WO2012064090A2 - Chemical preparation method of skeletal muscle-derived multipotent cells and use thereof - Google Patents

Chemical preparation method of skeletal muscle-derived multipotent cells and use thereof Download PDF

Info

Publication number
WO2012064090A2
WO2012064090A2 PCT/KR2011/008473 KR2011008473W WO2012064090A2 WO 2012064090 A2 WO2012064090 A2 WO 2012064090A2 KR 2011008473 W KR2011008473 W KR 2011008473W WO 2012064090 A2 WO2012064090 A2 WO 2012064090A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
differentiated
cell
differentiation
pluripotent
Prior art date
Application number
PCT/KR2011/008473
Other languages
French (fr)
Korean (ko)
Other versions
WO2012064090A9 (en
WO2012064090A3 (en
Inventor
알. 윌리암스다런
정다운
Original Assignee
광주과학기술원
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 광주과학기술원 filed Critical 광주과학기술원
Publication of WO2012064090A2 publication Critical patent/WO2012064090A2/en
Publication of WO2012064090A3 publication Critical patent/WO2012064090A3/en
Publication of WO2012064090A9 publication Critical patent/WO2012064090A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • 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
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/34Muscles; Smooth muscle cells; Heart; Cardiac stem cells; Myoblasts; Myocytes; Cardiomyocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • 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
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources
    • 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
    • C12N2501/06Anti-neoplasic drugs, anti-retroviral drugs, e.g. azacytidine, cyclophosphamide
    • 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
    • C12N2501/40Regulators of development
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1323Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from skeletal muscle cells

Definitions

  • the present invention provides a method for chemical preparation of skeletal muscle-derived pluripotent cells.
  • Dedifferentiation is a phenomenon in which differentiated cells acquire 'stem cells'-like or multipotent state before they differentiate into one or more different tissue forms [1]. All species have natural differentiation as part of their ability to regenerate tissues [2]. For example, acinar cells or duct eel is present in the pancreas of mammals can be reversed to produce insulin-expressing beta-cells [3].
  • retrodifferentiation varies among species, and systematically primitive vertebrates have greater retrodifferentiation.
  • the cells of the central nervous system of amphibians and fish can dedifferentiate [4].
  • the retina [5], lens [6] and limbs [groups] of the urodele amphibians undergo differentiation during the entire regeneration of the tissues.
  • the greatest retrodifferentiation is found in plants. For example, tobacco / coi / aa MeS0 phyll protoplasts cause reverse differentiation as part of the reprogramming process to generate whole plants [8].
  • Phenotype-based screening methods for small molecular libraries have been carried out to find novel regulators associated with intracellular dedifferentiation [14, 15].
  • An excellent example is the discovery of myoseverin, a tubulin-binding molecule identified from a 2,6,9-trisubstituted purines library [14, 16].
  • the molecular scaffold of myosevers is based on olomoucine, a known inhibitor of cyclin-dependent kinase 2, a cell cycle regulatory protein.
  • Myose verbin has been reported to promote reverse differentiation in a C2C12 mouse myoblast model for skeletal muscle differentiation.
  • CDKNs cycl in-dependent kinase inhibitors
  • pl8 also known as CDKN2C or INK4C
  • p21 also known as CIPl or CDK 1A
  • p27 also known as CDKNlB or KIP1
  • P57 also known as CDK 1C or KIP2 [23-25].
  • the inventors have tried to produce pluripotent cells (nrnltipotent eel Is). As a result, the present inventors induced down-regulation of p21 expression in differentiated cells obtained from skeletal muscle tissue having a very high refractory to obtain pluripotent cells and their differentiation capacity (eg, back-differentiation into muscle cells). The present invention was completed by finally confirming dedifferentiation, transdifferentiation into adipocytes or bone cells.
  • Another object of the present invention is to provide a pluripotent cell.
  • Another object of the present invention is to provide a method for regenerating a mammalian tissue or organ.
  • the present invention provides a method for chemically preparing a pluripotent cell (mult i potent eel Is) from mammalian differentiated cells comprising the steps of: a) obtaining differentiated cells from a mammal; ; And (b) inducing down-regulation of p21 expression in the differentiated cells to treat pluripotent cells after treatment with a reverse differentiation agent ((16 £ ⁇ 6! 1 3 ⁇ 011 agent) after induction of p21 low expression.
  • a reverse differentiation agent ((16 £ ⁇ 6! 1 3 ⁇ 011 agent) after induction of p21 low expression.
  • the present inventors have tried to prepare multipotent cells.
  • the present inventors induced down-regulation of p21 expression in differentiated cells obtained from skeletal muscle tissue having a very high refractory to obtain pluripotent cells and their differentiation capacity (eg, back-differentiation into muscle cells). , Heterologous differentiation into adipocytes or bone cells) was finally confirmed.
  • the present invention describes for the first time a relatively simple and reversible chemical-based method that can induce reverse differentiation of mammalian skeletal muscle (eg, cell cycle re-entry and differentiation), and is a fully differentiated fire in mammals.
  • mammalian skeletal muscle eg, cell cycle re-entry and differentiation
  • the present invention obtains mononuclear cells that proliferate through the continuous treatment of small molecules (chemicals) and transient p21 suppression (suppress ions) in mammalian skeletal muscle tissue having very high refractory properties.
  • pluripotent cells can be obtained through step-wise treatment of reversin, a small molecule, to proliferating mononuclear cells derived from muscle fibers.
  • the chemical preparation of the pluripotent cells of the present invention will be described in detail by dividing each step as follows: Step (a): Obtaining differentiated cells from a mammal
  • differentiated cells are obtained from a mammal.
  • the term 'mammal' includes, but is not limited to, humans, mice, rats, guinea pigs, rabbits, monkeys, pigs, horses, cattle, sheep, antelopes, dogs, and cats.
  • the differentiated cells are terminally differentiated cells
  • the term 'terminal differentiated cells' refers to cells at the endpoint stage during development, and has a mature phenotype with a characterized phenotype. Says a cell. Differentiated cells are distinguished from other cell types but have a fishery characteristic or function and have a limited ability to proliferate.
  • the differentiated cells are one cell selected from the group consisting of skeletal muscle cells, cardiomyocytes, smooth muscle cells, skin cells, chondrocytes, adipocytes and osteocytes, more preferably the differentiated cells are skeletal muscle cells, One cell selected from the group consisting of cardiomyocytes and smooth muscle cells, most preferably said differentiated cells are skeletal muscle cells.
  • the differentiated cells are connective tissue, nerve cells, lymph cells, vasculature cells, kidney cells, pancreas cells, lung cells, urethral cells, bladder cells, gastric cells, liver cells, small intestine cells, colon cells and esophageal cells.
  • step ( a ) of the present invention comprises a method of incorporating a cell-derived agent into the differentiated fusion to segment the syncytia derived from the muscle cells of the mammal and obtain mononuclear cells or eel lulates. This is done by treating cellularization-inducing agent.
  • a mononuclear cell or a cell material is prepared by treating a skeletal muscle fusion with a cellization-inducing agent.
  • syncytia refers to a multinuclear cell having a large cell-like structure filled with cytoplasm containing many nuclei, and in eukaryotes, most cells have a single nucleus. In that sense the fusion is a very unusual form.
  • the cell-derived agent of the present invention is myosebine, myosebine B, colchicine, nocodazole, allocolchin taxane, grapephylolock, combretastatin, steg Nacin, dihydroxy-pentamethoxy ananone, 2- methoxyestradiol (2-ME) and its analogues, benzimidazole carbamate, ansamitocin, TN16, vinbla Stin, vincristine, dolastatin, curacin A, etoposide, teniposide, sanguinarine and Chelidonineol, more preferably, myosebergin, myosebergin B, colchicine and nocodazole, even more preferably myosebergin and myosebergin B; And most preferably is myo
  • the present invention identifies myosebers identified using a purine-based compound library to identify novel regulators associated with intracellular dedifferentiation.
  • myoseber a potent microtubule-degrading compound, using a 2,6,9-alternative purine library constructed by Combinatorial Chemistry [14].
  • the low cytotoxicity of myoseberg and its analogs allows it to be applied as a cytostatic antitumor agent.
  • Myoserine used in the present invention is a small chemical molecule identified from the 2,6,9-trisubstituted purines library, and has the following chemical structure ( ⁇ ):
  • Myoserine is based on olomoucine, a purine derivative known as an inhibitor of cyclin-dependent kinase (CDK) 2 in cell cycle regulatory proteins, whose chemical structure ( ⁇ ) is as follows:
  • Myosebern not only restrains the cell cycle in the G 2 / M transit ion by disrupting spindle assembly, but also reverses fully differentiated muscle cells to environmental factors, resulting in muscle regeneration and enjoyable cell differentiation. Can be applied.
  • the present invention induces cellularization of the multinucleated skeletal muscle root canal by treating the skeletal muscle fusion described above with a cytolation-inducing agent. More specifically, root canal cultures were treated with 10 uM myosebine, 15 iiM myosevery B, 1 yM colchicine, 1 nocodazole or 10 ⁇ Taxol for 24 hours to obtain a cellized root canal. To remove relatively large amounts of intracellular debris in addition to the cells, they were centrifuged after trypsinization and filtered through a 70 m mesh. As a result, treatment with 10 ⁇ myoseberg produced the largest number of monocytes (see Figure lc).
  • the myoseberg of the invention is treated with 5-50 ⁇ , more preferably 5-30 ⁇ , even more preferably 5 ⁇ 20 ⁇ , most preferably 10-15 ⁇ do.
  • the differentiated cells are then treated with p21 down-regulatory inducing compounds.
  • p21 plays an important role in regulating nuclear events due to environmental stress, p21 is induced at very high levels due to oxidative stress and DNA damage.
  • the p21 protein acts as an inhibitor of cyclin-dependent kinases and effectively blocks cell cycle progression, so it is overexpressed in aging or fully differentiated cells.
  • down-p21 expression Regulation induces myofibrillarization and cell cycle reentry to mimic the early stages of adnexal regeneration of Rhesus amphibians.
  • the p21 down-regulatory inducing compounds of the present invention are compounds that induce down-regulation by inhibiting the expression of the p21 gene, inhibiting the activity of the p21 protein or decreasing the stability of the p21 protein.
  • down-regulation of p21 expression of the present invention can be carried out using any method known in the art, for example using antisense oligonucleotides, siRNA, miRNA, or natural product extracts. It may be carried out by, but is not limited thereto.
  • siRNA in the present invention means a nucleic acid molecule capable of mediating RNA interference or gene silencing (see TO 00/44895, WO 01/36646, WO 99/32619, WO 01/29058, WO 99 / 07409 and W0 00/44914).
  • siRNA is provided as an efficient gene knock-down method or gene therapy method because it can inhibit the expression of the target gene.
  • siRNA was first discovered in plants, worms, fruit flies and parasites, but has recently been used in mammalian cell research by developing / using siRNAs (Degot S, et al. 2002; Degot S, et al. 2004; Ballut L, et al. 2005).
  • siRNA molecule of the present invention may have a structure in which a sense strand (corresponding sequence corresponding to a p21 m NA sequence) and an antisense strand (a sequence complementary to a p21 mRNA sequence) are positioned opposite to each other to form a double strand. Further, according to another embodiment, siRNA molecules of the present invention may have a single chain structure having self-complementary sense and antisense strands.
  • siRNAs are not limited to fully paired double-stranded RNA portions of RNA, but are paired by mismatches (bases not complementary), bulges (no bases on one chain), and the like. May be included.
  • the total length is 10 to 100 bases, preferably 15 to 80 bases, more preferably 20 to 70 bases.
  • the siRNA terminal structure is capable of both blunt terminal black and cohesive terminal as long as the expression of the p21 gene can be inhibited by the R Ai effect. Cohesive end structures are possible with both 3'-end protrusion structures and 5'-end protrusion structures.
  • the siRNA molecules of the present invention may have a form in which a short nucleotide sequence (eg, about 5-15 nt) is inserted into a self-complementary sense and antisense strand, wherein the siRNA molecule formed by expression of the nucleotide sequence is a molecule Due to the internalization, the hairpin structure is formed, and the stem-and-loop structure as a whole is formed. This stem-and-loop structure can be processed in vitro or in ⁇ to produce an active siRNA molecule capable of mediating RNAi.
  • antisense oligonucleotide refers to DNA or RNA or a derivative thereof containing a nucleic acid sequence complementary to a sequence of a particular mRNA, and binds to a complementary sequence in the mRNA to inhibit translation of the mRNA into a protein. It works.
  • An antisense sequence of the invention refers to a DNA or RNA sequence that is complementary to P 21 and capable of binding to p21 mRNA, and is responsible for the translation of p21 mRNA, translocation into the cytoplasm, maturation or any other overall biological function. May inhibit essential activity.
  • the antisense nucleic acid has a length of 6 to 100 bases, preferably 8 to 60 bases, and more preferably 10 to 40 bases.
  • miRNA refers to a single-stranded RNA molecule of 21-25 nucleotides, and is a modulator that controls gene expression in eukaryotes through inhibition of the disruption or translation of target mRNAs.
  • This miRNA consists of two stages of processing.
  • the first miRNA transcript (primary miRNA) is made into the stem-loop structure of 70-90 bases, or pre-miRNA, by the RNaseEI type enzyme called Drosha in the nucleus, and then migrates into the cytoplasm and is called Dicer. Cleaved to make up to 21-25 bases of mature miRNA.
  • the miRNA thus produced complementarily binds to the target mRNA and acts as a post-transcript ional gene suppressor, inducing translation inhibition and mRNA destabilization. miRNAs are involved in a variety of physiological phenomena and diseases.
  • down-regulation of p21 expression in the present invention induces the proliferation of cellized root canal or re-entry and differentiation into the cell cycle.
  • the cellized root canal of the present invention maintains its inherent myogenic potential even after down-regulation of p21 expression.
  • the pluripotent cells obtained by the method of the invention are cells of the same lineage or germ layer as the differentiated cells used in step (a).
  • the pluripotent cells obtained by the present invention are cells of the same germ cells of skeletal muscle, such as skeletal muscle, bone, dermis, connective tissue, urogenital system, heart, blood (lymph cells), kidney and It is a pluripotent cell that can differentiate into the cells that make up the spleen.
  • the differentiated cells used in step (a) are skeletal muscle cells, and the pluripotent cells obtained in step (b) can be differentiated into skeletal muscle cells, adipocytes or osteocytes. to be.
  • p21 down-regulated differentiated cells are treated with a reverse differentiation agent.
  • the term “dedifferentiation agent” refers to an agent that promotes the dedifferentiation of one or more cell types, and includes chemicals, nucleic acids, peptides, polypeptides, small organic molecules, antibody antisense oligonucleotides, RNAi constructs. And ribozymes.
  • Various retrodifferentiating agents can induce retrodifferentiation by contacting one or more retrodifferentiating factors with cells in vivo or int ⁇ for a time sufficient to induce retrodifferentiation in fully differentiated mammalian cells.
  • the reverse differentiation agent which can be used in the present invention is reversin (N6-cyclonucleus-N2- -morpholin-4-yl-phenyl) -9H-purine-2 6-diamine) Trichostatin A, valproic acid, buphenyl, and 5-azacytidine (5_azacyt idine), but are not limited thereto. Most preferably, it is reversin.
  • Reversin is a 2,6-replaced purine compound with the following chemical structure (IV) and capable of proliferating and re-differentiating myogenic cells (eg, proliferation into bone and fat). And re-differentiation).
  • the pluripotent cells of the present invention can differentiate into myogenic lineage, adipogenic lineage or osteogenic lineage.
  • Yamanaka and co-workers produced embryonic stem cells similar to embryonic stem cells in 2006 and named them iPSCs (Kazutoshi Takahashi and Shiny a Yamanaka, Cell, 126: 663-676 (2006)).
  • the study provided the basis for producing patient- and disease-specific customized pluripotent pluripotent cells without ethical problems such as embryo destruction or egg donation, but the stability and differentiation ability of established pluripotent stem cells were significantly reduced. It is true.
  • small molecules eg, myose verbine of the present invention.
  • the chemical-based approach of the present invention is important for the development of strategies for inducing limb regeneration in mammals, and a novel approach for the development of iPSCs from fully differentiated afferent tissues. present.
  • the method is followed by treatment of the differentiation inducing agent to the cells obtained in step (b) after treatment with the dedifferentiation agent of step (b) to another cell lineage of the same three germ lineage as the differentiated cells of step (a). And further comprising the step of differentiating.
  • the three germ layers are composed of mesoderm mesoderm and ectoderm, endothelial cells in the stomach, colon, liver pancreas, bladder, the inside of the urethra, the epithelial part of the airway, lung, pharynx, thyroid gland, parathyroid gland and small intestine tissues,
  • the mesoderm develops the cells that make up skeletal muscle, bone, dermis, connective tissue, urogenital system, heart, blood (lymph cells), kidneys and spleen. Pigment cells, head connective tissues, epidermis, hair and cells that make up the mammary gland develop.
  • the term 'differentiation inducing agent' refers to growth factors, hormones, and the like included in the culture medium to differentiate into the desired cells.
  • the differentiation inducing agent used to obtain the lipogenic cell lineage from the pluripotent pleasant cells obtained in step (b) is inslin, texamethasone, rosiglitazone,
  • IBMX Isobutylmethylxanthine
  • the differentiation inducer used to obtain osteogenic cell lineage from the pluripotent stem cells obtained in step (b) is ascorbic acid-2-phosphate, dexamethasone, ⁇ -glycerophosphate or a combination thereof. .
  • the pluripotent cells of the present invention can differentiate into myogenic lineage, adipogenic lineage or osteogenic lineage.
  • the invention provides a pluripotent cell prepared according to the method described above.
