were plated together with 2 ml of a medium (FBS 10%) containing no gentamicin in a 6-well plate (ColI coated plate), and the siRNA mixture was added thereto.
  • FBS 10% fetal bovine serum
  • Human primary chondrocytes were cultured in a collagen I-coated flask (175, Col I Straight Vent 356487, Corning) under conditions of DMEM (17-205-CVR Corning), FBS Qualified (USA origin 500 mL 26140-079, Gibco), L-glutamine (200 mM) (100 ⁇ 25030-081, Gibco), and gentamicin (5 ug/ml) (10 mL 15700-060, Thermofisher).
  • Knockout was performed by targeting 20 nt on the genome of MAST4 (target sequences are marked in bold), and specifically, #1 and #3 target Exon5, and #2 targets Exon 8. #1 and #3 were prepared in the reverse direction, and #2 was prepared in the forward direction.
  • the human MAST4 gene used in the preparation of CRISPR/Cas9 system was with reference to MAST4 ENSG00000069020 (http://asia.ensembl.org/). Information of targeted Exon sequences and NGG PAM sequences (grey box) on which CRISPR deletion occurred are shown in detail in FIG. 7 .
  • hMAST4 CR#1 F (SEQ ID NO: 73) 5′-TAATACGACTCACTATAG GAGTGTGGTCGAGGCAATGC-3′ hMAST4 CR#1 R (SEQ ID NO: 74) 5′-TTCTAGCTCTAAAAC GCATTGCCTCGACCACACTC -3′ hMAST4 CR#2 F (SEQ ID NO: 75) 5′-TAATACGACTCACTATAG GTAACTCGTCTGGTGTTGGT-3′ hMAST4 CR#2 R (SEQ ID NO: 76) 5′-TTCTAGCTCTAAAAC ACCAACACCAGACGAGTTAC -3′ hMAST4 CR#3 F (SEQ ID NO: 77) 5′-TAATACGACTCACTATAG AGCAACCGGAAAAGCTTAAT -3′ hMAST4 CR#3 R (SEQ ID NO: 78) 5′-TTCTAGCTCTAAAAC ATTAAGCTTTTCCGGTTG
  • Example 4-1 It was examined whether suppression of MAST4 expression as confirmed in Example 4-1 was induced by TGF- ⁇ 1 and expression of extracellular matrix factors was affected thereby.
  • Example 4-1 human primary chondrocytes of Example 4-1 were treated with TGF- ⁇ 1, and an expression level thereof was measured by RT-PCR as in Examples 1-2 and 1-3 and Western blotting.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Rheumatology (AREA)
  • Virology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Mycology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a composition for producing an extracellular matrix from a eukaryotic cell, the composition comprising a polypeptide or compound capable of specifically binding to a microtubule associated serine/threonine kinase family member 4 (MAST4) protein or a fragment thereof or a polynucleotide, polypeptide or compound capable of specifically binding to a nucleic acid coding for the MAST4 protein or a fragment thereof, and a composition for promoting chondrogenesis, comprising the same composition.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • The present application is a continuation of U.S. patent application Ser. No. 16/492,477, filed Sep. 9, 2019, which is a National Stage Entry of PCT Patent Application No. PCT/US2020/025689, filed Mar. 8, 2018, each of which is hereby incorporated by reference in their entirety.
  • TECHNICAL FIELD
  • The present disclosure relates to a composition for producing an extracellular matrix from eukaryotic cells using Microtubule Associated Serine/Threonine Kinase Family Member 4 (MAST4) gene, a method of producing the extracellular matrix from the eukaryotic cells, and a composition for promoting chondrogenesis, the composition including the above composition.
  • BACKGROUND ART
  • Since most bone formation begins from a cartilaginous template, successful skeletal development requires a perfect cooperation in both structural and molecular aspects. Articular cartilage is a highly organized tissue, and the mechanism of in-vivo chondrogenesis involved therein is still unknown. Interactions between collagen microfibers and other extracellular matrix component proteins are known to maintain the structural integrity of the cartilage, but the signaling mechanisms regulating their complex processes have not yet been clearly revealed. Therefore, identification of the existence and function of a master regulator that leads chondrogenesis is not only academically meaningful, but also contributes to the public health as well as to development of innovative therapeutics.
  • Microtubule associated serine/threonine kinase (MAST) 4 is known to be expressed in cartilage (BMC Genomics 2007, 8: 165), but its role has not been clearly elucidated. CN 105636614 discloses that MAST4 may be used for the treatment of cartilage, but this is based only on the stochastic results of MAST4 expression in the cartilage, and does not elucidate specific roles thereof.
  • The present inventors have found MAST4 as a novel central regulator that is involved in chondrogenesis, and provide a source technology for the development of substances that modulate the activity of MAST4.
  • PRIOR ART DOCUMENTS
    • Non-Patent Document: BMC Genomics 2007, 8:165
    • Patent Document: CN 105636614
    DESCRIPTION OF EMBODIMENTS Technical Problem
  • An aspect provides a composition for promoting production of an extracellular matrix from eukaryotic cells, the composition including a compound capable of specifically binding to Microtubule Associated Serine/Threonine Kinase Family Member 4 (MAST4) protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • Another aspect provides a composition for promoting chondrogenesis of chondrocytes, the composition including a compound capable of specifically binding to MAST4 protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • Still another aspect provides a method of producing an extracellular matrix from eukaryotic cells, the method including contacting the eukaryotic cells with the composition for promoting production of the extracellular matrix from eukaryotic cells.
  • Solution to Problem
  • According to an aspect, provided is a composition for promoting production of an extracellular matrix from eukaryotic cells, the composition including a compound capable of specifically binding to Microtubule Associated Serine/Threonine Kinase Family Member 4 (MAST4) protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • According to another aspect, provided is a composition for promoting chondrogenesis of chondrocytes, the composition including a compound capable of specifically binding to MAST4 protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
  • In a specific embodiment, the compound capable of specifically binding to the MAST4 protein or the fragment thereof, or the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof includes those capable of at least partially binding to the protein or the fragment thereof, or the nucleic acid. Here, the compound may be a chemically synthesized compound, polypeptide, or polynucleotide, or a combination thereof. These compounds may inhibit activity or expression of MAST4 protein.
  • In a specific embodiment, the composition for promoting production of extracellular matrix from eukaryotic cells may be a composition for promoting chondrogenesis from eukaryotic cells.
  • In the composition, the activity inhibitor of MAST4 protein or the expression inhibitor of MAST4 protein includes any one, as long as it is able to inhibit expression of MAST4 gene or activity of MAST4 protein. The activity inhibitor or the expression inhibitor may be a polynucleotide complementary to the entire or a part of the MAST4 gene. The polynucleotide sequence may be RNA, DNA, or a hybrid thereof.
  • In a specific embodiment, the activity inhibition of the MAST4 protein may be kinase activity inhibition of the MAST4 protein.
  • MAST4 is a kinase capable of phosphorylating Ser or Thr of a target substrate, and the kinase activity inhibition of the MAST4 protein means blocking of phosphorylation of a target substrate of MAST4, specifically, blocking of phosphorylation of Ser or Thr.
  • In a specific embodiment, the polypeptide specifically binding to MAST4 protein or the fragment thereof, or the polypeptide specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof may be an antibody or an antigen-binding fragment thereof.
  • The term “antibody” means a specific immunoglobulin directed against an antigenic site. MAST4 gene is cloned into an expression vector to obtain the MAST4 protein encoded by the gene, and the antibody may be prepared from the protein according to a common method in the art. A type of the antibody includes a polyclonal antibody or a monoclonal antibody, and includes all immunoglobulin antibodies. The antibody includes not only complete forms having two full-length light chains and two full-length heavy chains but also functional fragments of antibody molecules which have a specific antigen binding site (binding domain) directed against an antigenic site to retain an antigen-binding function, although they do not have the intact complete antibody structure having two light chains and two heavy chains.
  • The term “polynucleotide” may be used in the same meaning as a nucleotide or a nucleic acid, unless otherwise mentioned, and refers to a deoxyribonucleotide or a ribonucleotide. The polynucleotide may include an analog of a natural nucleotide and an analog having a modified sugar or base moiety, unless otherwise mentioned. The polynucleotide may be modified by various methods known in the art, as needed. Examples of the modification may include methylation, capping, substitution of a natural nucleotide with one or more homologues, and modification between nucleotides, for example, modification to uncharged linkages (e.g., methylphosphonate, phosphotriester, phosphoroamidate, carbamate, etc.) or charged linkages (e.g., phosphorothioate, phosphorodithioate, etc.).
  • In a specific embodiment, as the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof, the polynucleotide capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof may be microRNA (miRNA), small interfering RNA (siRNA), short hairpin RNA (shRNA), Piwi-interacting RNA (piRNA), small nuclear RNA (snRNA), or antisense oligonucleotide, each specific to the nucleic acid encoding the MAST4 protein or the fragment thereof, or a combination thereof.
  • In another specific embodiment, the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof may include the polynucleotide capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof, and may be CRISPR-Cas including guide RNA specific to the nucleic acid encoding the MAST4 protein or the fragment thereof.
  • In a specific embodiment, the Cas may be Cas9.
  • Clustered Regularly Interspaced Short Palindromic Repeats (CRISPRs) mean loci including many short direct repeats found in the genome of bacteria or archaea, of which genetic sequences are revealed. The CRISPR-Cas system includes Cas9 as an essential protein element which forms a complex with guide RNA (specifically, two RNAs, called CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA), included in guide RNA), and it serves as an active endonuclease.
  • In a specific embodiment, for the CRISPR-Cas system to specifically act on the target gene MAST4, the guide RNA may have a form of a dual RNA including CRISPR RNA (crRNA) and transactivating crRNA (tracrRNA) specific to the nucleic acid encoding the MAST4 protein, or a single strand guide RNA including parts of the crRNA and the tracrRNA and hybridizing with the nucleic acid encoding the MAST4 protein. The dual RNA and the single strand guide RNA may at least partially hybridize with the polynucleotide encoding the MAST4 protein, and specifically, may hybridize with a region corresponding to “5′-TACCCTGCCGCTGCCGCACC-3′ (SEQ ID NO: 17)” in the polynucleotide sequence encoding the amino acid sequence of MAST4 protein.
  • Specifically, the guide RNA may be a dual RNA including crRNA and tracrRNA that hybridize with a target sequence selected from the nucleotide sequence encoding the MAST4 protein, or a single strand guide RNA including parts of the crRNA and the tracrRNA and hybridizing with the nucleotide encoding the MAST4 protein. The MAST4 gene which is the target sequence includes a polynucleotide sequence at least partially complementary to the crRNA or sgRNA, and a sequence including a protospacer-adjacent motif (PAM). The PAM may be a sequence well-known in the art, which may have a sequence suitable to be recognized by a nuclease protein. The MAST4 gene targeted by the CRISPR-Cas system may be endogenous DNA or artificial DNA. The nucleotide encoding the MAST4 protein may be specifically endogenous DNA of a eukaryotic cell, and more specifically, endogenous DNA of a chondrocyte.
  • In a specific embodiment, the crRNA or sgRNA may include twenty consecutive polynucleotides complementary to the target DNA. The target DNA of the complementary twenty consecutive polynucleotides may be 5′-TACCCTGCCGCTGCCGCACC-3′ (SEQ ID NO: 17), and may be selected from the sequences marked in bold in SEQ ID NOS: 74, 76, and 77 of Table 6. A nucleic acid encoding the Cas9 protein or the Cas9 protein may be derived from a microorganism of the genus Streptococcus. The microorganism of the genus Streptococcus may be Streptococcus pyogenes. The PAM may mean 5′-NGG-3′ trinucledotide, and the Cas9 protein may further include a nuclear localization signal (NLS) at the C-terminus or N-terminus to enhance the efficiency.
  • In the composition for promoting production of an extracellular matrix from eukaryotic cells of the present disclosure, the eukaryotic cells may be yeast cells, fungal cells, protozoa cells, plant cells, higher plant cells, insect cells, amphibian cells, or mammalian cells. The mammal may vary such as humans, monkeys, cows, horses, pigs, etc. The eukaryotic cells may include cultured cells (in vitro) isolated from an individual, graft cells, in vivo cells, or recombinant cells, but are not limited thereto. The eukaryotic cells isolated from an individual may be eukaryotic cells isolated from an individual the same as an individual into which the product including extracellular matrix produced from the eukaryotic cells is injected. In this case, it is advantageous in that side effects such as unnecessary hyperimmune reactions or rejection reactions including graft-versus-host reaction generated by injecting a product produced from a different individual may be prevented.
  • In a specific embodiment, the eukaryotic cells may be fibroblasts or chondrocytes.
  • In a specific embodiment, the composition for promoting the production of extracellular matrix from the eukaryotic cells and/or the composition for promoting chondrogenesis of chondrocytes may further include TGF-β1. The present inventors confirmed that MAST4 expression in human chondrocytes is reduced by TGF-β1, and as a result, production of extracellular matrix is promoted. Therefore, to more effectively and easily promote extracellular matrix in MAST4 knockout cells of eukaryotic cells (or chondrocytes), combination treatment with TGF-β1 may be advantageous.
  • The MAST4 is a protein derived from a human (Homo sapiens) or a mouse (Musmusculus), but the same protein may also be expressed in other mammals such as monkeys, cows, horses, etc.
  • The human-derived MAST4 may include all of seven isoforms present in human cells. The seven isoforms may include amino acid sequences of NP_055998.1 (SEQ ID NO: 1), NP_942123.1 (SEQ ID NO: 2), NP_001158136.1 (SEQ ID NO: 3), NP_001277155.1 (SEQ ID NO: 4), NP_001277156.1 (SEQ ID NO: 5), NP_001277157.1 (SEQ ID NO: 6), or NP_001284580.1 (SEQ ID NO: 7), based on NCBI reference sequence, and a protein or a polypeptide having each of the amino acid sequences may be translated from mRNA including polynucleotide sequences of SEQ ID NOS: 8 to 14 each encoding the amino acid sequences of SEQ ID NOS: 1 to in the sequence of NM_015183.2, NM_198828.2, NM_001164664.1, NM_001290226.1, NM_001290227.1, NM_001290228.1, or NM_001297651.1.
  • The mouse-derived MAST4 may include an amino acid sequence of NP_780380.2 (SEQ ID NO: 15), based on NCBI reference sequence, and a protein or a polypeptide having the amino acid sequence may be translated from mRNA including a polynucleotide sequence of SEQ ID NO: 16 encoding the amino acid sequence of SEQ ID NO: 15 in the sequence of NM_175171.3.
  • An amino acid sequence or a polynucleotide sequence having biologically equivalent activity, even though it is not identical to the amino acid sequences of SEQ ID NOS: 1 to 7 and 15 and the polynucleotide sequences of SEQ ID NOs: 8 to 14 and 16, may also be regarded as the MAST4 protein or mRNA thereof.
  • Therefore, in a specific embodiment, the MAST4 protein may include any one sequence of SEQ ID NOS: 1 to 7 and 15, and the nucleotide sequence encoding the MAST4 protein may include any one sequence of SEQ ID NOS: 8 to 14 and 16.
  • The MAST4 protein or polypeptide may include an amino acid sequence having 60% or more, for example, 70% or more, 80% or more, 90% or more, 95% or more, 99% or more, or 100% sequence identity to SEQ ID NOS: 1 to 7 and 15. Further, the MAST4 protein may have an amino acid sequence having modification of 1 or more amino acids, 2 or more amino acids, 3 or more amino acids, 4 or more amino acids, 5 or more amino acids, 6 or more amino acids, or 7 or more amino acids in the amino acid sequences of SEQ ID NOS: 1 to 7 and 15.