  • composition for treating myopathy comprising muscle forming cell lineage or muscle cells differentiated from pluripotent cells prepared according to the method of the present invention as an active ingredient.
  • the term 'muscle disease' refers to muscle fatigue, muscular dystrophy, accidental or surgical wounds or muscular dystrophy
  • the term 'muscular dystrophy' refers to Becker's muscul r dystrophy, congenital muscular dystrophy (congenital) muscular dystrophy), Duc ' henne muscular dystrophy, distal muscular dystrophy, Emery-Dreifuss muscular dystrophy, facial shoulder dystrophic muscular dystrophy, muscular dystrophy, muscular dystrophy Muscular dystrophy, myotonic muscular dystrophy, sarcoglycanism, spinal muscular atrophy or Brown-Vial etto-Van Laere syndrome (BL).
  • BL Brown-Vial etto-Van Laere syndrome
  • composition for treating bone diseases comprising osteogenic cell lineage differentiated from pluripotent cells prepared according to the method of the present invention as an active ingredient.
  • the term 'bone disease' refers to osteoporosis, juvenile osteoporosis, osteogenic imperfecta, hyperosclerosis, hypercalcemia, hyperparathyroidism, osteomalacia, soluble bone disease, osteonecrosis, Paget's disease of bone, bone development Diseases, bone fractures, bone loss due to rheumatoid arthritis, inflammatory rheumatoid arthritis, osteomyelitis, metastatic bone disease, periodontal bone loss, rickets, bone loss by cancer or senile loss of bone.
  • the composition of the present invention comprises ( a ) a pharmaceutically effective amount of cells differentiated from the pluripotent or pluripotent cells described above (eg, myogenic cell groups, osteogenic cell groups); And (b) a pharmaceutically acceptable carrier.
  • pharmaceutically effective amount means an amount sufficient to achieve the efficacy or activity of cells differentiated from the pluripotent cells described above.
  • the pharmaceutical composition of the present invention includes a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers included in the pharmaceutical composition of the present invention are those commonly used in the preparation, lactose, dextrose, sucrose, sorbbi, manny, starch, acacia rubber, phosphate chame, alginate, Gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyridone, cellulose, water, syrup, metal cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil Including, but not limited to.
  • the pharmaceutical composition of the present invention may further include lubricants, wetting agents, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, etc. in addition to the above components.
  • lubricants such as talc, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, kaolin, sorbitol, sorbitol, sorbitol, etc.
  • compositions of the present invention may be administered parenterally, for example, by subcutaneous injection, intramuscular injection, transdermal administration, intraarticular injection, and the like.
  • Appropriate dosages of the pharmaceutical compositions of the present invention will vary depending on factors such as formulation method, mode of administration, age, weight, sex, morbidity of food, time of administration, route of administration, rate of excretion and response sensitivity. Can be prescribed. Typical dosages of the pharmaceutical compositions of the invention are 10 2 -10 10 cells per day on an adult basis.
  • composition of the present invention is known in the art
  • the invention provides a method of regeneration 20 of a mammalian tissue or organ comprising the following steps:
  • the differentiation is performed by adding a differentiation inducing agent of a desired tissue or organ to the culture medium of the pluripotent cells obtained in step (b).
  • the present invention relates to a method for chemical preparation of muscle-derived pluripotent cells and uses thereof.
  • the method of the present invention obtains pluripotent mononuclear cells or cells proliferating from differentiated mammalian skeletal muscle through continuous treatment of small molecules and transient p21 suppression, followed by stepwise treatment of reversin. It is a very simple and efficient way of obtaining cells.
  • the pluripotent cells of the present invention are capable of heterodifferentiation into adipogenic or bone forming cell groups as well as dedifferentiation into muscle forming cell groups.
  • FIG. 1 is a result showing the effects of AraC, 5-FU, and myoserine on C2C12 myoblasts.
  • FIG. La is a cell fusion index of the culture medium that differentiates after 50 ⁇ M AraC for 72 hours in differentiating C2C12 myoblast culture. increase the fusion index (ie, the number of cell nuclei inserted into the multinucleated root canal
  • FIG. 2 shows the effect of ⁇ 21 down-regulation in the cellized root canal.
  • 2A shows a schematic of an experimental protocol for observing the proliferative response of cellized root canals following p21 down-regulation.
  • FIG. 2B is a flow cytometric analysis of PKpropidium iodide staining showing that p21 down-regulation for 48 hours can induce proliferation in cellularized, male muscle fibers. That is, the shift to the G 2 / M phase of the cell cycle was observed.
  • 2C is a graph showing that p21 down-regulation for 48 hours induces short term cell division of the cellized C2C12 root canal. The data is representative of three independent experiments.
  • FIG. 2D shows the flow cytometry for BrdU insertion in the cellized C2C12 root canal, indicating that p21 downregulation for 48 hours induces DNA synthesis.
  • FIG. 2E shows that treatment of p21 down-regulation or reversin induces differentiation in cellized C2C12 root canal, as a reduced label (fluorescence) of FTTC- ⁇ -Burorogroxine, a ligand for acetylcholine receptor Detected.
  • FIG. 3 is a result showing that p21 down-regulation reverses the EMG decreased by reversin treatment.
  • 3B is a graph showing that p21 knockdown reverses the inhibitory effect of reversin in C2C12 cell water.
  • 3C is an optical microscopic analysis of muscle formation in C2C12 cultures derived from cellized root canals treated with 50 nM reversin or 50 nM reversin and 80 pmol p21 siRNA. Hemafocillin staining shows that more nuclei are located in the root canal fusions in cultures derived from C2C12 cells treated with p21 siRNA. Exemplary nuclei are indicated by arrows in the continuous tract of the root canal cytoplasm.
  • FIG. 4 shows that p21 down-regulation promotes the conversion of cellularized root canal to adipocytes.
  • FIG. 4A shows C2C12 cell water treated with p21 knock-down and reversin and cultured in adipose media can accumulate lipids.
  • C2C12 myoblasts treated with reversin and cultured in lipogenic medium also accumulated lipids [15].
  • C2C12 cell water treated with reversin and cultured in lipogenic medium did not accumulate lipid.
  • FIG. 4B is a microscopic analysis of lipid accumulation in C2C12 cells treated with p21 knock-down, reversin and lipogenic cocktails, similar to cytoplasmic staining in C2C12 myoblasts treated with reversin and lipogenic cocktails. Showed.
  • FIG. 4C shows C2C12 cells treated with p21 knock-down, reversin and lipogenesis cocktails after insulin stimulation (adipocyte-specific function). The results show an increased uptake of 6-NBDG, a fluorescent glucose analogue. Replacement of p21 siRNA with control siRNA failed to produce cells capable of ingesting insulin-stimulated glucose.
  • C2C12 myoblasts treated with reversin (500 nM) and lipogenic cocktails also showed increased uptake of 6-NBDG after insulin stimulation.
  • 6-NBDG intake was less than that observed in 3T3-L1 adipocytes after insulin stimulation.
  • the data is representative of three independent experiments. 4D shows the results of visualizing fluorescence microscopy of 6-NBDG uptake after insulin stimulation. 6-NBDG was found to accumulate in the membranes of C2C12 myoblasts and cell-derived adipocytes.
  • FIG. 4E shows C2C12 cells treated with p21 knock-down reversin (500 nM) and an lipogenesis cocktail showed insulin-sensitive free fatty acid release after epinephrine-stimulated (fat cell-specific function).
  • Replacement of p21 siRNA with control siRNA failed to produce cells with the adipocyte-specific function.
  • FIG. 5 shows that p21 down-regulation promotes bone formation in cellized root canals.
  • 5A is a graph showing increased alkaline phosphatase activity in C2C12 cell cultures cultured in bone formation conditions after reversin treatment and p21 knock-down. Replacing p21 siRNA with control siRNA resulted in a significant decrease in alkaline phosphatase activity.
  • ATCD5 cells which were cartilage progenitor cells cultured under osteogenic conditions, were used as positive controls.
  • Figure 5b is added to the cell lysate, and AttoPhos substrate solution ® picture obtained after 300 seconds. Experimental groups are described in wells (numbers) floating under microplates.
  • 5C shows Alizarin Red S staining visualizing mineralization in ATDC5 cells and C2C12 cells after incubation for 10 days in osteogenic medium.
  • C2C12 cells treated with reversin (500 nM) and p21 siRNA were 500 nil Reversin induced a degree of mineralization similar to C2C12 myoblasts treated. Substitution of p21 siRNA with control siRNA reduced mineralization.
  • 5D is the semi-quantitative result of mineralization via the color assay.
  • ATCD5 cells cartilage progenitor cells cultured under the bone formation cocktail.
  • FIG. 6 shows a schematic diagram of a chemical approach for pluripotent cells to pluripotency from fully differentiated skeletal muscle tissue. Proliferating myoblasts can be treated with AraC to enhance segmentation into multinucleated root canal fusions. [Specific contents to carry out invention]
  • p21 siRNA and control siRNA were purchased from Santa Cruz Biotechnology (CA, USA), ⁇ -bungarotoxin and 6- ( ⁇ — Nitrobenz-2-oxa-1, 3-diazol-4-yl) amino) -6-deoxyglucose (6-NBDG) was purchased from Molecular Probes (OR, USA).
  • C2C12 mouse Oros musculus myoblasts contained 10% fetal bovine serum (FBS; Life Technologies, CA, USA), 50 units / mL penicillin (Pen; Life Technologies., CA, USA) and 50 jug / ml strepto Mycin (Strep; Life Technologies., CA, USA) was maintained in a growth medium consisting of DMEM (Dulbecco's Modified Eagle's Medium; Life Technologies., CA, 0 USA) supplemented.
  • Myoblasts are 5% horse serum, --- 50 units / mL . Penicillin and -50- ⁇ g / mL -streptomycin supplemented with DMEM
  • Differentiation of skeletal muscle into myotubes was induced by incubation for 8 days in the growth medium. Differentiation medium was changed every 48 hours.
  • myoblasts were cultured in collagen-coated 6-well 5 tissue culture plates (Sigma, M0, USA). To enhance differentiation and remove contaminants, myoblast cultures were treated with 50 ⁇ AraC [26] for 72 hours or 10 ⁇ 5-FU [2 phases for 48 hours as previously reported. AraC or 5-FU treatment was performed 18 hours after the addition of differentiation culture medium.
  • 3T3-L1 mouse (/ s musculus) embryonic fibroblasts were maintained in proliferation medium consisting of DMEM supplemented with 10% FBS, 505 units / mL penicillin and 50 ⁇ g / mL straptomycin.
  • Differentiation of 3T3-L1 fibroblasts into adipocytes was induced as follows: Post-confluent cells at 48 hours (considered day 0) were treated with 10% FBS, 0.5 mM 3-isobutyl-1-methyl-xanthine.
  • ATDC5 cells were provided by Professor Jeon Jang-soo from the School of Life Sciences, Gwangju Institute of Science and Technology. ATDC5 cells were cultured in a maintenance medium consisting of a 1: 1 complex of 53 ⁇ 4> FBS, 10 / g / mL human transferrin and 3 ⁇ 1 (DMEM with ⁇ 8 M sodium selenite and Ham's F-12 medium). To induce chondrogenesis, 10 // g / mL bovine insulin was added to the cell culture medium, the batch was changed every 48 hours and chondrocytes were used for the experiment 8 days later.
  • P19 mouse embryonic carcinoma cells were provided by Professor Song Mi-Ryung, School of Life Sciences, Gwangju Institute of Technology. As reported previously [28], neurogenesis of P19 cells was performed through cell ball formation and treatment of 500 nM retinoic acid in non-adherent culture dishes. . To inhibit dividing growth into glial-like cells, 50 ⁇ AraC was complemented 48 hours after plating in culture medium. Skeletal Muscle Root Canal Cellization
  • Root canal cultures were treated with 10 ⁇ myoserine, 15 ⁇ myoserine ⁇ , 1 yM colchicine, 1 ⁇ nocodazole or 10 ⁇ taxol for 24 hours (treatment as reported in IC 50 mice for root canal degradation). Method [16]).
  • Cellization was found to produce a relatively large amount of intracellular debris in addition to monocytes. Trypsin was treated to the cell monolayer and gently centrifuged for 5 min at 750 rpm. The cell pellet was gently resuspended in 10 mL growth medium and passed through a 70 IM cell strainer (BD Biosciences, CA, USA) to remove debris.
  • the obtained cells were resuspended in DMEM at a density of 1 ⁇ 10 6 cells AnL and stained with 0.4% trypan blue die (Sigma ⁇ Aldrich, Mo, USA). 100 cells were counted with a hemocytometer (Mariefeld GmbH, Germany) and cells not stained with trypan blue were considered viable cells.
  • siRNA-mediated gene silencing Mononuclear cells were transfected with 50 pmol of the target siRNA or control siRNA according to the manufacturer's protocol (Santa Cruz Biotechnology, CA, USA). Monocytes 48 hours after transfection were used for the experiment. Quantification of Myoblast Differentiation Using Fusion Fusion Index
  • the cells or cells were reacted with 10 ⁇ BrdU for 24 hours and then obtained with trypsin / EDTA. . Afterwards, cells were allowed to spin at 1,000 rpm for 5 minutes. After centrifugation and washing with PBS, 1 ⁇ 10 6 cells were resuspended in 500 ⁇ 0.5% paraformaldehyde (Sigma, MO, USA) (in PBS). The cells were reacted for 20 minutes under ice, washed once with PBS, and then centrifuged at 1500 rpm for 5 minutes. The obtained cells were resuspended in 1 mL of freshly prepared 3N HCl / 0.5% Tween 20 (Sigma, MO, USA) and allowed to permeate at 25 ° C.
  • 3T3-L1 adipocytes or muscle-derived adipocytes were dispensed into 24-well plates (BD Bioscience, CA, USA) at a concentration of 2 ⁇ 10 5 cells / well. After 48 hours, cells were incubated for 3 hours in serum-free low glucose DMEM. After addition of 100 nM insulin and 100 ⁇ 6- ( ⁇ _ (7_nitrobenzium 2-oxa-1,3-diazol-4-yl) amino) -2-deoxyglucose (6—NBDG) to the cells , Reacted at 37 ° C for 30 minutes. Cells were then washed with PBS and lysed with 100 ⁇ potassium phosphate buffer ( ⁇ 10) containing 1% Triton X-100.
  • muscle-derived adipocytes were obtained by microscopic analysis of NBDG uptake.
  • epinephrine-stimulated free fatty acid (FFA) release from adipocytes was measured using the free fatty acid quantification kit (Biovision, CA, USA). 3T3-L1 adipocytes grown until reaching confluence in 24-well plates were incubated for 3 hours in serum-free culture medium. Then, 10 ⁇ dissolved in salt in fat cells Epinephrine (Sigma, MO, USA) was treated to induce FFA release. The desired drug was added 10 minutes before the epinephrine treatment. Cell lysates were obtained and a 50 ⁇ organic phase was used for the chromogenic assay (VERSA Max microplate reader, Molecular Devices, Calif., USA). Palmitic acid was used to make a standard curve. Alkaline phosphatase assays
  • Alizarin red staining was quantified as described previously [33]. Briefly, after Alizarin Red staining, 800 ⁇ of 10% (v / v) acetic acid was added to each well and the plates were reacted with stirring for 30 minutes at room temperature. A monolayer was obtained from the plate using a cell scraper and vortexed by transferring to a 1.5-mL microcentr i fuge tube with 10 (v / v) acetic acid. Load 500 ⁇ mineral oil into the slurry and for 10 minutes After heating to 85 ° C, it cooled rapidly on ice. The slurry was then centrifuged at 20,000 g for 15 minutes and the 500 ⁇ supernatant was transferred to a new microcentrifuge tube.
  • the scrambled siRNA and p21 siRNA were treated 24 hours after the monocytes generated by C2C12 root canal cell division were dispensed (FIG. 2A).
  • PKpropidium iodide) staining on the cell nuclei showed that p21 down-regulation induced a shift from refractory G 0 phase to G 2 -M phase of the cell cycle (FIG. 2B).
  • This effect was also confirmed in counting the number of cells observed at 40 ⁇ magnification with p21 down-regulation leading to a small, but significant increase in cell number (FIG. 2C).
  • the above results indicate that p21 down-regulation It was again confirmed by the observation that BrdU, an indicator of increased DNA synthesis, caused an increase in the site of the inserted cells (FIG. 2D).
  • the treatment of p21 induces proliferation of the cellized root canal without a decrease in myoelectric potential.
  • the root canal showed a higher cell fusion index (p. 3b and 3c) after p21 knock-down.
  • p21 down-regulation promotes the conversion of cellularized root canal to adipocytes:
  • Reversin-treated myoblasts can be differentiated into osteoblasts by culturing under a cocktail of osteogenic factors [15].
  • Gaining alkaline phosphatase activity results in conversion of bone formation in C2C12 myoblasts transfected with regulators of the bone formation process such as Wnt-1-induced secreted protein 1 and Bone morphogenet ic protein 2 Has been used as an indicator [38].
  • Wnt-1-induced secreted protein 1 and Bone morphogenet ic protein 2 has been used as an indicator [38].
  • culture of C2C12 root canal cellized under osteogenic factors did not induce conversion to osteogenic cells (FIGS. 5A and 5B).
  • alkaline phosphatase activity was shown to be relatively small but significantly increased compared to untreated C2C12 cells in the experimental group of the present study, the acquisition of the alkaline phosphatase activity was achieved by control of the cultivated root canal treated with scrambled siRNA. It is even more markedly increased than the activity seen in.
  • C2C12 cells after culture in P 21 knock-down, reversin treatment and bone formation medium showed an increase in mineralization to a similar extent as C2C12 myoblasts cultured under the same conditions. Mineralization levels were lower than observed in ATDC5 cells after osteogenic culture. Moreover, C2C12 cells treated with control siRNA instead of p21 siRNA resulted in a significantly reduced level of mineralization after bone formation culture.