  • Each polynucleotide encoding MAST4 may have a sequence having 60% or more, for example, 70% or more, 80% or more, 90% or more, 95% or more, 99% or more, or 100% sequence identity to SEQ ID NOS: 8 to 14 and 16. Further, the polynucleotide encoding MAST4 may be a polynucleotide having a different sequence of 1 or more nucleotides, 2 or more nucleotides, 3 or more nucleotides, 4 or more nucleotides, 5 or more nucleotides, 6 or more nucleotides, or 7 or more nucleotides in the sequences of SEQ ID NOS: 8 to 14 and 16.
  • The present inventors first demonstrated that production of extracellular matrix is increased and chondrogenesis is promoted by inhibiting MAST4 gene expression in chondrocytes.
  • Therefore, in a specific embodiment, the composition for promoting production of an extracellular matrix from eukaryotic cells or the composition for promoting chondrogenesis of chondrocytes of the present disclosure may prevent or treat a joint disease, or improve symptoms thereof.
  • Further, in a specific embodiment, the composition for promoting the production of extracellular matrix from the eukaryotic cells and/or the composition for promoting chondrogenesis of chondrocytes of the present disclosure may be to induce chondrogenesis.
  • Further, in a specific embodiment, the composition for promoting the production of extracellular matrix from the eukaryotic cells may be used for tissue regeneration or anti-aging.
  • The tissue regeneration refers to regeneration of the skin damaged or deformed by wounds, burns, injury, aging, chronic inflammation, diseases, genetic factors, etc., and includes all those used for medical or skin cosmetic purposes. The damage or deformation is caused by the loss or reduced production of extracellular matrix in a tissue, or impossibility of recovery of the extracellular matrix in the tissue by the above factors, and the damage or deformation means symptoms improved, alleviated, recovered, or cured by promoting the production of extracellular matrix by the composition of the present disclosure.
  • As a tissue including the skin ages, the production of extracellular matrix decreases, resulting in reduced elasticity of the tissue, and the tissue is easily deformed or damaged by external stimuli, and its recovery becomes slow. Accordingly, the composition of the present disclosure may promote the production of extracellular matrix, thereby preventing or recovering reduced elasticity, deformation, or damage of tissues caused by aging.
  • In another specific embodiment, the composition for tissue regeneration or anti-aging may be used as a component of fillers or collagen supplement cosmetics. In still another specific embodiment, the composition for tissue regeneration or anti-aging may be used as a component of functional cosmetics to block the adsorption of fine dust or minerals.
  • The composition for promoting the production of extracellular matrix from the eukaryotic cells or the composition for promoting chondrogenesis of chondrocytes of the present disclosure may further include a pharmaceutically acceptable salt or carrier.
  • The term “pharmaceutically acceptable salt” means any organic or inorganic addition salt of the compound in the composition of the present disclosure, whose concentration has effective action because it is relatively non-toxic and harmless to patients and whose side effects do not degrade the beneficial efficacy of the composition of the present disclosure. These salt may be selected from any one known to those skilled in the art.
  • The composition of the present disclosure may further include a pharmaceutically acceptable carrier. The composition including the pharmaceutically acceptable carrier may have various formulations for oral or parenteral administration. When formulated, the composition may be prepared using commonly used diluents or excipients such as fillers, extenders, binders, wetting agents, disintegrating agents, surfactants, etc. Solid formulations for oral administration may include tablets, pills, powders, granules, capsules, troches, etc., and these solid formulations may be prepared by mixing one or more compounds of the present disclosure with at least one excipient such as starch, calcium carbonate, sucrose, lactose, or gelatin. Moreover, in addition to simple excipients, lubricants such as magnesium stearate, talc, etc. may be used. Liquid formulations for oral administration may include suspensions, liquids for internal use, emulsions, syrups, etc. Various excipients such as wetting agents, sweeteners, flavoring agents, preservatives, etc. may be included, in addition to commonly used simple diluents such as water, liquid paraffin, etc.
  • Formulations for parenteral administration may include sterilized aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried preparations, suppositories, etc. The non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, injectable esters such as ethyl oleate, etc. As a base of a suppository, witepsol, macrogol, Tween 61, cocoa butter, laurin butter, glycerol, gelatin, etc. may be used.
  • An aspect provides a method of preventing, treating, or improving a joint disease, the method including administering the composition to a subject.
  • Another aspect provides a method of producing an extracellular matrix, the method including contacting eukaryotic cells with the composition for producing the extracellular matrix from eukaryotic cells of the present disclosure.
  • In a specific embodiment, the eukaryotic cells may be isolated from a subject. In a specific embodiment, the eukaryotic cells may be chondrocytes.
  • In a specific embodiment, the contacting with the eukaryotic cells may include co-transfecting or serial-transfecting the composition into the eukaryotic cells. To effectively deliver the composition of the present disclosure to the eukaryotic cells, various methods known in the art, such as microinjection, electroporation, DEAE-dextran treatment, lipofection, nanoparticle-mediated transfection, protein transduction domain-mediated transduction, virus-mediated gene delivery, and PEG-mediated transfection in protoplast, etc. may be used, but are not limited thereto.
  • In a specific embodiment, the contacting with the eukaryotic cells may include culturing the eukaryotic cells in the presence of the composition.
  • In a specific embodiment, the culturing includes culturing in the presence of a chondrogenic inducer.
  • In a specific embodiment, the method of producing the extracellular matrix of the present disclosure may further include isolating the extracellular matrix from the contacting product.
  • In another specific embodiment, the method of producing the extracellular matrix may include contacting chondrocytes with the composition for promoting chondrogenesis of the present disclosure.
  • Still another aspect provides a method of forming a cartilage, the method including contacting chondrocytes with the composition for promoting chondrogenesis of the present disclosure.
  • In a specific embodiment, the chondrocytes may be isolated from a subject.
  • In a specific embodiment, the chondrocytes may be derived from a subject to be transplanted with the produced cartilage.
  • Still another aspect provides a method of producing ECM, the method including culturing eukaryotic cells having increased extracellular matrix productivity of the present disclosure to produce ECM; and isolating ECM from the culture.
  • In a specific embodiment, the culturing may be culturing in the presence of a chondrogenic inducer.
  • In a specific embodiment, the chondrogenic inducer may be BMP.
  • Advantageous Effects of Disclosure
  • A composition for promoting production of an extracellular matrix according to an aspect may be injected into a subject who requires supply of the extracellular matrix, thereby preventing or treating diseases including a joint disease, and improving symptoms thereof, and the composition may be applied to a method of efficiently producing the extracellular matrix from eukaryotic cells.
  • A composition for promoting chondrogenesis of chondrocytes according to another aspect may be injected into a subject, thereby preventing or treating diseases including a joint disease, and improving symptoms thereof. The composition may promote chondrogenesis of chondrocytes isolated from the subject, and thus it may be applied to a method of efficiently producing various components including extracellular matrice which are produced by chondrogenesis.
  • According to a method of producing an extracellular matrix from eukaryotic cells according to still another aspect, the extracellular matrix may be efficiently produced from eukaryotic cells.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 illustrates a method of preparing MAST4 knockout mice using a CRISPR/Cas9 system;
  • FIG. 2A shows RT-PCR results of examining changes in expression levels of respective genes in MAST4 knockout mouse type A and B, and FIG. 2B shows protein expression patterns in MAST4 knockout mice;
  • FIG. 3 shows identification of MAST4 knockout in C3H10T1/2 cells in which MAST4 was knocked out using the CRISPR/Cas9 system;
  • FIG. 4 shows RT-PCR results of examining changes in expression levels of respective genes in C3H10T1/2 cells in which MAST4 was knocked out using the CRISPR/Cas9 system;
  • FIG. 5 shows RT-PCR results of examining changes in expression levels of respective genes in a micromass culture to confirm chondrogenesis;
  • FIG. 6 shows alcian blue staining results of examining a difference in cartilage differentiation in C3H10T1/2 cells in which MAST4 was knocked out using the CRISPR/Cas9 system;
  • FIG. 7 shows sequence information of target genes used to knockout MAST4 of human cells;
  • FIG. 8A shows human chondrocytes in which MAST4 was knocked out using siRNA, and FIG. 8B shows expression levels of extracellular matrix factors in human chondrocytes in which MAST4 was knocked out using the CRISPR/Cas9 system;
  • FIG. 9A shows changes in the expression level of MAST4 after treatment of human primary chondrocytes with TGF-β1, and expression levels of extracellular matrix factors thereby, FIG. 9B shows changes in the expression level of MAST4 after treatment of human primary chondrocytes with TGF-β1, and expression levels of extracellular matrix factors thereby; and
  • FIG. 10 shows chondrogenesis and regeneration effects in the tibia of the MAST4 knockout mouse.
  • MODE OF DISCLOSURE
  • Hereinafter, the present disclosure will be described in more detail with reference to embodiments. However, these embodiments are for illustrative purposes only, and the scope of the present disclosure is not intended to be limited by these embodiments.
  • Example 1. Confirmation of Increased Expression of Cartilage Component in MAST4 Knockout Mouse
  • 1-1. Preparation of MAST4 Knockout Mouse Using CRISPR/Cas9 System
  • To examine whether an extracellular matrix as a cartilage component was increased by suppressing MAST4 expression, MAST4 knockout mice were prepared using a CRISPR/Cas9 system.
  • In detail, to prepare CRISPR knockout mice, pX330-U6-Chimeric_BB-CBh-hSpCas9 (Addgene, #42230), donated by Dr. Feng Zhang (Cong et al., 2013), was used as a plasmid capable of expressing Cas9 mRNA and guide RNA. Since MAST4 is a large protein of 7 kb or more, it was designed such that the gene editing was allowed to target two parts, exon 1 and exon 15. A guide RNA sequence targeting exon 1 of MAST4 is 5′-GGAAACTCTGTCGGAGGAAGGGG-3′ and a sequence targeting exon 15 is 5′-GGCACAAAGAGTCCCGCCAGAGG-3′. The guide RNA sequence was used to prepare oligomers as in MAST4 CRISPR oligomers of the following Table in accordance with the manufacturer's protocol (http://crispr.mit.edu/, Zhang Feng Lab), and each oligomer was inserted into a px330 plasmid to clone two plasmids targeting exon 1 and exon 15, respectively.
  • TABLE 1
    MAST4 exon 1 CRISPR F (SEQ ID NO: 18) 5′-caccGGAAACTCTGTCGGAGGAAG-
    3
    MAST4 exon
     1 CRISPR R (SEQ ID NO: 19) 5′-aaacCTTCCTCCGACAGAGTTTCC-3
    MAST4 exon
     15 CRISPR F (SEQ ID NO: 5′-caccGGCACAAAGAGTCCCGCCAG-
    20) 3′
    MAST4 exon 15 CRISPR R (SEQ ID NO: 5′-aaacCTGGCGGGACTCTTTGTGCC-
    21) 3′
  • To obtain embryos, 5 IU of pregnant mare serum gonadotrophin (PMSG; Prospec, cat. No. HOR-272) was administered to a C57BL/6J female mouse 2 days before mating, and after 47 hours, 5 IU of humanchorionic gonadotrophin (hCG, Prospec, cat. HOR-250) was administered thereto . . . . Thereafter, the mouse was mated with C57BL/6J male mouse, and embryos were obtained from fallopian tubes. A microinjection mixture including 5 ng/μl of the prepared plasmid and 10 ng of ssDNA donor was injected into the pronuclei of the embryos at a one-cell-stage with reference to an existing standard protocol (Gordon and Ruddle, 1981). The injected one-cell-embryos were transferred to pseudopregnant ICR mice.
  • Phenotypic analysis of born mice was performed for exon 1 and exon 15. Finally, two types of MAST4 knockout mice were obtained. Information about the two types of MAST4 knockout mice, type A and type B are shown as in FIG. 1 and the following Table 2 (5′→3′).
  • TABLE 2
    Type A MAST4 KO ATGGGGGAGAAAGTTTCCGAGGCGCCTGAGCCCGT
    (71 bp deletion in exon 1) GCCCCGGGGCTGCAGCGGACACGGCGCCCGGACCC
    (SEQ ID NO: 22) TAGTCTCTTCGGCGGCAGCCGTGTCCTCGGAGGGCG
    CTTCCTCAGCGGAGTCATCCTCTGGCTCGGAAACTCT
    GTCGGAGGAAGGGGAGCCCAGCCGCTTCTCCTGCA
    GGTCGCAGCCGCCGCGGCCGCCGGGCGGCGCCCT
    GGGAACCCGGCTACCCGCCGCGTGGGCTCCCGCGC
    GCGTGGCTCTGGAGCGTGGAGTCCCTACCCTGCCG
    CTGCCGCACCCGGGAGGAGCGGTGCTGCCGGTGCC
    CCAGGTCAGCAGCGCATCCCAAGAGGAGCAGGATGA
    AGAG
    Type B MAST4 KO ATGGGGGAGAAAGTTTCCGAGGCGCCTGAGCCCGT
    (90 bp deletion in exon 1) GCCCCGGGGCTGCAGCGGACACGGCGCCCGGACCC
    (SEQ ID NO: 23) TAGTCTCTTCGGCGGCAGCCGTGTCCTCGGAGGGCG
    CTTCCTCAGCGGAGTCATCCTCTGGCTCGGAAACTCT
    GTCGGAGGAAGGGGAGCCCAGCCGCTTCTCCTGCA
    GGTCGCAGCCGCCGCGGCCGCCGGGCGGCGCCCT
    GGGAACCCGGCTACCCGCCGCGTGGGCTCCCGCGC
    GCGTGGCTCTGGAGCGTGGAGTCCCTACCCTGCCG
    CTGCCGCACCCGGGAGGAGCGGTGCTGCCGGTGCC
    CCAGGTCAGCAGCGCATCCCAAGAGGAGCAGGATGA
    AGAG
    type A MAST4 KO GGCAGTCTACTTTGTTCGGCACAAAGAGTCCCGCCA
    (3 bp deletion in exon 15) GAGGTTTGCCATGAAGAAGATCAA
    (SEQ ID NO: 24) CAAGCAGAACCTCATCCTTCGGAACCAGATCCAGCA
    GGCCTTCGTGGAGCGAGACATCCT
    GACTTTCGCAGAGAACCCCTTTGTGGTCAGCATGTAT
    TGCTCCTTTGAAACGAGGCGTCA
    CTTATGCATGGTCATGGAGTATGTAGAAG
    type B MAST4 KO GGCAGTCTACTTTGTTCGGCACAAAGAGTCCCGCCA
    (13 bp deletion in exon 15) GAGGTTTGCCATGAAGAAGATCAA
    (SEQ ID NO: 25) CAAGCAGAACCTCATCCTTCGGAACCAGATCCAGCA
    GGCCTTCGTGGAGCGAGACATCCT
    GACTTTCGCAGAGAACCCCTTTGTGGTCAGCATGTAT
    TGCTCCTTTGAAACGAGGCGTCA
    CTTATGCATGGTCATGGAGTATGTAGAAG
  • Bases to be deleted in Table 2 are shown in bold.
  • 1-2. RNA-Sequencing for Confirmation of Change of Cartilage Component Expression in MAST4 Knockout Mouse
  • To examine changes in the extracellular matrix as a cartilage component in MAST4 knockout mice prepared in Example 1-1, RNA-sequencing was performed for respective genes.
  • In detail, 1 day-old-MAST4 knockout mice prepared in Example 1-1, hetero-type mice, and wild-type mice were sacrificed, and then their tibia was excised. Each of the excised tibias was placed in a dish containing DEPC-PBS on ice, and cartilage and bone in the tibia were separated using a needle under a dissecting microscope. The tissues separated from each group was immersed in 500 μl of TRIzol (purchased from Invitrogen), which were then used as samples. RNA was extracted according to a method well known in the art, and quantified using a nanodrop (Thermo scientific).
  • RNA-sequencing was performed by Theragen Etex. In detail, mRNA was isolated from 2 μg of total RNA extracted from the mouse of each group using oligo(dT). After fragmentation of the mRNA, single-stranded cDNA was synthesized through random hexamer priming. This single-strand cDNA was used as a template to synthesize a second strand, thereby synthesizing a double-stranded cDNA. To prepare blunt-ends, end repair was performed, and to ligate an adapter, A-tailing and adapter ligation were performed. Thereafter, cDNA library was amplified by polymerase chain reaction (PCR). A concentration and size of the final product were examined using 2100 BioAnalyzer. The produced library was finally quantified using a KAPA library quantification kit, and then sequence interpretation was performed using Hiseq2500. To remove low-quality sequences from the interpreted sequences, filtering was performed such that reads containing 10% or more of bases marked as ‘N’s in the sequence information or reads containing 40% or more of bases less than Q20 were removed, and reads whose average quality is Q20 or less were also removed. The whole filtering process was performed using the in-house program. The filtered sequences were aligned to a reference genome sequence (hg19) of the corresponding species using STAR v2.4.0b (Dobin et al, 2013).