  • the results presented in this study are the first description of a chemical-based protocol that mimics the early stages of limb regeneration in amphibians.
  • Previously Experimental approaches have been developed to induce adult cells to form into pluripotent or stem cells. Examples include somatic cell nuclear transfer and transfection of transcription factors associated with omnipotence [45].
  • the above approaches did not utilize fully differentiated adult cells, which accounted for the majority of the eel hilar population in humans [46].
  • the above approaches currently face technical problems such as low efficiency.
  • the chemical approach to obtaining pluripotency described in the present invention can de-differentiate and cell lineage conversion ⁇ -in skeletal muscle tissue, which is considered to have a very high atrophy [22].
  • reversin-treated muscle cells It may be possible to additionally 'push' the adipocyte-forming process by modifying the cocktail of lipogenic factors, e.g. to thiazolidine, which activates the nuclear chamber adipogenic factor, peroxysome growth factor-activated receptor. Treatment of triazolidinediones [48].
  • the chemically defined method of the present invention for producing pluripotent cells from differentiated muscle has numerous viable fields for future research. For example, it has been reported that myoblasts can form neuronal cells by reversin treatment and culture in neuronal differentiation medium [43]. Similar 'jumps' from mesodermal lineage cells (eg, adipocytes and bone cells) to neuroectodermal lineage cells could be possible in cellized muscle fibers. Optionally... Muscle biopsies can be performed according to the chemical-based protocols described in this study in an attempt to produce patient-specific pluripotent cells. For example, lateral myoskeletal muscle strips can be obtained from a patient under local anesthesia.
  • Tsonis PA Regeneration of the vertebrate lens and other eye structures. London .: Nature Pub 1. [www.els.net], 1999.
  • Tsonis PA Regeneration of the lens in amphibia. . Heidelberg- ' Springer ' Ver lag, 2000.
  • Gal ant amine prevents apoptosis induced by beta- amyloid and thapsigargin: involvement of nicotinic acetylcholine receptors. Neuropharmacology 2004; 46: 103-114.

Abstract

The present invention provides a chemical preparation method of skeletal muscle-derived multipotent cells comprising: (a) a step for obtaining differentiated cells from mammals; and (b) a step for obtaining multipotent cells by treating the differentiated cells with a dedifferentiation agent after inducing the down-regulation of p21 expression of the differentiated cells. The multipotent cells of the invention enable not only dedifferentiation into a musculogenesis cell population but also the transdifferentiation into an adipogenesis cell population or an osteogenesis cell population. Therefore, the chemical-based approach method of the invention is important in the development of strategies for inducing limb regeneration in mammals. In addition, the invention discloses a new approach method for the development of induced pluripotent stem cells (iPSCs) from completely differentiated refractory tissues.

Description

【명세서】  【Specification】
【발명의 명칭】  [Name of invention]
골격근-유래된 다능성 세포의 화학적 제조방법  Chemical preparation of skeletal muscle-derived pluripotent cells
【기술 분야】 [Technical field]
본 발명은 골격근-유래된 다능성 세포의 화학적 제조방법  The present invention provides a method for chemical preparation of skeletal muscle-derived pluripotent cells.
용도에 관한 것이다. It is about a use.
【배경 기술】 [Background technology]
역분화 (dedifferentiation)는 분화된 세포가 하나 이상의 서로 다른 조직 형태로 분화하기 전에 '줄기세포' -유사 또는 다능성 세포 상태 (multipotent state)를 획득하는 현상에 대한 것이다 [1]. 모든 종은 조직을 재생하는 능력의 일부로서 자연적인 분화능을 가진다 [2]. 예를 들어, 포유동물의 췌장에서 존재하는 선방세포 (acinar cells) 또는 관세포 (duct eel Is)는 인술린 -발현 베타-세포를 생산하기 위해 역분화할 수 있다 [3]. 하지만, 역분화능은 종들 간에 차이가 있으며, 계통학적으로 원시적인 척추동물들이 더 큰 역분화능을 보유하고 있다. 예를 들어, 양서류 및 어류의 중추신경계의 세포들은 역분화할 수 있다 [4]. 더욱이, 유미목 (urodele) 양서류의 망막 [5], 수정체 [6] 및 사지 [기는 상기 조직들의 전체 재생 과정 동안 역분화를 거친다. 하지만, 가장 큰 역분화능은 식물에서 발견된다. 예를 들어, 담배 /coi/a a
Figure imgf000003_0001
엽육 원형질체 (meS0phyll protoplasts)는 전체 식물체를 발생시키기 위한 재프로그래밍 과정의 일부로서 역분화를 일으킨다 [8].
Dedifferentiation is a phenomenon in which differentiated cells acquire 'stem cells'-like or multipotent state before they differentiate into one or more different tissue forms [1]. All species have natural differentiation as part of their ability to regenerate tissues [2]. For example, acinar cells or duct eel is present in the pancreas of mammals can be reversed to produce insulin-expressing beta-cells [3]. However, retrodifferentiation varies among species, and systematically primitive vertebrates have greater retrodifferentiation. For example, the cells of the central nervous system of amphibians and fish can dedifferentiate [4]. Moreover, the retina [5], lens [6] and limbs [groups] of the urodele amphibians undergo differentiation during the entire regeneration of the tissues. However, the greatest retrodifferentiation is found in plants. For example, tobacco / coi / aa
Figure imgf000003_0001
MeS0 phyll protoplasts cause reverse differentiation as part of the reprogramming process to generate whole plants [8].
1990 년대에, 조합화학 (combinatorial chemistry) 분야에서의 진보는 알려진 생물활성 화합물 (bioactive compounds)의 분자 스캐폴드에 기초한 작은 분자 라이브러리의 대규모 생산을 촉진시켰다 [9]. 최근에, 본 발명자들 및 다른 연구자들이 다양한 생물학적 과정들의 신규한 조절인자들 (regulators)을 동정하기 위해 상기 접근방법을 이용하였다 [1으 12]ᅳ 세포내 과정들을 조절함에 있어 작은 분자들의 이용은 수많은 장점들을 가진다 [13]. 전형적으로, 작은 분자들은 단백질 기능에 대한 일시적인 조절을 제공하고 종종 이의 역도 가능하며, 이들의 효과는 작은 분자의 농도를 변화시킴으로써 정밀하게 조절될 수 있다. 또한, 단일 작은 분자는 세포 내 다양한 특이 타겟들을 동시에 조절하는 기능 (potential)을 가진다. 상술한 접근방법은 바람직한 시너지 효과를 통해 소망하는 표현형을 생산할 수 있다. In the 1990s, advances in the field of combinatorial chemistry have facilitated large-scale production of small molecular libraries based on molecular scaffolds of known bioactive compounds [9]. Recently, we and other researchers have used this approach to identify novel regulators of various biological processes. [1] 12 The use of small molecules in regulating intracellular processes has Has numerous advantages [13]. Typically, small molecules provide temporary regulation of protein function and are often vice versa, and their effects are small. It can be precisely controlled by changing the concentration of the molecule. In addition, a single small molecule has the potential to simultaneously regulate various specific targets in the cell. The above approach can produce the desired phenotype through desirable synergies.
작은 분자 라이브러리에 대한 표현형-기반된 스크리닝 방법은 세포내 역분화와 관련된 신규한 조절인자들을 발견하기 위해 실시되었다 [14, 15]. 그 탁월한 예가 2,6,9-대체된 퓨린 (2,6,9-trisubstituted purines) 라이브러리로부터 동정된 튜블린 -결합 분자인 미오세버린 (myoseverin)의 발견이었다 [14, 16]. 미오세버린의 분자적 스캐폴드는 세포주기 조절 단백질인 사이클린-의존성 키나제 2 의 알려진 억제제인 올로뮤신 (olomoucine)에 기초한다. 미오세버린은 골격근 분화에 대한 C2C12 마우스 근육모세포 모델에서 역분화를 촉진하는 것으로 보고되었다. 상술한 결과는 다핵화된 근섬유 (myofibers)의 단편화 (fragmentation)를 통한 단핵세포의 증식이 유미목 양서류에서 사지 재생의 중요한 특징이기 때문에 유의하였다 [17-19]. 하지만, 골격근 분화에 대한 pmi28 마우스 근육모세포 모델에서의 이후 연구는 미오세버린만이 근육 섬유 (muscle fibers)의 세포화 (cellularization)를 유도할 수 있다는 것을 제안하였다 [20]. 세포화된 단핵세포는 증식하지 않은 채로 남아있어 역분화를 일으킬 수 없었다.  Phenotype-based screening methods for small molecular libraries have been carried out to find novel regulators associated with intracellular dedifferentiation [14, 15]. An excellent example is the discovery of myoseverin, a tubulin-binding molecule identified from a 2,6,9-trisubstituted purines library [14, 16]. The molecular scaffold of myosevers is based on olomoucine, a known inhibitor of cyclin-dependent kinase 2, a cell cycle regulatory protein. Myose verbin has been reported to promote reverse differentiation in a C2C12 mouse myoblast model for skeletal muscle differentiation. The above results were significant because proliferation of monocytes through fragmentation of multinucleated myofibers is an important feature of limb regeneration in Rhesus amphibians [17-19]. However, a later study in the pmi28 mouse myoblast model for skeletal muscle differentiation suggested that only myoseber can induce cellularization of muscle fibers [20]. Cellized monocytes remained unproliferated and could not cause reverse differentiation.
유미목 양서류에서 사지 재생 과정 동안, 다핵성 골격근 섬유의 핵들은 세포주기에 재진입한다 [21]. 이와 대조적으로, 최종적으로 분화된 포유동물의 골격근 세포의 핵들은 실험적으로 세포주기의 재활성화를 유도하는 것이 상대적으로 어렵다 [22]. 많은 사이클린-의존성 키나제 억제제들 (cycl in-dependent kinase inhibitors, CDKNs)은 골격근 최종 분화의 유지에 포함되어 있는데, 예를 들어 pl8(CDKN2C 또는 INK4C 로도 알려짐), p21(CIPl 또는 CDK 1A 로도 알려짐), p27(CDKNlB 또는 KIP1 로도 알려짐) 및 P57(CDK 1C 또는 KIP2 로도 알려짐)을 포함한다 [23-25] . 최근 연구결과는 외인성 성장인자들의 부재 하에서도 p21 CDKN 의 억제만으로 포유동물 골격근에서 세포주기를 재활성화시키는 데 충분하다는 것을 나타냈다 [24]. 본 명세서 전체에 걸쳐 다수의 논문 및 특허문헌이 참조되고 그 인용이 표시되어 있다. 인용된 논문 및 특허문헌의 개시 내용은 그 전체로서 본 명세서에 참조로 삽입되어 본 발명이 속하는 기술 분야의 수준 및 본 발명의 내용이 보다 명확하게 설명된다. During limb regeneration in Rhesus amphibians, the nuclei of multinucleated skeletal muscle fibers reenter the cell cycle [21]. In contrast, the nuclei of finally differentiated mammalian skeletal muscle cells are relatively difficult to experimentally induce cell cycle reactivation [22]. Many cycl in-dependent kinase inhibitors (CDKNs) are involved in the maintenance of skeletal muscle final differentiation, for example pl8 (also known as CDKN2C or INK4C), p21 (also known as CIPl or CDK 1A), p27 (also known as CDKNlB or KIP1) and P57 (also known as CDK 1C or KIP2) [23-25]. Recent studies have shown that in the absence of exogenous growth factors, inhibition of p21 CDKN alone is sufficient to reactivate the cell cycle in mammalian skeletal muscle [24]. Throughout this specification, many papers and patent documents are referenced and their citations are indicated. The disclosures of cited papers and patent documents are incorporated herein by reference in their entirety, and the level of the technical field to which the present invention belongs and the contents of the present invention are more clearly explained.
【발명의 내용】 [Content of invention]
【해결하려는 과제】  [Problem to solve]
본 발명자들은 다능성 세포 (nrnltipotent eel Is)를 제조하기 위해 연구 노력하였다. 그 결과, 본 발명자들은 불웅성 (refractory)이 매우 높은 골격근 조직으로부터 얻은 분화세포에 p21 발현의 하향-조절을 유도하면, 다능성 세포를 얻을 수 있고 이들의 분화능 [예컨대, 근육세포로의 역분화 (dedifferentiation), 지방세포 또는 골세포로의 이형분화 (transdifferentiation)]을 최종적으로 확인함으로써, 본 발명을 완성하게 되었다.  The inventors have tried to produce pluripotent cells (nrnltipotent eel Is). As a result, the present inventors induced down-regulation of p21 expression in differentiated cells obtained from skeletal muscle tissue having a very high refractory to obtain pluripotent cells and their differentiation capacity (eg, back-differentiation into muscle cells). The present invention was completed by finally confirming dedifferentiation, transdifferentiation into adipocytes or bone cells.
따라서, 본 발명의 목적은 다능성 세포의 화학적 제조방법을 제공하는 것에 있다.  Accordingly, it is an object of the present invention to provide a method for chemically preparing pluripotent cells.
본 발명의 다른 목적은 다능성 세포를 제공하는 데 있다.  Another object of the present invention is to provide a pluripotent cell.
본 발명의 또 다른 목적은 포유동물 조직 또는 기관의 재생방법을 제공하는 데 있다. 본 발명의 다른 목적 및 이점은 하기의 발명의 상세한 설명, 청구범위 및 도면에 의해 보다 명확하게 된다.  Another object of the present invention is to provide a method for regenerating a mammalian tissue or organ. Other objects and advantages of the present invention will become apparent from the following detailed description, claims and drawings.
【과제의 해결 수단】 [Measures of problem]
본 발명의 일 양태에 따르면, 본 발명은 다음의 단계를 포함하는 포유동물 분화세포로부터 다능성 세포 (mult i potent eel Is)의 화학적 제조방법을 제공한다: ) 포유동물부터 분화세포를 수득하는 단계; 및 (b) 상기 분화세포에 p21 발현의 하향-조절을 유도하여 p21 저발현 유도 후 역분화제((16 £^6!1 3^011 agent)를 처리하여 다능성 세포를 수득하는 단계 . 본 발명자들은 다능성 세포 (multipotent cells)를 제조하기 위해 연구 노력하였다. 그 결과, 본 발명자들은 불웅성 (refractory)이 매우 높은 골격근 조직으로부터 얻은 분화세포에 p21 발현의 하향-조절을 유도하면, 다능성 세포를 얻을 수 있고 이들의 분화능 (예컨대, 근육세포로의 역분화, 지방세포 또는 골세포로의 이형분화)을 최종적으로 확인하였다. According to one aspect of the present invention, the present invention provides a method for chemically preparing a pluripotent cell (mult i potent eel Is) from mammalian differentiated cells comprising the steps of: a) obtaining differentiated cells from a mammal; ; And (b) inducing down-regulation of p21 expression in the differentiated cells to treat pluripotent cells after treatment with a reverse differentiation agent ((16 £ ^ 6! 1 3 ^ 011 agent) after induction of p21 low expression. The present inventors have tried to prepare multipotent cells. As a result, the present inventors induced down-regulation of p21 expression in differentiated cells obtained from skeletal muscle tissue having a very high refractory to obtain pluripotent cells and their differentiation capacity (eg, back-differentiation into muscle cells). , Heterologous differentiation into adipocytes or bone cells) was finally confirmed.
본 발명은 포유동물 골격근의 역분화 (예컨대, 세포주기 재 -진입 및 분화)를 유도할 수 있는 상대적으로 간단하고 가역적인 화학물질-기반된 방법을 최초로 기재한 것으로, 포유동물의 완전 분화된 불웅성 조직으로부터 유도만능줄기세포 (iPSCs)의 개발을 위한 새로운 접근방법을 제시한다. - 본 발명에 따르면 본 발명은 매우 높은 불응성을 가지는 포유동물의 골격근 조직에 작은 분자 (화학물질)의 연속적인 처리 및 일시적인 p21 억제 (suppress ion)를 통해 증식하는 단핵세포를 얻는다. 이후, 근섬유로부터 유래된 증식하는 단핵세포에 작은 분자인 리버신의 단계적 처리 (step-wise treatment)를 통해 다능성 세포를 수득할 수 있다. 본 발명의 다능성 세포의 화학적 제조방법을 각각의 단계로 나누어 상세하게 설명하면 다음과 같다: 단계 (a): 포유동물로부터 분화세포를 수득  The present invention describes for the first time a relatively simple and reversible chemical-based method that can induce reverse differentiation of mammalian skeletal muscle (eg, cell cycle re-entry and differentiation), and is a fully differentiated fire in mammals. We present a new approach for the development of iPSCs from male tissues. According to the present invention the present invention obtains mononuclear cells that proliferate through the continuous treatment of small molecules (chemicals) and transient p21 suppression (suppress ions) in mammalian skeletal muscle tissue having very high refractory properties. Thereafter, pluripotent cells can be obtained through step-wise treatment of reversin, a small molecule, to proliferating mononuclear cells derived from muscle fibers. The chemical preparation of the pluripotent cells of the present invention will be described in detail by dividing each step as follows: Step (a): Obtaining differentiated cells from a mammal
본 발명에 따르면, 포유동물로부터 분화세포를 수득한다.  According to the present invention, differentiated cells are obtained from a mammal.
본 명세서에서, 용어 '포유동물' 은 인간, 마우스, 래트, 기니어 피그, 토끼, 원숭이, 돼지, 말, 소, 양, 영양, 개 및 고양이를 포함하지만, 이에 한정되는 것은 아니다.  As used herein, the term 'mammal' includes, but is not limited to, humans, mice, rats, guinea pigs, rabbits, monkeys, pigs, horses, cattle, sheep, antelopes, dogs, and cats.