  • Expression level was measured using Cufflinks v2.1.1 (Trapnell C. et al, 2010), and the calculated expression values were expressed as fragments read per kilobase of exon per million fragments mapped (FPKM). Ensemble 72 was used as a genetic information database, and a non-coding gene region was removed with expression-mask option. To increase measurement accuracy of the expression levels, multi-read-correction and frag-bias-correct options were additionally used, and all other options were set to default values.
  • To examine genes which were changed by MAST4 knockout, expression values of the samples of each group, which were obtained through Cufflinks, were used. Genes, of which expression values were twice or more, as compared with those of wild-type MAST4, and which had a significance of P value <0.01, were selected, and the expression values of the selected genes and their differences are listed in Table 3.
  • As a result, it was confirmed that expression of many genes associated with extracellular matrix as a cartilage component was increased as in the following Table 3. However, in all of the two types of MAST4 knockout mice, reduced expression of mmp8 and mmp9 which are extracellular matrix-degrading enzymes was observed.
  • 1-3. RT-PCR for Confirmation of Change of Cartilage Component Expression in MAST4 Knockout Mouse
  • To more specifically examine changes in the extracellular matrix as a cartilage component in MAST4 knockout mice prepared in Example 1-1, a part of genes showing changes in the expression in the RNA sequencing results of Example 1-2 was selected and subjected to RT-PCR.
  • In detail, RT-PCR was performed using a set of primers of the following Table 4 and AccuPower PCR premix (BIONEER, Korea) according to the manufacturer's instructions.
  • As a result, results were consistent with the RNA sequencing results of Example 1-2, and it was confirmed that expression of genes associated with extracellular matrix as a cartilage component was increased (FIG. 2).
  • 1-4. Confirmation of Expression Level of Chondrocyte Marker in MAST4 Knockout Mouse
  • To examine the effect of MAST4 knockout on chondrocytes, Col2a1 which is known as a chondrocyte marker was stained with fluorescence in the mouse tibia.
  • In detail, the tibia tissue was obtained from the mouse model of Example 1-1, and fixed with 4% paraformaldehyde (PFA, Wako, Osaka, JAPAN) in 0.01 M phosphate buffer saline (PBS, pH 7.4) at 4° C. overnight. The tissue was decalcified with 10% EDTA, and embedded in paraffin (Leica Biosystems, MO, USA), and sectioned 6 mm in thickness. The sample slide was stained with hematoxylin and eosin, and the tissue section was incubated with a primary antibody at 4° C. overnight. The primary antibody targets ColI2a1 (Abcam, Cambridge, UK). After washing with PBS, the tissue section was sequentially incubated with AlexaFluor 488 (Invitrogen, CA, USA) at room temperature for 2 hours. Each image was obtained using a confocal microscope LSM700 (Carl Zeiss, Oberkochen, Germany), and a representative sample section was stained with freshly prepared Russell-Movatmodified pentachrome (American MasterTech, CA, USA).
  • As a result, FIG. 10 is an enlargement of a specific area of the observed sample, where Col2a1 (fluorescent green zone/grey background zone) was significantly increased in the tibia of the MAST4 knockout mouse model. TOPRO-3 (areas marked by red dots/gray dots) shows staining of the nuclei of chondrocytes. Therefore, it was confirmed that chondrogenesis and cartilage regeneration may be promoted by MAST4 knockout.
  • Example 2. Confirmation of Increased Expression of Cartilage Component in MAST4 Knockout Cells
  • 2-1. Preparation of MAST4 Knockout Cells Using CRISPR/Cas9 System
  • To examine whether increased extracellular matrix in the MAST4 knockout mice is also reproduced in vitro, MAST4 knockout cells were prepared using the CRISPR/Cas9 system.
  • In detail, C3H/10T1/2, Clone 8 (ATCCCCL-226™) which is a mouse-derived fibroblast cell and is able to differentiate into chondrocytes was purchased ((C3H10T1/2 cell) provided by prof. Seon-Yong Jeong's lab, Department of Medical Genetics, School of Medicine, Ajou University). To knockout the cells, lentiCRISPR v2 (Plasmid #52961), pVSVg (AddGene 8454), and psPAX2 (AddGene 12260) were purchased from Addgene, and oligomers of the following Table 5 were used to insert guide RNA targeting exon 1 of mouse MAST4 gene (ENSMUSG00000034751) into LentiCRISPR v2 plasmid according to the manufacturer's instructions (lentiCRISPRv2 and lentiGuide oligo cloning protocol), thereby preparing a plasmid expressing guide RNA and Cas9 enzyme at the same time (as a control group, a plasmid having no guideRNA and expressing only Cas9 was used).
  • TABLE 5
    Oligomer Sequence
    mMAST4 CRISPR exon 1 sgRNA F 5′-CACCGTACCCTGCCGCTGCCGCACC-3′
    (SEQ ID NO: 70)
    mMAST4 CRISPR exon 1 sgRNA R 5′-AAACGGTGCGGCAGCGGCAGGGTAC-3′
    (SEQ ID NO: 71)
    mouse MAST4 exon 1 5′-
    (SEQ ID NO: 72) ATGGGGGAGAAAGTTTCCGAGGCGCCTG
    AGCCCGTGCCCCGGGGCTGCAGCGGACA
    CGGCGCCCGGACCCTAGTCTCTTCGGCG
    GCAGCCGTGTCCTCGGAGGGCGCTTCCT
    CAGCGGAGTCATCCTCTGGCTCGGAAACT
    CTGTCGGAGGAAGGGGAGCCCAGCCGCT
    TCTCCTGCAGGTCGCAGCCGCCGCGGCC
    GCCGGGCGGCGCCCTGGGAACCCGGCT
    ACCCGCCGCGTGGGCTCCCGCGCGCGT
    GGCTCTGGAGCGTGGAGTCCCTACCCTG
    CCGCTGCCGCACCCGGGAGGAGCGGTG
    CTGCCGGTGCCCCAGGTCAGCAGCGCAT
    CCCAAGAGGAGCAGGATGAAGAG-3′
  • This method is a lentivirus-based CRISPR knockout method. To prepare a virus, the three plasmids prepared above (LentiCRISPR v2 (+guide RNA): guide RNA+Cas9 expressing plasmid, pVSVg: Virus envelop plasmid, psPAX2: Virus packaging plasmid) were transfected into 293T cells using a polyethyenimine (PEI) reagent. 18 hours later, the medium was replaced with a fresh medium, and only the medium was collected, and viruses were obtained using a 0.45 μm filter. The obtained viruses were transfected into a 6-well dish to which C3H10T/12 was seeded. 24 hours after treatment with 1 ml of virus+1 ml of DMEM/FBS+2 μl of polybren, the medium was replaced with fresh DMEM/FBS. 24 hours later, only infected cells were selected by treatment with puromycin, and subcultured to 40% confluency in a 10 cm dish. Since gene editing by CRISPR may randomly occur in cells, single colony selection was performed. Cells were seeded in 10 cm dishes such that 50 cells existed in each dish. When cells formed colonies over time, these colonies were defined as one clone, and genomic DNA was extracted from each clone. PCR was performed using primers specifically amplifying exon 1 (F: 5′->3′ CTGTGGTCCAACCTCTGTCA, R: 5′->3′ ATCGGCTCAGTGACACTTCC). The amplified PCR products were analyzed by the sequencing company. As a result of sequencing analysis, cells in which gene editing by frameshift was identified were used in the experiment, together with control cells. The sequences targeted by the prepared guide RNA were are in bold in Table 5. As a result of sequencing the MAST4 knockout results, deletion of two nucleotides occurred in mouse MAST4 exon 1, indicating frameshift induction.
  • 2-2. RT-PCR for Confirmation of Change of Cartilage Component Expression in MAST4 Knockout Cells
  • To examine changes in the extracellular matrix as a cartilage component in MAST4 knockout mice prepared in Example 1-1, RT-PCR was performed for respective genes.
  • 10 μl of a medium containing total 105 cells was put in the center of 12 wells, and incubated for 2 hours. 1 ml of DMEM containing 10% FBS was added to each well. 24 hours later, cells were harvested, and RNA was extracted using an easy-BLUE™ Total RNA Extraction Kit (Intron, Cat 17061) according to the manufacturer's instructions. Next, cDNA was synthesized using M-MLV reverse transcriptase (Promega, M1705) according to the manufacturer's instructions. Primers used in RT-PCR are as described in Table 4.
  • As a result, increased expression of extracellular matrix-associated genes was also found in MAST4 knockout cells, as consistent with the results of Example 1-2 and Example 1-3 (FIG. 4), indicating that the same results as in the MAST4 knockout mouse were also obtained in vitro.
  • Example 3. Micromass Culture of MAST4 Knockout Cells and Confirmation of Increased Cartilage Differentiation Activity