바람직하게는, 상기 분화세포는 말기 분화세포 (terminally differentiated cell)이다,  Preferably, the differentiated cells are terminally differentiated cells,
본 명세서에서, 용어 '말기 분화세포' 는 발생 (development) 과정 중 종점 (endpoint) 단계의 세포로, 특성화된 표현형을 갖는 성숙한 (mature) 세포를 말한다. 분화세포는 다른 세포 종류와 구별되는 하나 어상의 특징 또는 기능을 갖으며, 증식할 수 있는 능력이 제한적이다. As used herein, the term 'terminal differentiated cells' refers to cells at the endpoint stage during development, and has a mature phenotype with a characterized phenotype. Says a cell. Differentiated cells are distinguished from other cell types but have a fishery characteristic or function and have a limited ability to proliferate.
바람직하게는, 상기 분화세포는 골격근 세포, 심근 세포, 평활근 세포, 피부 세포, 연골 세포, 지방 세포 및 골세포로 구성된 군으로부터 선택되는 하나의 세포이고, 보다 바람직하게는 상기 분화세포는 골격근 세포, 심근 세포 및 평활근 세포로 구성된 군으로부터 선택되는 하나의 세포이며, 가장 바람직하게는 상기 분화세포는 골격근 세포이다.  Preferably, the differentiated cells are one cell selected from the group consisting of skeletal muscle cells, cardiomyocytes, smooth muscle cells, skin cells, chondrocytes, adipocytes and osteocytes, more preferably the differentiated cells are skeletal muscle cells, One cell selected from the group consisting of cardiomyocytes and smooth muscle cells, most preferably said differentiated cells are skeletal muscle cells.
바람직하게는, 상기 분화세포는 결합 조직, 신경 세포, 림프 세포, 맥관 세포, 신장 세포, 췌장 세포, 폐 세포, 요도 세포, 방광 세포, 위 세포, 간 세포, 소장 세포, 대장 세포 및 식도 세포로 구성된 군으로부터 선택되는 하나의 세포이다.  Preferably, the differentiated cells are connective tissue, nerve cells, lymph cells, vasculature cells, kidney cells, pancreas cells, lung cells, urethral cells, bladder cells, gastric cells, liver cells, small intestine cells, colon cells and esophageal cells. One cell selected from the group consisting of:
바람직하게는, 본 발명의 단계 (a)는 포유동물의 근육세포로부터 유래된 융합체 (syncytia)를 분절시키고 단핵세포 또는 세포물 (eel lulate)을 얻기 위하여 상기 분화된 융합체에 세포화 -유도 제제 (cellularization- inducing agent)를 처리하여 실시한다. Preferably, step ( a ) of the present invention comprises a method of incorporating a cell-derived agent into the differentiated fusion to segment the syncytia derived from the muscle cells of the mammal and obtain mononuclear cells or eel lulates. This is done by treating cellularization-inducing agent.
본 발명의 바람직한 구현예에 따르면, 골격근 융합체에 세포화 -유도 제제 (cellularization-inducing agent)를 처리하여 단핵세포 또는 세포물을 제조한다.  According to a preferred embodiment of the present invention, a mononuclear cell or a cell material is prepared by treating a skeletal muscle fusion with a cellization-inducing agent.
본 명세서의 용어 "융합체 (syncytia)" 는 많은 핵을 포함하는 세포질로 가득찬 거대 세포 -유사 (large cell-like) 구조를 가지는 다핵세포를 의미하며, 진핵생물에서 대부분의 세포는 단일 핵을 가진다는 점에서 융합체는 매우 특이한 형태이다.  As used herein, the term "syncytia" refers to a multinuclear cell having a large cell-like structure filled with cytoplasm containing many nuclei, and in eukaryotes, most cells have a single nucleus. In that sense the fusion is a very unusual form.
본 발명의 세포화—유도 제제는 마이크로튜블 어셈블리를 억제할 수 있는 당업계에 알려진 어떠한 화학물질도 사용할 수 있다. 본 발명의 바람직한 구현예에 따르면, 본 발명의 세포화 -유도 제제는 미오세버린, 미오세버린 B, 콜치신, 노코다졸, 알로콜친 탁세인, 포도필로록식, 콤브레타스타틴, 스테그나신, 다이하이드록시- 펜타메톡시플라나논 (dihydroxy—pentamethoxy ananone) , 2- 메록시에스트라디올 (2-ME) 및 이의 유사체, 벤지미다졸 카바메이트, 안사미토신 (ansamitocin), TN16, 빈블라스틴, 빈크리스틴, 돌라스타틴, 쿠라신 A, 에토포사이드, 테니포사이드, 산구이나린 (sanguinarine) 및 첼리도닌 (chelidonine)올 포함하며, 보다 바람직하게는, 미오세버린, 미오세버린 B, 콜치신 및 노코다졸을 포함하고, 보다 더 바람직하게는 미오세버린 및 미오세버린 B를 포함하며, 가장 바람직하게는 미오세버린이다. The cellularization-inducing formulations of the present invention can use any chemical known in the art that can inhibit microtubule assembly. According to a preferred embodiment of the present invention, the cell-derived agent of the present invention is myosebine, myosebine B, colchicine, nocodazole, allocolchin taxane, grapephylolock, combretastatin, steg Nacin, dihydroxy-pentamethoxy ananone, 2- methoxyestradiol (2-ME) and its analogues, benzimidazole carbamate, ansamitocin, TN16, vinbla Stin, vincristine, dolastatin, curacin A, etoposide, teniposide, sanguinarine and Chelidonineol, more preferably, myosebergin, myosebergin B, colchicine and nocodazole, even more preferably myosebergin and myosebergin B; And most preferably is myosebourne.
본 발명은 세포내 역분화와 관련된 신규한 조절인자들을 동정하기 위해 퓨린-기반된 화합물 라이브러리를 이용하여 동정된 미오세버린을 이 I )는 다음과 같다:  The present invention identifies myosebers identified using a purine-based compound library to identify novel regulators associated with intracellular dedifferentiation.
Figure imgf000008_0001
Figure imgf000008_0001
Schultz 및 공동연구자들은 조합화학적 합성 방법 (Combinatorial Chemistry)으로 구축된 2,6,9-대체 퓨린 라이브러리를 이용하여 강력한 마이크로튜블 -분해 화합물인 미오세버린을 동정하였다 [14]. 미오세버린 및 이의 유사체들의 낮은 세포독성은 세포증식억제성 항종양 제제로서 적용될 수 있도록 한다. 본 발명에서 이용되는 미오세버린은 2,6,9- 대체된 퓨린 (2,6,9-trisubstituted purines) 라이브러리로부터 동정된 작은 화학분자로서 다음과 같은 화학구조 (Π)를 가진다:  Schultz and co-workers identified myoseber, a potent microtubule-degrading compound, using a 2,6,9-alternative purine library constructed by Combinatorial Chemistry [14]. The low cytotoxicity of myoseberg and its analogs allows it to be applied as a cytostatic antitumor agent. Myoserine used in the present invention is a small chemical molecule identified from the 2,6,9-trisubstituted purines library, and has the following chemical structure (Π):
Figure imgf000008_0002
Figure imgf000008_0002
미오세버린은 세포주기 조절 단백질안 사이클린-의존성 키나제 (CDK) 2의 억제제로 알려진 퓨린 유도체인 올로뮤신 (olomoucine)에 기초하며, 이의 화학구조 (ΙΠ)는 다음과 같다:  Myoserine is based on olomoucine, a purine derivative known as an inhibitor of cyclin-dependent kinase (CDK) 2 in cell cycle regulatory proteins, whose chemical structure (ΙΠ) is as follows:
6 6
대체용지 (규칙 제 26조) Alternative Site (Article 26)
Figure imgf000009_0001
미오세버린은 스핀들 어셈블리를 방해함으로써 G2/M 이동 (transit ion)에서 세포주기를 어레스트할 뿐 아니라, 완전 분화된 근육세포를 환경적 요인에 반웅하는 상태로 역전시켜 근육 재생 및 즐기세포 분화에 적용시킬 수 있다.
Figure imgf000009_0001
Myosebern not only restrains the cell cycle in the G 2 / M transit ion by disrupting spindle assembly, but also reverses fully differentiated muscle cells to environmental factors, resulting in muscle regeneration and enjoyable cell differentiation. Can be applied.
본 발명은 상술한 골격근 융합체에 세포화 -유도 제제를 처리하여 다핵성 골격근 근관의 세포화를 유도하였다. 보다 상세하게는, 근관 배양액에 10 uM 미오세버린, 15 iiM 미오세버린 B, 1 yM 콜치신, 1 노코다졸 또는 10 μΜ 탁솔을 24시간 동안 처리하여 세포화된 근관을 얻었으며, 이때 단핵세포 이외에 상대적으로 많은 양의 세포내 잔해물 (debris)을 제거하기 위해, 트립신 처리 후 원심분리하고 70 m 메쉬를 통해 여과시켰다. 그 결과, 10 μΜ 미오세버린의 처리가 가장 많은 수의 단핵세포를 생산하였다 (참고: 도 lc).  The present invention induces cellularization of the multinucleated skeletal muscle root canal by treating the skeletal muscle fusion described above with a cytolation-inducing agent. More specifically, root canal cultures were treated with 10 uM myosebine, 15 iiM myosevery B, 1 yM colchicine, 1 nocodazole or 10 μΜ Taxol for 24 hours to obtain a cellized root canal. To remove relatively large amounts of intracellular debris in addition to the cells, they were centrifuged after trypsinization and filtered through a 70 m mesh. As a result, treatment with 10 μΜ myoseberg produced the largest number of monocytes (see Figure lc).
본 발명의 바람직한 구현예에 따르면, 본 발명의 미오세버린은 5-50 μΜ, 보다 바람직하게는 5-30 μΜ, 보다 더 바람직하게는 5ᅳ20 μΜ, 가장 바람직하게는 10-15 μΜ으로 처리한다. 단계 (b): p21 발현의 하향 -조절 유도  According to a preferred embodiment of the invention, the myoseberg of the invention is treated with 5-50 μΜ, more preferably 5-30 μΜ, even more preferably 5 × 20 μΜ, most preferably 10-15 μΜ do. Step (b): Induce down-regulation of p21 expression
이어, 분화세포에 p21 하향—조절 유도 화합물을 처리한다.  The differentiated cells are then treated with p21 down-regulatory inducing compounds.
p21 은 환경적 스트레스에 따른 핵 이벤트를 조절하는 과정들에서 중요한 기능을 한다。 p21은 산화적 스트레스 및 DNA손상에 따라 매우 높은 레벨로 유도된다. p21 단백질은 사이클린-의존성 키나제의 억제제로 기능하며 세포주기 진행을 효과적으로 차단하기 때문에, 노화 또는 완전 분화된 세포들에서 과다발현 된다. 본 발명에 따르면, p21 발현의 하향- 조절은 근섬유 세포화 및 세포주기 재ᅳ진입을 유도하여 유미목 양서류의 부속기관 재생의 초기 단계를 모방하도록 유도한다. p21 plays an important role in regulating nuclear events due to environmental stress, p21 is induced at very high levels due to oxidative stress and DNA damage. The p21 protein acts as an inhibitor of cyclin-dependent kinases and effectively blocks cell cycle progression, so it is overexpressed in aging or fully differentiated cells. According to the present invention, down-p21 expression Regulation induces myofibrillarization and cell cycle reentry to mimic the early stages of adnexal regeneration of Rhesus amphibians.
본 발명의 p21 하향 -조절 유도 화합물은 p21 유전자의 발현 억제, p21 단백질의 활성 억제 또는 p21 단백질의 안정성을 감소시켜 하향-조절을 유도하는 화합물이다ᅳ  The p21 down-regulatory inducing compounds of the present invention are compounds that induce down-regulation by inhibiting the expression of the p21 gene, inhibiting the activity of the p21 protein or decreasing the stability of the p21 protein.
본 발명의 바람직한 구현예에 따르면, 본 발명의 p21 발현의 하향- 조절은 당업계에 알려진 어떠한 방법을 이용하여 실시할 수 있으며, 예를 들어 안티센스 을리고뉴클레오타이드, siRNA, miRNA, 또는 천연물 추출물을 이용하여 실시할 수 있으나, 이에 한정되는 것은 아니다.  According to a preferred embodiment of the present invention, down-regulation of p21 expression of the present invention can be carried out using any method known in the art, for example using antisense oligonucleotides, siRNA, miRNA, or natural product extracts. It may be carried out by, but is not limited thereto.
본 발명에서 용어 "siRNA" 는 RNA 방해 또는 유전자 사일런싱을 매개할 수 있는 핵산 분자를 의미한다 (참조: TO 00/44895, WO 01/36646, WO 99/32619, WO 01/29058, WO 99/07409 및 W0 00/44914) . siRNA 는 표적 유전자의 발현을 억제할 수 있기 때문에 효율적인 유전자 넉 -다운 방법으로서 또는 유전자치료 방법으로 제공된다. siRNA 는 식물, 벌레, 초파리 및 기생충에서 처음으로 발견되었으나, 최근에 siRNA 를 개발 /이용하여 포유류 세포 연구에 웅용되었다 (Degot S, et al. 2002; Degot S, et al. 2004; Ballut L, et al. 2005).  The term "siRNA" in the present invention means a nucleic acid molecule capable of mediating RNA interference or gene silencing (see TO 00/44895, WO 01/36646, WO 99/32619, WO 01/29058, WO 99 / 07409 and W0 00/44914). siRNA is provided as an efficient gene knock-down method or gene therapy method because it can inhibit the expression of the target gene. siRNA was first discovered in plants, worms, fruit flies and parasites, but has recently been used in mammalian cell research by developing / using siRNAs (Degot S, et al. 2002; Degot S, et al. 2004; Ballut L, et al. 2005).
본 발명의 siRNA 분자는, 센스 가닥 (p21 m NA 서열에 상응하는 (corresponding) 서열)과 안티센스 가닥 (p21 mRNA 서열에 상보적인 서열)이 서로 반대쪽에 위치하여 이중쇄를 이루는 구조를 가질 수 있다. 또한, 다른 구현 예에 따르면, 본 발명의 siRNA 분자는, 자기-상보성 (self- complementary) 센스 및 안티센스 가닥을 가지는 단일쇄 구조를 가질 수 있다.  The siRNA molecule of the present invention may have a structure in which a sense strand (corresponding sequence corresponding to a p21 m NA sequence) and an antisense strand (a sequence complementary to a p21 mRNA sequence) are positioned opposite to each other to form a double strand. Further, according to another embodiment, siRNA molecules of the present invention may have a single chain structure having self-complementary sense and antisense strands.
siRNA 는 RNA 끼리 짝을 이루는 이중사슬 RNA 부분이 완전히 쌍을 이루는 것에 한정되지 않고 미스매치 (대웅하는 염기가 상보적이지 않음), 벌지 (일방의 사슬에 대웅하는 염기가 없음) 등에 의하여 쌍을 이루지 않는 부분이 포함될 수 있다. 전체 길이는 10 내지 100 염기, 바람직하게는 15 내지 80 염기, 더욱 바람직하게는 20 내지 70 염기이다.  siRNAs are not limited to fully paired double-stranded RNA portions of RNA, but are paired by mismatches (bases not complementary), bulges (no bases on one chain), and the like. May be included. The total length is 10 to 100 bases, preferably 15 to 80 bases, more preferably 20 to 70 bases.
siRNA 말단 구조는 p21 유전자의 발현을 R Ai 효과에 의하여 억제할 수 있는 것이면 평활 (blunt) 말단 흑은 점착 (cohesive) 말단 모두 가능하다. 점착 말단 구조는 3'—말단 돌출 구조와 5'-말단 돌출 구조 모두 가능하다. 본 발명의 siRNA 분자는, 자기-상보성 센스 및 안티센스 가닥 사어에 짧은 뉴클레오타이드 서열 (예컨대, 약 5-15 nt)이 삽입된 형태를 가질 수 있으며, 이 경우 뉴클레오타이드 서열의 발현에 의해 형성된 siRNA 분자는 분자내 흔성화에 의하여 헤어핀 구조를 형성하게 되며, 전체적으로는 스템- 앤드 -루프 구조를 형성하게 된다. 이 스템 -앤드 -루프 구조는 인 비트로 또는 인 ^에서 프로세싱되어 RNAi 를 매개할 수 있는 활성의 siRNA 분자를 생성한다. The siRNA terminal structure is capable of both blunt terminal black and cohesive terminal as long as the expression of the p21 gene can be inhibited by the R Ai effect. Cohesive end structures are possible with both 3'-end protrusion structures and 5'-end protrusion structures. The siRNA molecules of the present invention may have a form in which a short nucleotide sequence (eg, about 5-15 nt) is inserted into a self-complementary sense and antisense strand, wherein the siRNA molecule formed by expression of the nucleotide sequence is a molecule Due to the internalization, the hairpin structure is formed, and the stem-and-loop structure as a whole is formed. This stem-and-loop structure can be processed in vitro or in ^ to produce an active siRNA molecule capable of mediating RNAi.