  • 3-1. Micromass Culture of MAST4 Knockout Cells
  • To evaluate chondrogenic ability of the MAST4 knockout cells of Example 2-2, micromass culture was performed.
  • In detail, MAST4 knockout cells were prepared as in Example 2-1, and micromass culture was performed with reference to a known method (Differentiation and Mineralization of Murine Mesenchymal C3H10T1/2 Cells in Micromass Culture, 2010, Rani Roy). First, 10 μl of a medium containing total 105 fibroblast cells were put in the center of each well of a 12-well plate, and incubated for 2 hours. 1 ml of DMEM containing 10% FBS was added to each well. Thereafter, 100 ng/ml, 500 ng/ml, or 1000 ng/ml of BMP2 was added to each culture depending on the purpose of cartilage induction, respectively. Thereafter, the medium was replaced with a fresh medium every three days.
  • 3-2. Confirmation of Reproduction of Effects of Micromass-Cultured MAST4 Knockout Cells
  • To examine whether production of extracellular matrix as a cartilage component was also increased in the MAST4 knockout cells cultured according to Example 3-1, as in the MAST4 knockout cells of Example 2-2, and finally, chondrogenic ability was increased therein, RT-PCR was performed.
  • In detail, cells, which were cultured for 0 day, 3 days, and 6 days from the day when the cells were seeded in a plate for micromass culture, were harvested, respectively, and RNA was extracted therefrom on the same day. RT-PCR was performed for respective genes, as in Example 1-3, and whether or not production of the cartilage component was increased was examined.
  • As a result, as consistent with the results observed in the MAST4 knockout cells of Example 2-2, expression of extracellular matrix components was increased, and at the same time, differentiation into chondrocytes began with aggrecan expression on day 3 after induction using BMP2, and as a result, it was confirmed that chondrogenic ability was increased (FIG. 5). In particular, when MAST4 was knock-outed, some genes (hapIn1) showed no significant difference in the expression on day 3, but all of the indicated extracellular matrix-associated genes showed overexpression on day 6. In contrast, in the control group, some proteins were less expressed or rather decreased on day 6 (e.g., Matn3, or Comp). The MAST4 knockout cells were found to be useful in the overexpression of all various extracellular matrices.
  • 3-3. Confirmation of Chondrogenesis of Mass-Cultured MAST4 Knockout Cells
  • With regard to the overexpression of the respective extracellular matrix-associated genes observed in Example 3-2, to examine whether or not the expression was actually increased at the level of isolated proteins, not at the gene expression level, alcian blue staining was performed.
  • In detail, plates of cells corresponding to each date were washed twice with PBS and fixed for 15 minutes by adding 1 ml of 4% paraformaldehyde. Then, 1 ml of 1% alcian blue 8-GX (Sigma-Aldrich, A5268) dissolved in 0.1 N HCl (pH 1.0) was added and stained overnight. After washing twice with 500 μl of 0.1 N HCl, images were obtained.
  • As a result, in the case of MAST4 knockout cells, chondrogenesis was increased from day 3, and extracellular matrix secretion was increased, and the degree was increased with increasing BMP2 concentration (FIG. 6).
  • Example 4. Confirmation of Effect of Suppression of MAST4 Expression in Human Cells Example 4-1. Confirmation of Effect of Suppression of MAST4 Expression in Human Cells
  • It was examined whether the results confirmed in the knockout mouse model and mouse cells were also induced in human cells.
  • In detail, human primary chondrocytes (donated by College of Medicine, Inha University) were knocked-out by transient transfection with MAST4 siRNA(h) (sc-106201; Santa Cruz biotechnology) (FIG. 8A) or MAST4 expression was knocked-out by the CRISPR/Cas9 system. MAST4 siRNA was transfected using a Lipofectamine RNAiMAX transfection reagent of ThermoFisher SCIENTIFIC, and information of primers used herein is as described in the following Table 6. Preparation and treatment of the CRISPR/Cas9 system were performed in the same manner as in Example 1-1 with reference to GeneArt™ Precision gRNA Synthesis Kit (A29377) of ThermoScientific, and information of primers used herein is as described in the following Table 6.
  • For high transfection efficiency, siRNA transfection was performed by a reverse transfection technique in which cell planting and transfection are performed at the same time, and a transfection reagent was Lipofectamine RNAiMAX transfection reagent of ThermoFisher SCIENTIFIC. In detail, 15 nM of MAST4 siRNA and 4.5 μl of Lipofectamine RNAiMax were mixed in 40 μl of Gibco™ Opti-MEM™, and incubated for 15 minutes. Thereafter, human primary chondrocytes of 1.5×105 cell/well were plated together with 2 ml of a medium (FBS 10%) containing no gentamicin in a 6-well plate (ColI coated plate), and the siRNA mixture was added thereto. 72 hours later, the cells were harvested and RNA was isolated. Human primary chondrocytes were cultured in a collagen I-coated flask (175, Col I Straight Vent 356487, Corning) under conditions of DMEM (17-205-CVR Corning), FBS Qualified (USA origin 500 mL 26140-079, Gibco), L-glutamine (200 mM) (100×25030-081, Gibco), and gentamicin (5 ug/ml) (10 mL 15700-060, Thermofisher).
  • Knockout was performed by targeting 20 nt on the genome of MAST4 (target sequences are marked in bold), and specifically, #1 and #3 target Exon5, and #2 targets Exon 8. #1 and #3 were prepared in the reverse direction, and #2 was prepared in the forward direction. The human MAST4 gene used in the preparation of CRISPR/Cas9 system was with reference to MAST4 ENSG00000069020 (http://asia.ensembl.org/). Information of targeted Exon sequences and NGG PAM sequences (grey box) on which CRISPR deletion occurred are shown in detail in FIG. 7.
  • TABLE 6
    hMAST4 CR#1 F (SEQ ID NO: 73) 5′-TAATACGACTCACTATAG
    GAGTGTGGTCGAGGCAATGC-3′
    hMAST4 CR#1 R (SEQ ID NO: 74) 5′-TTCTAGCTCTAAAAC
    GCATTGCCTCGACCACACTC-3′
    hMAST4 CR#2 F (SEQ ID NO: 75) 5′-TAATACGACTCACTATAG
    GTAACTCGTCTGGTGTTGGT-3′
    hMAST4 CR#2 R (SEQ ID NO: 76) 5′-TTCTAGCTCTAAAAC
    ACCAACACCAGACGAGTTAC-3′
    hMAST4 CR#3 F (SEQ ID NO: 77) 5′-TAATACGACTCACTATAG
    AGCAACCGGAAAAGCTTAAT-3′
    hMAST4 CR#3 R (SEQ ID NO: 78) 5′-TTCTAGCTCTAAAAC
    ATTAAGCTTTTCCGGTTGCT-3′
    HumanAcanRT Forward (336) 5′-gaatcaactgctgcagacca-3′
    (SEQ ID NO: 82)
    HumanAcan RT Reverse (336) 5′-gtgccagatcatcaccacac-3′
    (SEQ ID NO: 83)
    HumanCol9a1RT Forward (467) 5′-CGTGGATTTCCAGGCCGTGG-3′
    (SEQ ID NO: 84)
    HumanCol9a1RT Reverse (467) 5′-TCGCTGTCCTTGATCACCAG-3′
    (SEQ ID NO: 85)
    HumanGapdhRT Forward (156) 5′-TGGCAAAGTGGAGATTGTTGCC-3′
    (SEQ ID NO: 86)
    HumanGapdhRT Reverse (156) 5′-AAGATGGTGATGGGCTTCCCG-3′
    (SEQ ID NO: 87)
  • As a result, as shown in FIG. 8A, when MAST4 siRNA was transfected, MAST4 expression was decreased, and at this time, expression of extracellular matrix factors such as Acan was increased. Further, as shown in FIG. 8B, when MAST4 was knocked out, expression of extracellular matrix factors such as Acan and Col9a1 was increased. These results are the same as those demonstrated in the previous mouse models and mouse cells. Therefore, with regard to other extracellular matrix factors and chondrogenic effects, the same results as those demonstrated in the mouse may be also obtained by suppressing MAST4 expression in human cells.
  • Example 4-2. Suppression of MAST4 Expression by TGF-β1 in Human Cells and Confirmation of Effect Thereof
  • It was examined whether suppression of MAST4 expression as confirmed in Example 4-1 was induced by TGF-β1 and expression of extracellular matrix factors was affected thereby.
  • In detail, the human primary chondrocytes of Example 4-1 were treated with TGF-β1, and an expression level thereof was measured by RT-PCR as in Examples 1-2 and 1-3 and Western blotting.
  • As a result, as shown in FIG. 9, when TGF-β1 (5 ng/ml) was treated for 24 hours, 48 hours, or 72 hours, respectively, MAST4 expression was suppressed, and as a result, expression of extracellular matrix factors was increased. When co-treatment with TGF-β1 (5 ng/ml) and TEW-7197 which is a TGF-β1 inhibitor was performed (FIG. 9B), Acan expression increased by TGF-β1 was suppressed and the inhibitory effect on MAST4 expression was also decreased, as compared with single treatment with TGF-β1.