본 발명에서 용어 "안티센스 올리고뉴클레오타이드" 란 특정 mRNA 의 서열에 상보적인 핵산 서열을 함유하고 있는 DNA 또는 RNA 또는 이들의 유도체를 의미하고, mRNA 내의 상보적인 서열에 결합하여 mRNA 의 단백질로의 번역을 저해하는 작용을 한다. 본 발명의 안티센스 서열은 P21 에 상보적이고 p21 mRNA 에 결합할 수 있는 DNA 또는 RNA 서열을 의미하고, p21 mRNA 의 번역, 세포질 내로의 전위 (translocation) , 성숙 (maturation) 또는 다른 모든 전체적인 생물학적 기능에 대한 필수적인 활성을 저해할 수 있다. 안티센스 핵산의 길이는 6 내지 100 염기이고, 바람직하게는 8내지 60 염기이고, 보다 바람직하게는 10 내 40 염기이다. 본 명세서의 용어 "miRNA(microRNA)" 는 21-25 뉴클레오타이드의 단일 가닥 RNA 분자로써, 타겟 mRNA 의 파쇄 또는 해독단계에서의 억제를 통하여 진핵생물의 유전자 발현을 제어하는 조절물질이다. 이러한 miRNA 는 두 단계의 프로세싱으로 이루어진다. 최초의 miRNA 전사체 (primary miRNA)가 핵 안에서 Drosha 라는 RNaseEI type 효소에 의해 70-90 염기 정도의 스템 -루프 구조, 즉 pre-miRNA 로 만들어지고, 이후 세포질로 이동하여 다이서 (Dicer)라는 효소에 의해 절단되어 21-25 염기의 성숙한 miRNA 로 만들어진다. 이렇게 생성된 miRNA 는 표적 mRNA 에 상보적으로 결합하여 전사 후 유전자 억압자 (post-transcript ional gene suppressor)로써 작용하며, 번역 억제와 mRNA 불안정화를 유도한다. miRNAs는 다양한 생리학적 현상 및 질환에 관여한다. As used herein, the term "antisense oligonucleotide" refers to DNA or RNA or a derivative thereof containing a nucleic acid sequence complementary to a sequence of a particular mRNA, and binds to a complementary sequence in the mRNA to inhibit translation of the mRNA into a protein. It works. An antisense sequence of the invention refers to a DNA or RNA sequence that is complementary to P 21 and capable of binding to p21 mRNA, and is responsible for the translation of p21 mRNA, translocation into the cytoplasm, maturation or any other overall biological function. May inhibit essential activity. The antisense nucleic acid has a length of 6 to 100 bases, preferably 8 to 60 bases, and more preferably 10 to 40 bases. As used herein, the term “miRNA (microRNA)” refers to a single-stranded RNA molecule of 21-25 nucleotides, and is a modulator that controls gene expression in eukaryotes through inhibition of the disruption or translation of target mRNAs. This miRNA consists of two stages of processing. The first miRNA transcript (primary miRNA) is made into the stem-loop structure of 70-90 bases, or pre-miRNA, by the RNaseEI type enzyme called Drosha in the nucleus, and then migrates into the cytoplasm and is called Dicer. Cleaved to make up to 21-25 bases of mature miRNA. The miRNA thus produced complementarily binds to the target mRNA and acts as a post-transcript ional gene suppressor, inducing translation inhibition and mRNA destabilization. miRNAs are involved in a variety of physiological phenomena and diseases.
본 발명의 바람직한 구현예에 따르면, 본 발명에서 p21 발현의 하향- 조절은 세포화된 근관의 증식 또는 세포주기로의 재 -진입 (re-entry) 및 분화를 유도한다. 본 발명의 바람직한 구현예에 따르면, 본 발명의 세포화된 근관은 p21 발현의 하향 -조절 후에도 고유한 근전위 (myogenic potential)를 유지한다. According to a preferred embodiment of the present invention, down-regulation of p21 expression in the present invention induces the proliferation of cellized root canal or re-entry and differentiation into the cell cycle. According to a preferred embodiment of the present invention, the cellized root canal of the present invention maintains its inherent myogenic potential even after down-regulation of p21 expression.
본 발명의 일 양태에 따르면, 본 발명의 방법에 의해 수득되는 다능성 세포는 단계 (a)에서 이용되는 분화세포와 동일한 리니지 또는 배엽의 세포이다. 예컨대, 분화세포로서 골격근 세포를 이용하는 경우, 본 발명에 의해 수득되는 다능성 세포는 골격근 동일한 배엽의 세포, 예컨대 골격근, 뼈, 진피, 결합 조직, 비뇨생식계, 심장, 피 (림프 세포), 신장 및 비장을 구성하는 세포로 분화될 수 있는 다능성 세포이다.  According to one aspect of the invention, the pluripotent cells obtained by the method of the invention are cells of the same lineage or germ layer as the differentiated cells used in step (a). For example, in the case of using skeletal muscle cells as differentiated cells, the pluripotent cells obtained by the present invention are cells of the same germ cells of skeletal muscle, such as skeletal muscle, bone, dermis, connective tissue, urogenital system, heart, blood (lymph cells), kidney and It is a pluripotent cell that can differentiate into the cells that make up the spleen.
본 발명의 바람직한 구현예에 따르면, 단계 (a)에서 이용되는 분화세포는 골격근 세포이고, 단계 (b)에서 수득한 다능성 세포는 골격근 세포, 지방세포 또는 골세포로 분화될 수 있는 다능성 세포이다.  According to a preferred embodiment of the present invention, the differentiated cells used in step (a) are skeletal muscle cells, and the pluripotent cells obtained in step (b) can be differentiated into skeletal muscle cells, adipocytes or osteocytes. to be.
본 발명의 제조방법에 따라, p21 하향-조절된 분화세포에 역분화제를 처리한다.  According to the preparation method of the present invention, p21 down-regulated differentiated cells are treated with a reverse differentiation agent.
본 발명의 용어 "역분화제 (dedifferentiation agent)" 는 하나 이상의 세포형태들의 역분화를 촉진하는 제제를 의미하며, 화학물질, 핵산, 펩타이드, 폴리펩타이드, 작은 유기분자, 항체 안티센스 올리고뉴클레오타이드, RNAi 컨스트럭트 및 리보자임을 포함한다. 다양한 역분화제는 완전 분화된 포유동물 세포에서 역분화를 유도하기에 층분한 시간 동안 하나 이상의 역분화 인자들과 인 비보 또는 인 트 ^에서 세포와 접촉시킴으로써 역분화를 유도할 수 있다.  As used herein, the term “dedifferentiation agent” refers to an agent that promotes the dedifferentiation of one or more cell types, and includes chemicals, nucleic acids, peptides, polypeptides, small organic molecules, antibody antisense oligonucleotides, RNAi constructs. And ribozymes. Various retrodifferentiating agents can induce retrodifferentiation by contacting one or more retrodifferentiating factors with cells in vivo or int ^ for a time sufficient to induce retrodifferentiation in fully differentiated mammalian cells.
본 발명의 바람직한 구현예에 따르면, 본 발명에서 이용될 수 있는 역분화제는 리버신 (N6-사이클로핵실 -N2- -모폴린 -4-일-페닐 )-9H-퓨린 -2 6- 다이아민), 트리코스타틴 A(trichostatin A), 발프론산 (valproic acid), 부페닐 (buphenyl) 및 5-아자시티딘 (5_azacyt idine)을 포함하지만, 이에 한정되는 것은 아니다. 가장 바람직하게는, 리버신이다.  According to a preferred embodiment of the present invention, the reverse differentiation agent which can be used in the present invention is reversin (N6-cyclonucleus-N2- -morpholin-4-yl-phenyl) -9H-purine-2 6-diamine) Trichostatin A, valproic acid, buphenyl, and 5-azacytidine (5_azacyt idine), but are not limited thereto. Most preferably, it is reversin.
리버신은 2,6-대체된 퓨린 화합물로 하기의 화학구조 (IV)를 가지며, 근육형성 세포군 (myogenic)을 증식 및 재-분화가 가능한 다능성 중간엽전구세포 (예컨대, 골 및 지방으로의 증식 및 재-분화)로 유도하는 놀라운 특성을 가진다: Reversin is a 2,6-replaced purine compound with the following chemical structure (IV) and capable of proliferating and re-differentiating myogenic cells (eg, proliferation into bone and fat). And re-differentiation).
Figure imgf000013_0001
Figure imgf000013_0001
상술한 바와 같이, 본 발명의 화학물질-기반된 접근방법 (chemically defined approaches)에 따라 얻어진 본 발명의 다능성 세포를 화학적으로 잘 정립된 배양 조건에서 배양시킴으로써, 골격근 융합체로부터 근육형성세포로의 분화가 가능할 뿐 아니라 연골형성세포 및 지방형성 세포로의 이형분화도 가능하였다 (참고: 도 3 내지 도 6).  As described above, differentiation from skeletal muscle fusion to myogenic cells by culturing the pluripotent cells of the invention obtained according to the chemically defined approaches of the present invention in chemically well-established culture conditions. Not only was possible but also heterologous differentiation into chondrogenic and adipogenic cells (see Figures 3 to 6).
본 발명의 바람직한 구현예에 따르면, 본 발명의 다능성 세포는 근육형성 세포군 (myogenic lineage), 지방형성 세포군 (adipogenic lineage) 또는 골형성 세포군 (osteogenic lineage)으로 분화할 수 있다.  According to a preferred embodiment of the present invention, the pluripotent cells of the present invention can differentiate into myogenic lineage, adipogenic lineage or osteogenic lineage.
야마나카 및 공동연구자들은 2006 년에 배아줄기세포와 유사한 원시세포를 생성하여, 이를 유도만능줄기세포 (iPSCs)로 명명하였다 (Kazutoshi Takahashi and Shiny a Yamanaka, Cell, 126: 663- 676(2006)). 상기 연구는 배아 파괴나 난자 제공과 같은 윤리적 문제가 없는 환자별 /질환별 맞춤식 역분화 만능 즐기세포를 만들어낼 수 있는 기반을 제공하였지만, 확립된 역분화 만능줄기세포의 안정성 및 분화능이 현저히 떨어지고 있는 실정이다. 상기 문제점들을 극복하기 위해, 작은 분자 (예컨대, 본 발명의 미오세버린)를 이용하여 역분화 만능 줄기세포의 안정화 및 분화능을 증가시킬 수 있는 방법의 개발이 시급하다. 따라서 , 본 발명의 화학물질-기반된 접근방법은 포유동물에서 사지 재생을 유도하는 전략들의 개발에 중요하며, 완전 분화된 불웅성 조직으로부터 유도만능줄기세포 (iPSCs)의 개발을 위한 새로운 접근방법을 제시한다.  Yamanaka and co-workers produced embryonic stem cells similar to embryonic stem cells in 2006 and named them iPSCs (Kazutoshi Takahashi and Shiny a Yamanaka, Cell, 126: 663-676 (2006)). The study provided the basis for producing patient- and disease-specific customized pluripotent pluripotent cells without ethical problems such as embryo destruction or egg donation, but the stability and differentiation ability of established pluripotent stem cells were significantly reduced. It is true. In order to overcome the above problems, it is urgent to develop a method that can increase the stabilization and differentiation capacity of pluripotent stem cells using small molecules (eg, myose verbine of the present invention). Thus, the chemical-based approach of the present invention is important for the development of strategies for inducing limb regeneration in mammals, and a novel approach for the development of iPSCs from fully differentiated afferent tissues. present.
바람직하게는, 상기 방법은 단계 (b)의 역분화제 처리이후 분화 유도제를 단계 (b)에서 얻은 세포에 처리하여 단계 (a)의 분화세포와 동일 3 배엽 계통의 다른 세포 리니지 (cell lineage)로 분화시키는 단계를 추가적으로 포함한다。 상기 3 배엽은 내배엽 중배엽 및 외배엽으로 구성되며, 내배엽에서는 위, 결장, 간 췌장, 방광, 요도의 안쪽, 기도의 상피부분, 폐, 인두, 갑상선, 부갑상선 및 소장 조직을 구성하는 세포가 발생하고, 중배엽에서는 골격근, 뼈, 진피, 결합 조직, 비뇨생식계, 심장, 피 (림프 세포), 신장 및 비장을 구성하는 세포가 발생하며, 외배엽에서는 중주신경계, 눈의 수정체, 두개골 및 감각, 신경절 및 신경, 색소 세포, 머리결합조직 (head connective tissues), 표피, 털 및 유선을 구성하는 세포가 발생한다. Preferably, the method is followed by treatment of the differentiation inducing agent to the cells obtained in step (b) after treatment with the dedifferentiation agent of step (b) to another cell lineage of the same three germ lineage as the differentiated cells of step (a). And further comprising the step of differentiating. The three germ layers are composed of mesoderm mesoderm and ectoderm, endothelial cells in the stomach, colon, liver pancreas, bladder, the inside of the urethra, the epithelial part of the airway, lung, pharynx, thyroid gland, parathyroid gland and small intestine tissues, The mesoderm develops the cells that make up skeletal muscle, bone, dermis, connective tissue, urogenital system, heart, blood (lymph cells), kidneys and spleen. Pigment cells, head connective tissues, epidermis, hair and cells that make up the mammary gland develop.
본 명세서에서, 용어 '분화 유도제' 는 원하는 세포로 분화시키기 위해 배양배지에 포함시키는 성장인자 및 호르몬 등을 말한다.  As used herein, the term 'differentiation inducing agent' refers to growth factors, hormones, and the like included in the culture medium to differentiate into the desired cells.
바람직한 구현예에 따르면, 단계 (b)에서 수득한 다능성 즐기세포로부터 지방형성 세포 리니지를 수득하기 위하여 이용되는 분화 유도제는 인슬린, 텍사메타손, 로시글리타존, According to a preferred embodiment, the differentiation inducing agent used to obtain the lipogenic cell lineage from the pluripotent pleasant cells obtained in step (b) is inslin, texamethasone, rosiglitazone,
IBMX(isobutylmethylxanthine) 또는 이의 흔합물이다. Isobutylmethylxanthine (IBMX) or a combination thereof.
바람직한 구현예에 따르면, 단계 (b)에서 수득한 다능성 줄기세포로부터 골형성 세포 리니지를 수득하기 위하여 이용되는 분화 유도제는 아스코르브산 -2-포스페이트, 덱사메타손, β—글리세로포스페이트 또는 이의 흔합물이다.  According to a preferred embodiment, the differentiation inducer used to obtain osteogenic cell lineage from the pluripotent stem cells obtained in step (b) is ascorbic acid-2-phosphate, dexamethasone, β-glycerophosphate or a combination thereof. .
본 발명의 바람직한 구현예에 따르면, 본 발명의 다능성 세포는 근육형성 세포 리니지 (myogenic lineage), 지방형성 세포 리니지 (adipogenic lineage) 또는 골형성 세포 리니지 (osteogenic lineage)로 분화할 수 있다. 본 발명의 다른 양태에 따르면, 본 발명은 상술한 방법에 따라 제조된 다능성 세포를 제공한다.  According to a preferred embodiment of the present invention, the pluripotent cells of the present invention can differentiate into myogenic lineage, adipogenic lineage or osteogenic lineage. According to another aspect of the invention, the invention provides a pluripotent cell prepared according to the method described above.
본 발명의 방법에 따라 제조된 다능성 세포로부터 분화된 근육형성 세포 리니지 또는 근육세포를 유효성분으로 포함하는 근질환 (myopathy) 치료용 조성물을 제공할 수 있다.  It is possible to provide a composition for treating myopathy comprising muscle forming cell lineage or muscle cells differentiated from pluripotent cells prepared according to the method of the present invention as an active ingredient.
본 명세서에서, 용어 '근질환' 은 근육 피로, 근위축증, 사고 또는 수술에 의한 상처 또는 근이영양증이고, 용어 '근이영양증' 은 베커형 근이영양증 (Becker 's muscul r dystrophy) , 선천성 근이영양증 (congenital muscular dystrophy) , 뒤셴형근이영양증 (Duc'henne muscular dystrophy) , 원위 근이영양증 (distal muscular dystrophy), 에머리 -드라이푸스 근이영양증 (Emery-Dreifuss muscular dystrophy), 안면견갑상완 근이영양증 지대형 근이영양증, 근긴장성 근이영양증, 안인두성 근이영양증, 근긴장성 근이영양증, 사르코글리칸증, 척수성 근육위축 또는 B L (Brown-Vial etto- Van Laere syndrome)을 포함하지만, 이에 한정되는 것은 아니다. In the present specification, the term 'muscle disease' refers to muscle fatigue, muscular dystrophy, accidental or surgical wounds or muscular dystrophy, and the term 'muscular dystrophy' refers to Becker's muscul r dystrophy, congenital muscular dystrophy (congenital) muscular dystrophy), Duc ' henne muscular dystrophy, distal muscular dystrophy, Emery-Dreifuss muscular dystrophy, facial shoulder dystrophic muscular dystrophy, muscular dystrophy, muscular dystrophy Muscular dystrophy, myotonic muscular dystrophy, sarcoglycanism, spinal muscular atrophy or Brown-Vial etto-Van Laere syndrome (BL).
본 발명의 방법에 따라 제조된 다능성 세포로부터 분화된 골형성 세포 리니지을 유효성분으로 포함하는 골 질환 치료용 조성물을 제공할 수 있다.  It is possible to provide a composition for treating bone diseases comprising osteogenic cell lineage differentiated from pluripotent cells prepared according to the method of the present invention as an active ingredient.
본 명세서에서, 용어 '골 질환' 은 골다공증, 청소년 골다공증, 불완전 골형성증 (osteogenesis imperfecta), 과골화증, 고칼슘혈증, 부갑상선 기능항진증, 골연화증, 용해성 골질환, 골괴사증, 뼈의 파젯병, 골 발생 질환, 골 골절, 류마티스 관절염에 의한 골 손실, 염증성 류마티스 관절염, 골수염, 전이성 골질환, 치주성 골 소실, 구루병, 암에 의한 골 손실 또는 골의 노인성 손실을 포함하지만, 이에 한정되는 것은 아니다. 본 발명의 조성물은 (a) 상술한 다능성 또는 전능성 세포로부터 분화된 세포 (예컨대, 근육형성 세포군, 골형성 세포군)의 약제학적 유효량; 및 (b) 약제학적으로 허용되는 담체를 포함하는 약제학적 조성물이다. As used herein, the term 'bone disease' refers to osteoporosis, juvenile osteoporosis, osteogenic imperfecta, hyperosclerosis, hypercalcemia, hyperparathyroidism, osteomalacia, soluble bone disease, osteonecrosis, Paget's disease of bone, bone development Diseases, bone fractures, bone loss due to rheumatoid arthritis, inflammatory rheumatoid arthritis, osteomyelitis, metastatic bone disease, periodontal bone loss, rickets, bone loss by cancer or senile loss of bone. The composition of the present invention comprises ( a ) a pharmaceutically effective amount of cells differentiated from the pluripotent or pluripotent cells described above (eg, myogenic cell groups, osteogenic cell groups); And (b) a pharmaceutically acceptable carrier.
본 명세서에서 용어 "약제학적 유효량" 은 상술한 다능성 세포로부터 분화된 세포의 효능 또는 활성을 달성하는 데 충분한 양을 의미한다.  The term "pharmaceutically effective amount" as used herein means an amount sufficient to achieve the efficacy or activity of cells differentiated from the pluripotent cells described above.
본 발명의 조성물이 약제학적 조성물로 제조되는 경우, 본 발명의 약제학적 조성물은 약제학적으로 허용되는 담체를 포함한다. 본 발명의 약제학적 조성물에 포함되는 약제학적으로 허용되는 담체는 제제시에 통상적으로 이용되는 것으로서, 락토스, 덱스트로스, 수크로스, 솔비를, 만니를, 전분, 아카시아 고무, 인산 칼슴, 알기네이트, 젤라틴, 규산 칼슘, 미세결정성 셀를로스, 폴리비닐피를리돈, 셀롤로스, 물, 시럽, 메탈 셀를로스, 메틸히드록시벤조에이트, 프로필히드록시벤조에이트, 활석, 스테아르산 마그네슘 및 미네랄 오일 등을 포함하나, 이에 한정되는 것은 아니다. 본 발명의 약제학적 조성물은 상기 성분들 이외에 윤활제, 습윤제, 감미제, 향미제, 유화제, 현탁제, 보존제 등을 추가로 포함할 수 있다. 적합한 약제학적으로 허용되는 담체 및 제제는 Remington's Pharmaceutical Sciences (19th ed. , 1995)에 상세히 기재되어 있다. When the composition of the present invention is made into a pharmaceutical composition, the pharmaceutical composition of the present invention includes a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers included in the pharmaceutical composition of the present invention are those commonly used in the preparation, lactose, dextrose, sucrose, sorbbi, manny, starch, acacia rubber, phosphate chame, alginate, Gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyridone, cellulose, water, syrup, metal cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate and mineral oil Including, but not limited to. The pharmaceutical composition of the present invention may further include lubricants, wetting agents, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, etc. in addition to the above components. have. Suitable pharmaceutically acceptable carriers and formulations are described in detail in Remington's Pharmaceutical Sciences (19th ed., 1995).
본 발명의 약제학적 조성물은 비경구 투여할 수 있으며, 예를 들어 피하 주입, 근육 주입, 경피 투여, 관절강내 주사, 등으로 투여할 수 있다. 5 본 발명의 약제학적 조성물의 적합한 투여량은 제제화 방법, 투여 방식, 환자의 연령, 체중, 성, 병적 상태ᅳ 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감웅성과 같은 요인들에 의해 다양하게 처방될 수 있다. 본 발명의 약제학적 조성물의 일반적인 투여량은 성인 기준으로 1 일 당 102-1010 세포이다. The pharmaceutical compositions of the present invention may be administered parenterally, for example, by subcutaneous injection, intramuscular injection, transdermal administration, intraarticular injection, and the like. 5 Appropriate dosages of the pharmaceutical compositions of the present invention will vary depending on factors such as formulation method, mode of administration, age, weight, sex, morbidity of food, time of administration, route of administration, rate of excretion and response sensitivity. Can be prescribed. Typical dosages of the pharmaceutical compositions of the invention are 10 2 -10 10 cells per day on an adult basis.
10 본 발명의 약제학적 조성물은 당해 발명이 속하는 기술분야에서 10 The pharmaceutical composition of the present invention is known in the art
——통상의 지식을 가진 -자가 용이하게ᅳ실시할ᅳ수 았는 방법에- 따라, 약제학적으로 허용되는 담체 및 /또는 부형제를 이용하여 제제화함으로써 단위 용량 형태로 제조되거나 또는 다용량 용기 내에 내입시켜 제조될 수 있다. 이때 제형은 오일 또는 수성 매질중의 용액, 현탁액, 시럽제 또는 15 유화액 형태이거나 액스제, 산제, 분말제, 과립제, 정제 또는 캅샐제 형태일 수도 있으며, 분산제 또는 안정화제를 추가적으로 포함할 수 있다. 본 발명의 또 다른 양태에 따르면, 본 발명은 본 발명은 다음의 단계를 포함하는 포유동물 조직 또는 기관의 재생 (regeneration)방법을 20 제공한다: — Manufactured in unit dose form or incorporated into a multi-dose container by formulating with pharmaceutically acceptable carriers and / or excipients—according to how conventionally readily available ones can be practiced. Can be prepared. The formulation may be in the form of solutions, suspensions, syrups or emulsions in oil or aqueous media, or may be in the form of axes, powders, powders, granules, tablets or capsules, and may further comprise dispersants or stabilizers. According to another aspect of the invention, the invention provides a method of regeneration 20 of a mammalian tissue or organ comprising the following steps:
(a) 포유동물부터 분화세포를 수득하는 단계;  (a) obtaining differentiated cells from a mammal;
(b) 상기 분화세포에 p21 하향 -조절 유도 후 역분화제 (dedifferentiation agent)를 처리하여 다능성 세포를 수득하는 단계; 및  (b) treating the differentiated cells with p21 down-regulation followed by dedifferentiation agent to obtain pluripotent cells; And
25 (C) 상기 다능성 세포를 원하는 조직 또는 기관으로 분화시키는 단계.  25 (C) differentiating the pluripotent cells into the desired tissue or organ.
본 발명의 방법은 상기 제조방법을 이용하기 때문에, 이 둘 사이에 공통된 내용은 본 명세서의 과도한 복잡성을 피하기 위하여, 그 기재를 생략한다,  Since the method of the present invention uses the above manufacturing method, the common content between the two is omitted in order to avoid excessive complexity of the present specification,
30 단계 (c): 다능성 세포를 원하는 조직 또는 기관으로 분화 이어, 단계 (b)에서 수득한 다능성 세포를 원하는 조직 또는 기관으로 분화시킨다. · Step 30 (c): Differentiate pluripotent cells into desired tissues or organs The pluripotent cells obtained in step (b) are then differentiated into the desired tissues or organs. ·
상기 분화는 단계 (b)에서 수득한 다능성 세포의 배양배지에 원하는 조직 또는 기관의 분화 유도제를 첨가하여 실시한다.  The differentiation is performed by adding a differentiation inducing agent of a desired tissue or organ to the culture medium of the pluripotent cells obtained in step (b).
【발명의 효과】 【Effects of the Invention】
본 발명의 특징 및 이점을 요약하면 다음과 같다:  The features and advantages of the present invention are summarized as follows:
(a) 본 발명은 근육-유래된 다능성 세포의 화학적 제조방법 및 이의 용도에 관한 것이다.  (a) The present invention relates to a method for chemical preparation of muscle-derived pluripotent cells and uses thereof.
(b) 본 발명의 방법은 연속적인 작은 분자의 처리 및 일시적인 p21 억제 (suppress ion)를 통해 분화가 완료된 포유동물 골격근으로부터 증식하는 단핵세포 또는 세포물을 얻은 후 이에 리버신의 단계적 처리에 의해 다능성 세포를 수득하는 매우 간단하고 효율적인 방법이다.  (b) The method of the present invention obtains pluripotent mononuclear cells or cells proliferating from differentiated mammalian skeletal muscle through continuous treatment of small molecules and transient p21 suppression, followed by stepwise treatment of reversin. It is a very simple and efficient way of obtaining cells.
(c) 본 발명의 다능성 세포는 근육형성 세포군으로의 역분화 뿐 아니라, 지방형성 세포군 또는 골형성 세포군으로의 이형분화도 가능하다 .  (c) The pluripotent cells of the present invention are capable of heterodifferentiation into adipogenic or bone forming cell groups as well as dedifferentiation into muscle forming cell groups.
(d) 따라서, 본 발명의 화학물질-기반된 접근방법은 포유동물에서 사지 재생을 유도하는 전략들의 개발에 중요하며, 완전 분화된 불웅성 조직으로부터 유도만능즐기세포 (iPSCs)의 개발을 위한 새로운 접근방법을 제시한다.  (d) Therefore, the chemical-based approach of the present invention is important for the development of strategies for inducing limb regeneration in mammals, and is novel for the development of iPSCs from fully differentiated afferent tissue. Present your approach.
【도면의 간단한 설명】 [Brief Description of Drawings]
도 1 은 C2C12 근육모세포에 대한 AraC, 5-FU 및 미오세버린의 효과를 나타내는 결과이다ᅳ 도 la 는 분화하는 C2C12 근육모세포 배양에 50 μΜ AraC 를 72 시간 동안 처리하면 분화하는 배양액의 세포융합지수 (fusion index)를 증가시킨다 (즉ᅳ 다핵 근관 융합체로 삽입된 세포 핵의 수)는 것을 보여주는 결과이다. 분화하는 C2C12 근육모세포 배양에 다른 항-유사분열 약물인 5-FI 10 μΜ)를 72 시간 동안 처리하면 세포융합지수의 증가를 야기하지 않았다. 오차막대 = 표준편차; *, 비처리된 분화된 C2C12 배양과 비교하여 Ρ = < 0.05. 데이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 도 lb 는 분화하는 C2C12 근육모세포 배양에 50 μΜ AraC 를 72 시간 동안 처리하면 분화하는 배양액의 트위치 카운트 (twitch count)를 증가시킨다 (즉, 1초 당 최소 하나 이상의 비율로 자발적인 수축을 일으키는 근관)는 것을 보여주는 결과이다. 분화하는 C2C12 근육모세포 배양에 10 μΜ 5-FU 를 72 시간 동안 처리하면 트위치 카운트의 증가를 야기하지 않았다. 오차막대 = 표준편차; *, 비처리된 분화된 C2C12 배양과 비교하여 = < 0.05. 데이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 도 lc 는 10 μΜ 미오세버린, 15 μΜ 미오세버린 Β, 1 yM 콜치신, 1 μΜ 노코다졸 또는 10 μΜ 탁솔을 24 시간 동안 처리함으로써, 근관 배양이 세포화를 일으킨다는 것을 보여주는 결과이다. 살아있는 세포는 트립판 블루 다이로 염색되지 않은 세포로 분류하였다. 미오세버린은 가장 적은 세포독성을 가지며 세포화를 유도하는 것으로 확인되었다. - 오차막대ᅩ표준편차; *, 콜치신 처리구와 비교하여 Ρ = < 0.05; **, 노코다졸ᅳ 콜치신 및 미오세버린 Β 처리구와 비교하여 Ρ = < 0.05; #, 미오세버린 처리구와 비교하여 Ρ = < 0.05. 데이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 도 2 는 세포화된 근관에서 ρ21 하향-조절의 효과를 보여주는 결과이다. 도 2a 는 p21 하향-조절에 따른 세포화된 근관의 증식 반웅을 관찰하기 위한 실험적 프로토콜의 개략적인 도식을 보여준다. 도 2b 는 48 시간 동안의 p21 하향-조절이 세포화된, 불웅성 근섬유에서 증식을 유도할 수 있다는 것을 보여주는 PKpropidium iodide) 염색에 대한 유세포 분석 결과이다. 즉, 세포주기의 G2/M 기로의 쉬프트를 관찰할 수 있었다. 도 2c 는 48 시간 동안의 p21 하향-조절이 세포화된 C2C12 근관의 단기간 세포 분열을 유도한다는 것을 보여주는 그래프이다. 데이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 도 2d 는 세포화된 C2C12 근관에서 BrdU 삽입에 대한 유세포 분석을 통해 , 48 시간 동안의 p21 하향ᅳ조절이 DNA 합성을 유도한다는 것을 보여주는 결과이다. 도 2e 는 p21 하향 -조절 또는 리버신의 처리는 세포화된 C2C12 근관에서 역분화를 유도한다는 것을 보여주는 결과로, 아세틸콜린 수용체에 대한 리간드인 FTTC-α-붕가로록신의 감소된 표지 (형광)으로서 검출하였다. 도 2f 는 세포화된 C2C12 근관에서 FITC-a-붕가로특신의 시그널을 정량한 마이크로플레이트 판독기 분석 결과이다. p21 하향 -조절 또는 리버신의 처리는 세포화된 C2C12 근관에서 역분화를 유도하였다. 오차막대 = 표준편차; *, 근육모세포와 비교하여 P = < 0.05; **, 대조군 siRNA 가 처리된 C2C12 세포물과 비교하여 7° = < 0.05. 테이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 1 is a result showing the effects of AraC, 5-FU, and myoserine on C2C12 myoblasts. FIG. La is a cell fusion index of the culture medium that differentiates after 50 μM AraC for 72 hours in differentiating C2C12 myoblast culture. increase the fusion index (ie, the number of cell nuclei inserted into the multinucleated root canal fusion). Treatment of different C2C12 myoblast cultures with another anti-mitotic drug 5-FI 10 μΜ) for 72 hours did not cause an increase in the cell fusion index. Error bars = standard deviation; *, Ρ = <0.05 compared to untreated differentiated C2C12 culture. The data is representative of three independent experiments. FIG. Lb shows the differentiation of 50 μΜ AraC for 72 hours in differentiating C2C12 myoblast cultures. This results in increasing the twitch count of the culture (ie, root canal causing spontaneous contraction at least one rate per second). Treatment of 10 μΜ 5-FU in differentiated C2C12 myoblast cultures for 72 hours did not cause an increase in twitch counts. Error bars = standard deviation; *, Compared to untreated differentiated C2C12 culture = <0.05. The data is representative of three independent experiments. FIG. Lc is the result showing that root canal culture causes cellularization by treatment of 10 μM myosebergin, 15 μM myosebergin B, 1 yM colchicine, 1 μM nocodazole or 10 μM taxol for 24 hours. Living cells were classified as cells not stained with trypan blue die. Myosevery has been found to have the least cytotoxicity and induce cellularization. -Error bars 편 standard deviation; *, Ρ = <0.05 compared with colchicine treatment **, Δ = <0.05;#, compared with myocerine treated β and Ρ = <0.05 compared with treatment with nocazoleazole. The data is representative of three independent experiments. 2 shows the effect of ρ21 down-regulation in the cellized root canal. 2A shows a schematic of an experimental protocol for observing the proliferative response of cellized root canals following p21 down-regulation. FIG. 2B is a flow cytometric analysis of PKpropidium iodide staining showing that p21 down-regulation for 48 hours can induce proliferation in cellularized, male muscle fibers. That is, the shift to the G 2 / M phase of the cell cycle was observed. 2C is a graph showing that p21 down-regulation for 48 hours induces short term cell division of the cellized C2C12 root canal. The data is representative of three independent experiments. FIG. 2D shows the flow cytometry for BrdU insertion in the cellized C2C12 root canal, indicating that p21 downregulation for 48 hours induces DNA synthesis. FIG. 2E shows that treatment of p21 down-regulation or reversin induces differentiation in cellized C2C12 root canal, as a reduced label (fluorescence) of FTTC-α-Burorogroxine, a ligand for acetylcholine receptor Detected. FIG. 2F shows the results of a microplate reader assay that quantifies the signal of FITC-a-Bungaro in cellized C2C12 root canals. Treatment with p21 down-regulation or reversin induced reverse differentiation in the cellized C2C12 root canal. Error bar = Standard Deviation; *, P = <0.05 compared to myoblasts; **, 7 ° = <0.05 compared to C2C12 cells treated with control siRNA. Data is representative of three independent experiments.
도 3 은 p21 하향-조절이 리버신 처리에 의해 감소된 근전위를 역전시킨다는 것을 보여주는 결과이다. 도 3a 는 리버신 처리가 C2C12 근육모세포 및 C2C12 세포물의 근전위를 감소시킨다는 것을 보여주는 결과로, 분화 배지에서 10 일 동안 배양한 후 세포융합지수에서 감소가 나타났다. 오차막대 = 표준편차; *, 비처리된 근육모세포와 비교하여 P 0.05; **, 미오세버린 단독으로 처리된 C2C12 세포물과 비교하여 P = < 0.05. 도 3b 는 C2C12 세포물에서 p21 넉一다운이 리버신의 억제 효과를 역전시킨다는 것을 보여주는 그래프이다. 오차막대 = 표준편차; *, 미오세버린 단독으로 처리된 C2C12 세포물과 비교하여 P 0.05; **, 미오세버린 및 리버신이 처리된 C2C12 세포물과 비교하여 P 0.05. 데이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 도 3c 는 50 nM 리버신 또는 50 nM 리버신 및 80 pmol p21 siRNA 로 처리된 세포화된 근관으로부터 유래된 C2C12 배양에서 근육형성을 관찰한 광학 현미경 분석 결과이다. 헤마특실린 염색은 p21 siRNA 로 처리된 C2C12 세포물로부터 유래된 배양물에서 보다 많은 핵이 근관 융합체 내에 위치한다는 것을 보여준다. 예시적인 핵들이 근관 세포질의 연속관 (continuous tract) 내에 화살표로 표시되어 있다.  3 is a result showing that p21 down-regulation reverses the EMG decreased by reversin treatment. FIG. 3A shows that reversin treatment reduces the ETP of C2C12 myoblasts and C2C12 cells, resulting in a decrease in cell fusion index after incubation for 10 days in differentiation medium. Error bars = standard deviation; *, P 0.05 compared to untreated myoblasts; **, P = <0.05 compared to C2C12 cells treated with myoserine alone. 3B is a graph showing that p21 knockdown reverses the inhibitory effect of reversin in C2C12 cell water. Error bars = standard deviation; *, P 0.05 compared to C2C12 cells treated with myoserine alone; **, P 0.05 compared to C2C12 cells treated with myoserine and reversin. The data is representative of three independent experiments. 3C is an optical microscopic analysis of muscle formation in C2C12 cultures derived from cellized root canals treated with 50 nM reversin or 50 nM reversin and 80 pmol p21 siRNA. Hemafocillin staining shows that more nuclei are located in the root canal fusions in cultures derived from C2C12 cells treated with p21 siRNA. Exemplary nuclei are indicated by arrows in the continuous tract of the root canal cytoplasm.