Claims (20)

1. A composition for promoting production of an extracellular matrix from eukaryotic cells, the composition comprising a compound capable of specifically binding to Microtubule Associated Serine/Threonine Kinase Family Member 4 (MAST4) protein or a fragment thereof, or a compound capable of specifically binding to a nucleic acid encoding the MAST4 protein or the fragment thereof.
2. The composition of claim 1, wherein the MAST4 protein comprises any one amino acid sequence of SEQ ID NOS: 1 to 7 and 15, and the nucleic acid encoding the MAST4 protein comprises any one polynucleotide sequence of SEQ ID NOS: 8 to 14 and 16.
3. The composition of claim 1, wherein the compound capable of specifically binding to the MAST4 protein or the fragment thereof, or the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof is a chemically synthesized compound, polypeptide, or polynucleotide that inhibits activity or expression of the MAST4 protein, or a combination thereof.
4. The composition of claim 3, wherein the polypeptide is an antibody or an antigen-binding site thereof.
5. The composition of claim 1, wherein the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof is microRNA (miRNA), small interfering RNA (siRNA), short hairpin RNA (shRNA), Piwi-interacting RNA (piRNA), small nuclear RNA (snRNA), or antisense oligonucleotide, each specific to the nucleic acid encoding the MAST4 protein or the fragment thereof, or a combination thereof.
6. The composition of claim 1, wherein the compound capable of specifically binding to the nucleic acid encoding the MAST4 protein or the fragment thereof is CRISPR-Cas comprising guide RNA specific to the nucleic acid encoding the MAST4 protein or the fragment thereof.
7. The composition of claim 6, wherein the guide RNA is a dual RNA comprising CRISPR RNA (crRNA) and transactivating crRNA (tracrRNA) specific to the nucleic acid encoding the MAST4 protein or the fragment thereof, or a single strand guide RNA comprising parts of the crRNA and the tracrRNA and hybridizing with the nucleic acid encoding the MAST4 protein or the fragment thereof.
8. The composition of claim 1, wherein the eukaryotic cells are fibroblast cells or chondrocytes.
9. The composition of claim 1, wherein the composition is to promote chondrogenesis of the eukaryotic cells.
10. The composition of claim 1, wherein the composition is to prevent or treat a joint disease or to improve symptoms thereof.
11. The composition of claim 9, wherein to promote chondrogenesis is to induce chondrogenesis.
12. The composition of claim 1, wherein the composition is used for tissue regeneration or anti-aging.
13. The composition of claim 1, further comprising TGF-β1.
14. A method of preventing, treating, or improving a joint disease, the method comprising administering the composition of claim 1 to a subject.
15. A method of producing an extracellular matrix from eukaryotic cells, the method comprising contacting the eukaryotic cells with the composition for promoting production of extracellular matrix from eukaryotic cells of claim 1.
16. The method of claim 15, wherein the eukaryotic cells are isolated from a subject.
17. The method of claim 15, wherein the contacting with the eukaryotic cells comprises culturing the eukaryotic cells in the presence of the composition.
18. The method of claim 17, wherein the culturing is to culture in the presence of a chondrogenic inducer.
19. The method of claim 15, further comprising isolating the extracellular matrix from the contacting product.
20. The method of claim 15, wherein the eukaryotic cells are chondrocytes.
US17/494,765 2017-03-08 2021-10-05 Extracellular matrix-producing composition using mast4 gene and preparation method therefor Abandoned US20220220221A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/494,765 US20220220221A1 (en) 2017-03-08 2021-10-05 Extracellular matrix-producing composition using mast4 gene and preparation method therefor

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
KR10-2017-0029607 2017-03-08
KR20170029607 2017-03-08
KR10-2018-0027111 2018-03-07
KR1020180027111A KR102588627B1 (en) 2017-03-08 2018-03-07 Composition for preparing extracellular matrix using MAST4 gene and method for preparing the same
PCT/KR2018/002763 WO2018164507A2 (en) 2017-03-08 2018-03-08 Extracellular matrix-producing composition using mast4 gene and preparation method therefor
US201916492477A 2019-09-09 2019-09-09
US17/494,765 US20220220221A1 (en) 2017-03-08 2021-10-05 Extracellular matrix-producing composition using mast4 gene and preparation method therefor

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US16/492,477 Continuation US11180573B2 (en) 2017-03-08 2018-03-08 Extracellular matrix-producing composition using MAST4 gene and preparation method therefor
PCT/KR2018/002763 Continuation WO2018164507A2 (en) 2017-03-08 2018-03-08 Extracellular matrix-producing composition using mast4 gene and preparation method therefor

Publications (1)

Publication Number Publication Date
US20220220221A1 true US20220220221A1 (en) 2022-07-14

Family

ID=63718648

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/492,477 Active US11180573B2 (en) 2017-03-08 2018-03-08 Extracellular matrix-producing composition using MAST4 gene and preparation method therefor
US17/494,765 Abandoned US20220220221A1 (en) 2017-03-08 2021-10-05 Extracellular matrix-producing composition using mast4 gene and preparation method therefor

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/492,477 Active US11180573B2 (en) 2017-03-08 2018-03-08 Extracellular matrix-producing composition using MAST4 gene and preparation method therefor

Country Status (7)

Country Link
US (2) US11180573B2 (en)
EP (1) EP3594324A4 (en)
JP (3) JP7154238B2 (en)
KR (1) KR102588627B1 (en)
CN (1) CN110612348B (en)
AU (2) AU2018231559A1 (en)
CA (1) CA3055729A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021146566A1 (en) * 2020-01-16 2021-07-22 The Johns Hopkins University Engineered fibroblasts as cell therapy to treat cancer via tumor stroma stabilization
WO2023150305A1 (en) * 2022-02-04 2023-08-10 Kim Seong Jin Method of making bone and cartilage

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6528052B1 (en) * 2000-09-29 2003-03-04 The Board Of Trustees Of The Leland Stanford Junior University Method for in vivo ex vivo and in vitro repair and regeneration of cartilage and collagen and bone remodeling
EP1894581A1 (en) * 2006-08-31 2008-03-05 CellCoTec B.V. Repair of cartilage tissue using a matrix gel containing chondrocytes
KR20120104517A (en) 2009-09-03 2012-09-21 제넨테크, 인크. Methods for treating, diagnosing, and monitoring rheumatoid arthritis
WO2012083363A1 (en) * 2010-12-22 2012-06-28 Murdoch Childrens Research Institute A method of treatment
ES2727334T3 (en) * 2011-09-07 2019-10-15 3 D Matrix Ltd MicroRNA based methods and assays for osteosarcoma
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
KR102039311B1 (en) 2012-09-07 2019-11-04 주식회사 마크로젠 Fusion Protein containing AXL and composition for diagnosing cancer
AU2014317861B2 (en) 2013-09-09 2019-11-28 Figene, Llc Gene therapy for the regeneration of chondrocytes or cartilage type cells
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
CA3011306A1 (en) 2016-01-14 2017-07-20 Spinalcyte, Llc Cellular blend for the regeneration of chondrocytes or cartilage type cells

Also Published As

Publication number Publication date
KR102588627B1 (en) 2023-10-16
JP7154238B2 (en) 2022-10-17
CA3055729A1 (en) 2018-09-13
EP3594324A4 (en) 2021-01-20
JP2020513849A (en) 2020-05-21
US20200239596A1 (en) 2020-07-30
AU2018231559A1 (en) 2019-10-31
AU2023222944A1 (en) 2023-11-23
US11180573B2 (en) 2021-11-23
JP2022188178A (en) 2022-12-20
EP3594324A2 (en) 2020-01-15
CN110612348A (en) 2019-12-24
KR20180103015A (en) 2018-09-18
CN110612348B (en) 2024-02-09
JP7498239B2 (en) 2024-06-11
JP2024116197A (en) 2024-08-27

Similar Documents

Publication Publication Date Title
JP7498239B2 (en) Composition for producing extracellular matrix using MAST4 gene and method for producing same
CN102712927A (en) Treatment of membrane bound transcription factor peptidase, site 1 (MBTPS1) related diseases by inhibition of natural antisense transcript to MBTPS1
TWI516592B (en) An inducible gene expression composition for using eukaryotic pol-2 promoter-driven transcription in prokaryotes and the applications thereof
CN102482672A (en) Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
CN102439149A (en) Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
US20180362979A1 (en) Therapeutic and diagnostic molecules
JP2019520080A (en) Substances and methods for treating pain related disorders
KR20130131414A (en) Ips cells and method for generating same
KR20200141470A (en) Composition and method for adjusting somatic cell reprogramming and imprinting
JP2021528984A (en) In vitro induction of adult stem cell expansion and induction
TW201629227A (en) Production and utilization of a novel anti-cancer drug in therapy
KR101642202B1 (en) Composition comprising klf10 inhibitors for stimulating chondrogenesis or treatment of cartilage diseases, and method for stimulating chondrogenesis using the same
KR20140046339A (en) Method for differentiation into retinal cells from stem cells using inhibition of mirna-203
WO2018164507A2 (en) Extracellular matrix-producing composition using mast4 gene and preparation method therefor
US20230121720A1 (en) Diagnostic methods using pcg-1a expression
CN118147149A (en) Method for knocking out genes of mice and miR-455 gene knocked out mouse model constructed by same
WO2017053389A1 (en) Methods for inhibiting epithelial to mesenchymal transition by inhibition of foxs1
KR20160141677A (en) Method for inducing high activity of stem cells
Serey Elucidating Mechanisms of Alternative Splicing in Cancer and Cellular Stress
KR20220155585A (en) Diagnostic methods using SIRT1 expression
Flanagan Identification of genes responsible for maintenance of differentiation capability in dental pulp stem cells
JPWO2016009809A1 (en) Expression suppressing nucleic acid molecule for gene expression control and use thereof
KR20140101173A (en) Methods for Inducing Differentiation from Embryonic Stem Cells to Early Neuronal Precursor Cells by Introducing ACS4 Gene

Legal Events

Date Code Title Description
AS Assignment

Owner name: KIM, YPSSE, KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KIM, SEONG JIN;JUNG, HAN SUNG;TAKAHASHI, SATORU;SIGNING DATES FROM 20200916 TO 20200917;REEL/FRAME:057708/0764

Owner name: KIM, YEUNG WON JUNE, KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KIM, SEONG JIN;JUNG, HAN SUNG;TAKAHASHI, SATORU;SIGNING DATES FROM 20200916 TO 20200917;REEL/FRAME:057708/0764

Owner name: KIM, SAEROM, KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KIM, SEONG JIN;JUNG, HAN SUNG;TAKAHASHI, SATORU;SIGNING DATES FROM 20200916 TO 20200917;REEL/FRAME:057708/0764

Owner name: KIM, SEONG JIN, KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KIM, SEONG JIN;JUNG, HAN SUNG;TAKAHASHI, SATORU;SIGNING DATES FROM 20200916 TO 20200917;REEL/FRAME:057708/0764

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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