도 4 는 p21 하향-조절이 세포화된 근관에서 지방세포로의 전환을 촉진시킨다는 것을 보여주는 결과이다. 도 4a 는 p21 넉 -다운 및 리버신으로 처리되고 지방형성 배지에서 배양된 C2C12 세포물은 지질을 축적할 수 있다. 이전에 기재된 바와 같이, 리버신으로 처리되고 지방형성 배지에서 배양된 C2C12 근육모세포도 지질을 축적하였다 [15]. 하지만, 리버신으로 처리되고 지방형성 배지에서 배양된 C2C12 세포물은 지질을 축적하지 않았다. 도 4b 는 p21 넉-다운, 리버신 및 지방형성 칵테일로 처리된 C2C12 세포물에서의 지질 축적을 관찰한 현미경 분석 결과로, 리버신 및 지방형성 칵테일로 처리된 C2C12 근육모세포에서 세포질 염색과 유사한 패턴을 보였다. 도 4c는 p21 넉—다운, 리버신 및 지방형성 칵테일로 처리된 C2C12 세포물이 인술린 자극 (지방세포-특이적 기능) 후 형광 글루코오스 유사체인 6-NBDG의 증가된 섭취를 보인다는 것을 보여주는 결과이다. p21 siRNA 의 대조군 siRNA 로의 대체는 인슐린-자극된 글루코오스의 섭취가 가능한 세포를 생산하는 데 실패했다. 리버신 (500 nM) 및 지방형성 칵테일로 처리된 C2C12 근육모세포도 인슐린 자극 후 6- NBDG 의 증가된 섭취를 보였다. 6-NBDG 섭취량은 인슐린 자극 후 3T3-L1 지방세포에서 관찰된 양보다 적었다. 오차막대 = 표준편차; *, 6-NBDG 단독으로 처리된 군과 비교하여 尸 = < 0.05. 데이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 도 4d 는 인슐린 자극 후 6- NBDG 의 섭취를 형광 현미경으로 시각화한 결과이다. 6-NBDG 는 C2C12 근육모세포 및 세포물-유래된 지방세포의 막에 축적하는 것으로 확인되었다. 도 4e 는 p21 넉-다운 리버신 (500 nM) 및 지방형성 칵테일로 처리된 C2C12 세포물이 에피네프린 -자극 (지방세포—특이적 기능) 후 인슐린-민감성 유리 지방산 방출을 나타냈다. p21 siRNA 의 대조군 siRNA 로의 대체는 상기 지방세포-특이적 기능을 가지는 세포를 생산하는 데 실패했다. 리버신 (500 nM) 및 지방형성 칵테일로 처리된 C2C12 근육모세포도 에피네프린-자극후 인슐린-민감성 유리 지방산 방출을 나타냈다. 유리 지방산의 방출량 및 인슐린 민감성은 3Τ3-ίΓ지방세포에서 관찰된 것보다 낮았다. 오차막대 = 표준편차; *, 에피네프린 단독으로 처리된 군과 비교하여 P = < 0.05. 데이터는 세 개의 서로 독립적인 실험들에 대한 대표적인 결과이다. 4 shows that p21 down-regulation promotes the conversion of cellularized root canal to adipocytes. FIG. 4A shows C2C12 cell water treated with p21 knock-down and reversin and cultured in adipose media can accumulate lipids. As previously described, C2C12 myoblasts treated with reversin and cultured in lipogenic medium also accumulated lipids [15]. However, C2C12 cell water treated with reversin and cultured in lipogenic medium did not accumulate lipid. FIG. 4B is a microscopic analysis of lipid accumulation in C2C12 cells treated with p21 knock-down, reversin and lipogenic cocktails, similar to cytoplasmic staining in C2C12 myoblasts treated with reversin and lipogenic cocktails. Showed. FIG. 4C shows C2C12 cells treated with p21 knock-down, reversin and lipogenesis cocktails after insulin stimulation (adipocyte-specific function). The results show an increased uptake of 6-NBDG, a fluorescent glucose analogue. Replacement of p21 siRNA with control siRNA failed to produce cells capable of ingesting insulin-stimulated glucose. C2C12 myoblasts treated with reversin (500 nM) and lipogenic cocktails also showed increased uptake of 6-NBDG after insulin stimulation. 6-NBDG intake was less than that observed in 3T3-L1 adipocytes after insulin stimulation. Error bars = standard deviation; *, 尸 = <0.05 compared to group treated with 6-NBDG alone. The data is representative of three independent experiments. 4D shows the results of visualizing fluorescence microscopy of 6-NBDG uptake after insulin stimulation. 6-NBDG was found to accumulate in the membranes of C2C12 myoblasts and cell-derived adipocytes. 4E shows C2C12 cells treated with p21 knock-down reversin (500 nM) and an lipogenesis cocktail showed insulin-sensitive free fatty acid release after epinephrine-stimulated (fat cell-specific function). Replacement of p21 siRNA with control siRNA failed to produce cells with the adipocyte-specific function. C2C12 myoblasts treated with reversin (500 nM) and lipogenic cocktails also showed insulin-sensitive free fatty acid release following epinephrine-stimulation. The amount of free fatty acids released and insulin sensitivity were lower than those observed in 3Τ3-ίΓ adipocytes. Error bars = standard deviation; *, P = <0.05 compared to the group treated with epinephrine alone. The data is representative of three independent experiments.
도 5 는 p21 하향-조절은 세포화된 근관에서 골형성을 촉진시킨다는 것올 나타내는 결과이다. 도 5a는 리버신 처리 및 p21 넉 -다운 후 골형성 조건에서 배양된 C2C12 세포물에서 증가된 알칼린 포스파타제 활성을 보여주는 그래프이다. p21 siRNA 를 대조군 siRNA 로 대체하는 것은 알칼린 포스파타제 활성에서 현저한 감소를 초래하였다. 골형성 조건에서 배양된 연골형성 전구세포인 ATCD5 세포는 양성 대조군으로 이용하였다. 도 5b 는 세포 용해물을 첨가하고 300 초 후 얻은 AttoPhos® 기질 용액 사진이다. 실험군들은 마이크로플레이트 밑에 상웅하는 웰 (번호)에 기재되어 있다, 도 5c 는 골형성 촉진 배지에서 10 일 동안 배양한 후, ATDC5 세포 및 C2C12 세포물에서 무기화를 시각화한 알리자린 레드 S 염색 결과이다. 리버신 (500 nM) 및 p21 siRNA로 처리된 C2C12 세포물은 500 nil 리버신이 처리된 C2C12 근육모세포와 유사한 정도의 무기화를 유도하였다. p21 siRNA 의 대조군 siRNA 로의 대체는 무기화 ί· 감소시켰다. 도 5d 는 발색 어세이를 통한 무기화의 반 -정량 결과이다. 리버신 (500 nM) 및 p21 siRNA 로 처리된 C2C12 세포물은 골형성 칵테일을 첨가한 후 무기화를 유도하였다. 무기화 정도는 500 nM 리버신 및 골형성 칵테일이 처리된 C2C12 근육모세포와 유사하였다. 하지만, 상기 세포들의 무기화는 골형성 칵테일 하에서 배양된 연골형성 전구세포인 ATCD5 세포에서 관찰된 정도보다 낮았다. 오차막대 = 표준편차; *, C2C12 세포물+골형성 칵테일과 비교하여 P = < 0.05. 데이터는 세 개의 서로 독립적인 실험들에 전체 평균이다. FIG. 5 shows that p21 down-regulation promotes bone formation in cellized root canals. 5A is a graph showing increased alkaline phosphatase activity in C2C12 cell cultures cultured in bone formation conditions after reversin treatment and p21 knock-down. Replacing p21 siRNA with control siRNA resulted in a significant decrease in alkaline phosphatase activity. ATCD5 cells, which were cartilage progenitor cells cultured under osteogenic conditions, were used as positive controls. Figure 5b is added to the cell lysate, and AttoPhos substrate solution ® picture obtained after 300 seconds. Experimental groups are described in wells (numbers) floating under microplates. FIG. 5C shows Alizarin Red S staining visualizing mineralization in ATDC5 cells and C2C12 cells after incubation for 10 days in osteogenic medium. C2C12 cells treated with reversin (500 nM) and p21 siRNA were 500 nil Reversin induced a degree of mineralization similar to C2C12 myoblasts treated. Substitution of p21 siRNA with control siRNA reduced mineralization. 5D is the semi-quantitative result of mineralization via the color assay. C2C12 cells treated with reversin (500 nM) and p21 siRNA induced mineralization after addition of a bone formation cocktail. The degree of mineralization was similar to C2C12 myoblasts treated with 500 nM reversin and bone formation cocktails. However, the mineralization of these cells was lower than that observed in ATCD5 cells, which are cartilage progenitor cells cultured under the bone formation cocktail. Error bars = standard deviation; *, P = <0.05 compared to C2C12 cell water + bone formation cocktail. Data is the overall mean for three independent experiments.
도 6 은--완전 분화된 골격근 조직으로부터 -다능성—세포를 -앋기 위한 화학적 접근방법의 개략적인 도식을 보여준다. 증식하는 근육모세포는 다핵 근관 융합체로의 분절를 증진시키기 위해 AraC가 처리될 수 있다. 【발명을 실시하기 위한 구체적인 내용】  FIG. 6 shows a schematic diagram of a chemical approach for pluripotent cells to pluripotency from fully differentiated skeletal muscle tissue. Proliferating myoblasts can be treated with AraC to enhance segmentation into multinucleated root canal fusions. [Specific contents to carry out invention]
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 보다 구체적으로 설명하기 위한 것으로, 본 발명의 요지에 따라 본 발명의 범위가 이들 실시예에 의해 제한되지 않는다는 것은 당업계에서 통상의 지식을 가진 자에 있어서 자명할 것이다. 실시예  Hereinafter, the present invention will be described in more detail with reference to Examples. These examples are only for illustrating the present invention more specifically, it will be apparent to those skilled in the art that the scope of the present invention is not limited by these examples in accordance with the gist of the present invention. . Example
실험재료 및 실험방법 Experimental Materials and Methods
항체 및 시약 Antibodies and Reagents
미오세버린, 미오세버린 B, 콜치신, 노코다졸, 뉴로다진, 사이토신 β-D—아라비노퓨라노시드 (AraC), 5-플루오로우라실 (5-FU), 5-브로모 -2' - 데옥시우리딘 (BrdU), 인간 트랜스페린, 소듐 샐레나이트, 덱사메타손, 3- 이소뷰틸— 1-메틸 크산틴 (IBMX) 및 인술린은 Sigma(M0, USA)로부터 구입하였다. 리버신은 Santa Cruz Biotechnology(CA, USA)로부터 구밉하였다. p21 siRNA 및 대조군 siRNA는 Santa Cruz Biotechnology(CA, USA)로부터 구입하였다, α-붕가로톡신 ( α-bungarotoxin) 및 6-(Ν— (그 니트로벤즈 -2-옥사 -1, 3-디아졸 -4-일)아미노 )-6-데옥시글루코오스 (6-NBDG)는 Molecular Probes (OR, USA)로부터 구입하였다. 세포 배양 Myose verbine, myose verbine B, colchicine, nocodazole, neurodazine, cytosine β-D—arabinofuranoside (AraC), 5-fluorouracil (5-FU), 5-bromo- 2'-deoxyuridine (BrdU), human transferrin, sodium salenite, dexamethasone, 3-isobutyl- 1-methyl xanthine (IBMX) and insulin were purchased from Sigma (M0, USA). Reversin was guipped from Santa Cruz Biotechnology (CA, USA). p21 siRNA and control siRNA were purchased from Santa Cruz Biotechnology (CA, USA), α-bungarotoxin and 6- (Ν— Nitrobenz-2-oxa-1, 3-diazol-4-yl) amino) -6-deoxyglucose (6-NBDG) was purchased from Molecular Probes (OR, USA). Cell culture
5 C2C12 마우스 Oros musculus) 근육모세포 (myoblasts)는 10% 우태아혈청 (FBS; Life Technologies, CA, USA), 50 유니트 /mL 페니실린 (Pen; Life Technologies. , CA, USA) 및 50 jug/ml 스트렙토마이신 (Strep; Life Technologies., CA, USA)이 보층된 DMEM(Dulbecco' s Modified Eagle' s Medium; Life Technologies. , CA,0 USA)으로 이루어진 증식 배지에서 유지하였다. 근육모세포는 5% 말 혈청, --- 50 유니트 /mL .페니실린 및- 50-^g/mL -스트렙토마이신이 보층된 DMEM 으로5 C2C12 mouse Oros musculus myoblasts contained 10% fetal bovine serum (FBS; Life Technologies, CA, USA), 50 units / mL penicillin (Pen; Life Technologies., CA, USA) and 50 jug / ml strepto Mycin (Strep; Life Technologies., CA, USA) was maintained in a growth medium consisting of DMEM (Dulbecco's Modified Eagle's Medium; Life Technologies., CA, 0 USA) supplemented. Myoblasts are 5% horse serum, --- 50 units / mL . Penicillin and -50- ^ g / mL -streptomycin supplemented with DMEM
, 이루어진 증식 배지에서 8 일 동안 배양을 통해 골격근의 근관 (myotubes)으로의 분화가 유도되었다. 분화 배지는 48 시간 마다 교체되었다. 분화 단계를 위해, 근육모세포는 콜라겐-코팅된 6-웰5 조직배양 플레이트 (Sigma, M0, USA)에서 배양하였다. 분화를 증진시키고 오염물을 제거하기 위해, 이전에 보고된 바와 같이 근육모세포 배양액에 50 μΜ AraC[26]를 72 시간 동안 처리하거나 또는 10 μΜ 5-FU[2기를 48 시간 동안 처리하였다. AraC 또는 5-FU 처리는 분화용 배양배지의 첨가한 후 18 시간째에 실시하였다. 순수한 근관 배양액을 얻기 위해, 분화된 근관0 배양액에 트립신을 처리하고 10 분 동안 800 rpm 에서 원심분리한 후, 10 mL 분화 배지에 부드럽게 재현탁하고 70 m 메쉬 (BD bioscience, CA, USA)에 통과시켜 단핵세포를 분리하였다. 이후, 상기 메쉬를 20 mL 분화 배지로 세척하여 근관을 수득하였다. Differentiation of skeletal muscle into myotubes was induced by incubation for 8 days in the growth medium. Differentiation medium was changed every 48 hours. For the differentiation step, myoblasts were cultured in collagen-coated 6-well 5 tissue culture plates (Sigma, M0, USA). To enhance differentiation and remove contaminants, myoblast cultures were treated with 50 μΜ AraC [26] for 72 hours or 10 μΜ 5-FU [2 phases for 48 hours as previously reported. AraC or 5-FU treatment was performed 18 hours after the addition of differentiation culture medium. To obtain pure root canal culture, trypsinized the differentiated root canal culture and centrifuged at 800 rpm for 10 minutes, then gently resuspend in 10 mL differentiation medium and pass through a 70 m mesh (BD bioscience, CA, USA) Mononuclear cells were isolated. The mesh was then washed with 20 mL differentiation medium to obtain root canal.
3T3-L1 마우스 ( /s musculus) 배아섬유아세포는 10% FBS, 505 유니트 /mL 페니실린 및 50 ^g/mL 스트랩토마이신이 보층된 DMEM 으로 이루어진 증식 배지에서 유지하였다. 3T3-L1 섬유아세포의 지방세포로의 분화가 다음과 같이 유도되었다: 48 시간 째의 포스트컨플루언트 세포 (0 일째로 간주)를 10% FBS, 0.5 mM 3-이소뷰틸 -1-메틸-크산틴, 2 //g/mL 덱사메타손, 1 //g/mL 인슐린, 50 유니트 /mL 페니실린 및 50 /g/mL0 스트랩토마이신이 보층된 DMEM 에서 2 일 동안 배양하였다. 이후, 세포는 2 일 마다 10% FBS 및 1 ^g/mL 인술린이 보충된 신선한 DMEM 에서 배양하였다. 분화 유도 후 8일 째 3T3-L1 지방세포가 실험에 이용되었다. 3T3-L1 mouse (/ s musculus) embryonic fibroblasts were maintained in proliferation medium consisting of DMEM supplemented with 10% FBS, 505 units / mL penicillin and 50 ^ g / mL straptomycin. Differentiation of 3T3-L1 fibroblasts into adipocytes was induced as follows: Post-confluent cells at 48 hours (considered day 0) were treated with 10% FBS, 0.5 mM 3-isobutyl-1-methyl-xanthine. , 2 // g / mL dexamethasone, 1 // g / mL insulin, 50 units / mL penicillin and 50 / g / mL0 straptomycin were incubated for 2 days in DMEM. After that, the cell Every 2 days incubated in fresh DMEM supplemented with 10% FBS and 1 ^ g / mL insulin. 8 days after induction of differentiation, 3T3-L1 adipocytes were used for the experiment.
ATDC5 세포는 광주과기원 생명과학부 전장수 교수로부터 제공받았다. ATDC5 세포는 5¾> FBS, 10 /g/mL 인간 트랜스페린 및 3Χ1(Γ8 M 소듐 셀레나이트를 포함하는 DMEM 및 Ham's F-12 배지의 1:1 복합물로 구성된 유지 배지에서 배양하였다. 연골형성 과정 (chondrogenesis)의 유도를 위해, 세포 배양 배지에 10 //g/mL 소 인슐린이 첨가되었다. 배치는 48 시간 마다 교체되었으며 연골세포는 8일 후 실험에 이용되었다. ATDC5 cells were provided by Professor Jeon Jang-soo from the School of Life Sciences, Gwangju Institute of Science and Technology. ATDC5 cells were cultured in a maintenance medium consisting of a 1: 1 complex of 5¾> FBS, 10 / g / mL human transferrin and 3Χ1 (DMEM with Γ 8 M sodium selenite and Ham's F-12 medium). To induce chondrogenesis, 10 // g / mL bovine insulin was added to the cell culture medium, the batch was changed every 48 hours and chondrocytes were used for the experiment 8 days later.
P19 마우스 배아 암종세포는 광주과기원 생명과학부 송미령 교수로부터 제공받았다. 이전에 보고된 바와 같이 [28], P19 세포의 신경형성 (neurogenesis)은 비-부착성 배양 디쉬에서 세포구 형성 (cell ball formation) 및 500 nM 레티노산 (ret inoic acid)의 처리를 통해 실시하였다. 신경교세포 -유사 (glial-like) 세포로 분열하는 성장을 억제하기 위해, 배양 배지에 플레이팅 후 48시간 째 50 μΜ AraC가 보층되었다. 골격근 근관의 세포화  P19 mouse embryonic carcinoma cells were provided by Professor Song Mi-Ryung, School of Life Sciences, Gwangju Institute of Technology. As reported previously [28], neurogenesis of P19 cells was performed through cell ball formation and treatment of 500 nM retinoic acid in non-adherent culture dishes. . To inhibit dividing growth into glial-like cells, 50 μΜ AraC was complemented 48 hours after plating in culture medium. Skeletal Muscle Root Canal Cellization
근관 배양액에 10 μΜ 미오세버린, 15 μΜ 미오세버린 Β, 1 yM 콜치신, 1 μΜ 노코다졸 또는 10 μΜ 탁솔을 24 시간 동안 처리하였다 (근관 분해에 있어서 IC50 마우스에서 보고된 바와 같은 처리방법 [16]). 세포화는 단핵세포 이외에 상대적으로 많은 양의 세포내 잔해물 (debris)을 생산한다고 밝혀졌다ᅳ 세포 단일층에 트립신을 처리하고 750 rpm 에서 5 분 동안 부드럽게 원심분리하였다. 세포 펠렛을 10 mL 증식 배지에서 부드럽게 재현탁하고 잔해물을 제거하기 위해 70 IM 세포 여과기 (cell strainer; BD Biosciences, CA, USA)에 통과시켰다. 단핵세포의 생존율을 측정하기 위해, 수득된 세포를 1X106 세포 AnL 의 밀도로 DMEM 에 재현탁한 후 0.4% 트립판 블루 다이 (Sigmaᅳ Aldrich, M0, USA)로 염색하였다. 100 개의 세포를 혈구계수기 (Mariefeld GmbH, Germany) 로 카운팅하고 트립판 블루로 염색되지 않은 세포를 생존 세포로 간주하였다. siRNA-매개된 유전자사일런싱 제조자의 프로토콜에 따라 (Santa Cruz Biotechnology, CA, USA) 단핵세포에 50 pmol 의 목적 siRNA 또는 대조군 siRNA를 트랜스펙션시켰다. 트랜스펙션 후 48시간 째의 단핵세포를 실험에 이용하였다. 세포융합지수 (fusion index)를 이용한 근육모세포 분화의 정량 Root canal cultures were treated with 10 μΜ myoserine, 15 μΜ myoserine Β, 1 yM colchicine, 1 μΜ nocodazole or 10 μΜ taxol for 24 hours (treatment as reported in IC 50 mice for root canal degradation). Method [16]). Cellization was found to produce a relatively large amount of intracellular debris in addition to monocytes. Trypsin was treated to the cell monolayer and gently centrifuged for 5 min at 750 rpm. The cell pellet was gently resuspended in 10 mL growth medium and passed through a 70 IM cell strainer (BD Biosciences, CA, USA) to remove debris. To determine the viability of monocytes, the obtained cells were resuspended in DMEM at a density of 1 × 10 6 cells AnL and stained with 0.4% trypan blue die (Sigma ᅳ Aldrich, Mo, USA). 100 cells were counted with a hemocytometer (Mariefeld GmbH, Germany) and cells not stained with trypan blue were considered viable cells. siRNA-mediated gene silencing Mononuclear cells were transfected with 50 pmol of the target siRNA or control siRNA according to the manufacturer's protocol (Santa Cruz Biotechnology, CA, USA). Monocytes 48 hours after transfection were used for the experiment. Quantification of Myoblast Differentiation Using Fusion Fusion Index
세포융합지수는 이전에 보고된 바와 같이 산출하였다 [29]. 배양액을 70 (v/v) 및 96%(v/v) 에탄올에서 연속적으로 반웅시켜 고정한 후, 핵을 시각화하기 위해 헤마특실린 용액 (Sigma, M0, USA)으로 염색하였다. 염색된 배양액의 이미지들이 CKX41 도립형 현미경 (Olympus, Japan)을 이용하여 얻어 DigiEye 330 디지털 카메라 (Dewinter , India) 및 Biowizard 4.3 소프트웨어 (Dewinter, India)로 기록하였다. 세포융합지수는 다섯 개의 현미경 필드에서 결정하였으며 다음과 같이 계산하였다: 세포융합지수 = 골격근 근관에 위치한 핵의 수 /핵의 전체 수. 근관은 세 개 또는 그 이상의 핵을 포함하는 융합체 (syncytia)로 제한하였다. 유세포 분석기  Cell fusion index was calculated as previously reported [29]. Cultures were fixed by successive reactions in 70 (v / v) and 96% (v / v) ethanol and then stained with hemafecillin solution (Sigma, M0, USA) to visualize nuclei. Images of the stained culture were obtained using a CKX41 inverted microscope (Olympus, Japan) and recorded with a DigiEye 330 digital camera (Dewinter, India) and Biowizard 4.3 software (Dewinter, India). The cell fusion index was determined in five microscopic fields and calculated as follows: cell fusion index = number of nuclei located in the skeletal muscle root canal / total number of nuclei. The root canal was limited to a syncytia containing three or more nuclei. Flow cytometer
세포주기상태의 분석: Analysis of cell cycle state:
트립신-처리된 세포 또는 세포물 (cellulate)을 1,000 rpm 에서 5 분 동안 원심분리한 후, 2.5X106 세포를 1 mL PBS 에 재현탁하였다. 상기 세포들은 파이펫팅을 통해 2.5 mL 에탄올로 조심스럽게 옮겨지고 얼음 하에서 15분 동안 반응시켜 고정되었다. 세포를 1,500 rpm에서 5분 동안 원심분리하고 500 ,의 PI 용액 (50 g/m 프로피디움 아이오다이드 (Sigma, MO, USA, 0.1 mg/mL RNase A(Sigma, M0, USA), 0.05% 트라이톤 X-100( Sigma MO, USA)에 재현탁한 후, 얼음 하에서 40 분 동안 반웅시켰다. 이후, PBS(3 mL)를 처리하고 세포를 1,500 rpm 에서 5 분 동안 원심분리하였다. 유세포 분석 (Beckman Coulter EPICS® XL-MCL)을 위해, 상층액을 제거하고 세포를 1X106 세포 /500 ≠ PBS 완층액의 밀도로 재현탁하였다. Trypsin-treated cells or cells were centrifuged at 1,000 rpm for 5 minutes and then 2.5 × 10 6 cells were resuspended in 1 mL PBS. The cells were carefully transferred to 2.5 mL ethanol via pipetting and fixed by reaction for 15 minutes under ice. Cells were centrifuged at 1,500 rpm for 5 minutes and a PI solution of 500, 50 g / m propidium iodide (Sigma, MO, USA, 0.1 mg / mL RNase A (Sigma, M0, USA), 0.05% Triton After resuspension in X-100 (Sigma MO, USA), the reaction was allowed to react for 40 minutes under ice, after which PBS (3 mL) was treated and the cells were centrifuged at 1,500 rpm for 5 minutes Flow cytometry (Beckman Coulter EPICS). ® XL-MCL), supernatants were removed and cells were resuspended to a density of 1 × 10 6 cells / 500 ≠ PBS complete solution.
DNA 합성의 분석: Analysis of DNA Synthesis:
세포 또는 세포물은 10 μΜ BrdU 와 24 시간 동안 반웅시킨 후, 트립신 /EDTA 로 얻었다. . 이후, 세포를 1,000 rpm 에서 5 분 동안 원심분리하고 PBS 로 세척한 후, 1X106 세포를 500 ^의 0.5% 파라포름알데하이드 (Sigma, MO, USA) (in PBS)에 재현탁하였다. 세포를 얼음 하에서 20 분 동안 반웅시키고 PBS로 한번 세척한 후, 1,500 rpm 에서 5 분 동안 원심분리하였다. 수득한 세포를 1 mL 의 신선하게 제조된 3N HCl/0.5% 트윈 20(Sigma, MO, USA)에 재현탁하여 25°C에서 20 분 동안 반웅시킴으로써 투과가 가능하도록 하였다. 세포를 세척한 후, 1 mL 의 0.1 M 다이소듐 테트라보레이트 (Sigma, MO, USA)에 재현탁하였다. 이후, 세포를 1,500 rpm 에서 5분 동안 원심분리하고 FACS 완충액 (PBS + 0.5% FBS + 5 mM EDTA)으로 세척하였다. 세포에 항-마우스 FITC 와 컨쥬게이션된 마우스 항 -BrdU 항체 (Sigma, MO, USA) (1: 100 회석)를 포함하는 FACS 완충액 (500 )을 처리하고 얼음 하에서 20 분 동안 반웅시킴으로써 세포를 염색하였다. 세포를 FACS 완층액으로 두 번에 걸쳐서 세척한 후, 500 ιΛ FACS 완충액에 IX 106 세포 밀도로 재현탁하였다. 세포는 20 분 안에 유세포 분석을 통해 분석하였다. 아세틸콜린 수용체들 (AChRs)의 α-붕가로톡신 표지 The cells or cells were reacted with 10 μΜ BrdU for 24 hours and then obtained with trypsin / EDTA. . Afterwards, cells were allowed to spin at 1,000 rpm for 5 minutes. After centrifugation and washing with PBS, 1 × 10 6 cells were resuspended in 500 ^ 0.5% paraformaldehyde (Sigma, MO, USA) (in PBS). The cells were reacted for 20 minutes under ice, washed once with PBS, and then centrifuged at 1500 rpm for 5 minutes. The obtained cells were resuspended in 1 mL of freshly prepared 3N HCl / 0.5% Tween 20 (Sigma, MO, USA) and allowed to permeate at 25 ° C. for 20 minutes. The cells were washed and then resuspended in 1 mL of 0.1 M disodium tetraborate (Sigma, MO, USA). Cells were then centrifuged at 1,500 rpm for 5 minutes and washed with FACS buffer (PBS + 0.5% FBS + 5 mM EDTA). Cells were stained by treating them with FACS buffer (500) containing a mouse anti-BrdU antibody (Sigma, MO, USA) conjugated with anti-mouse FITC (1: 100 lime) and reacting for 20 minutes on ice. . Cells were washed twice with FACS complete solution and then resuspended in 500 ιΛ FACS buffer at IX 10 6 cell density. Cells were analyzed by flow cytometry within 20 minutes. Α-Bungarotoxin Labeling of Acetylcholine Receptors (AChRs)
이전에 기재된 바와 같이 [14], 단핵세포는 배양 배지 내 20 /g/mL 의 FITC-컨쥬게이션된 α—붕가로특신과 5) C02/95 공기, 30°C에서 30 분 동안 반웅시켰다. 세포를 2% 파라포름알데하이드 (in PBS)로 고정하고 세척한 후, 형광 마운팅 배지 (Sigma-Aldrich, M0, USA)로 마운팅하여 MetaMorph 7.5 이미지 캡쳐 소프트웨어 (Molecular Devices, CA, USA)가 장착된 형광 현미경 (Olympus 1X81, Japan)에서 시각화하였다. 얻어진 이미지들을 포토샵 CS4 소프트웨어 (Adobe Systems Incorporated, CA, USA)로 분석하였다. 알파 -붕가로록신 표지를 정량화하기 위해, 이전에 기재된 바와 같이 [30] 세포를 PBS 로 세척하고 형광을 형광 마이크로플레이트 판독기 (SpectraMax GeminiXS, Molecular Devices, CA, USA; 여기파장 /형광 방출 λ: 495 nm/521 nm)로 측정하였다. 골격근 근관의 세포화로부터 유래된 단핵세포의 역분화 As previously described in [14], and mononuclear cells were banung in particular god 5) C0 2/95 air, 30 ° C to the inside of the 20 / g / mL culture medium FITC- conjugated α- Bungah for 30 minutes. Cells were fixed with 2% paraformaldehyde (in PBS) and washed, then mounted with fluorescence mounting medium (Sigma-Aldrich, M0, USA) to fluorescence with MetaMorph 7.5 image capture software (Molecular Devices, CA, USA) Visualization was carried out under a microscope (Olympus 1X81, Japan). The obtained images were analyzed with Photoshop CS4 software (Adobe Systems Incorporated, CA, USA). In order to quantify the alpha-bogaroroxine label, [30] cells were washed with PBS and fluorescence was fluorescence microplate reader (SpectraMax GeminiXS, Molecular Devices, CA, USA; excitation wavelength / fluorescence emission λ: 495 as previously described). nm / 521 nm). De-differentiation of monocytes derived from the cellization of skeletal muscle root canal
리버신의 최초 보고는 4 일 동안 5 μΜ 의 투여량을 이용하였다 [15]. 하지만, 본 발명자들은 상술한 농도가 미오세버린에 의한 근관으로부터 유래된 단핵세포에서 광범위한 세포독성 (30% 이상의 세포 단일층이 파괴됨)을 초래한다는 것을 발견하였다. 따라서, 세포독성을 감소시키기 위해, 단핵세포에 저농도의 리버신을 짧은 시간 동안 처리하였다 (3 일 동안 500 nM). 반웅 3 일 후, 리버신을 제거하고 단핵세포에 지방세포형성 (adipogenesis), 골형성 (osteogenesis) 또는 신경형성 (neurogenesis) 과정을 유도시켰다 (하기에 기재된 바와 같이). 세포화된 골격근 근관에서 골형성의 유도 The initial report of reversin used a dose of 5 μΜ for 4 days [15]. However, the present inventors have found that the above-mentioned concentration is derived from the root canal by It has been found that in derived monocytes results in a wide range of cytotoxicity (more than 30% of the cell monolayers are destroyed). Thus, to reduce cytotoxicity, mononuclear cells were treated with low concentrations of reversin for a short time (500 nM for 3 days). After 3 days of reaction, reversin was removed and monocytes were induced to undergo adipogenesis, osteogenic or neurogenesis processes (as described below). Induction of Bone Formation in Cellized Skeletal Muscle Root Canal
리버신-처리된 근육모세포에서 세포화된 골격근 근관에서 골형성의 유도는 이전에 기재된 바와 같이 실시하였다 [15, 31] . 세포를 50 βg/mL 아스코르빈산 -2-포스페이트 (Sigma, M0, USA), 0.1 μΜ 덱사메타손 및 10 mM β-글라이세로포스페이트 (Sigma, M0, USA)가 첨가된 DMEM + 10% FBS 에서 10 일 동안 배양하였다. 이후, 배양 조건을 ATDC5 세포에서 내연골성 (endochondral) 골형성을 모방하기 위해 스위치되었다: 5% FBS, 10 g/mL 인간 트랜스페린, 3X10— 8 M 소듐 샐레나이트 및 10 /mL 소 인술린을 포함하는 a-MEM(mini隨 m essential medium)에서 97% 공기 /3¾> C02 하에서 추가적으로 10 일 동안 배양하였다. 배양 배지는 48 시간 마다 교체하였다. 세포화된 골격근 근관에서 지방세포형성의 유도 Induction of bone formation in skeletal muscle root canals that were cellized in reversin-treated myoblasts was performed as previously described [15, 31]. Cells were 10 in DMEM + 10% FBS with 50 βg / mL ascorbic acid-2-phosphate (Sigma, M0, USA), 0.1 μΜ dexamethasone and 10 mM β-glycerophosphate (Sigma, M0, USA) Incubated for days. Then, the cartilage culture conditions in ATDC5 cellular (endochondral) was switch to mimic bone formation: 5% FBS, 10 g / mL of human transferrin, 3X10- 8 M sodium saelre nitro and 10 / mL bovine including Insular Lin Incubated for an additional 10 days in a-MEM (mini ¾ m essential medium) under 97% air / 3¾> C0 2 . Culture medium was changed every 48 hours. Induction of Adipocyte Formation in Cellized Skeletal Muscle Root Canal
세포화된 골격근 근관에서 지방세포형성의 유도는 리버신-처리된 근육모세포에서 이전에 기재된 바와 같이 실시하였다 [15, 31] . 세포를 1.7 μΜ 인슐린, 5 μΜ 덱사메타손 및 0.5 mM 이소뷰틸 메틸 크산틴이 첨가된 DMEM + 10%소 혈청에서 7일 동안 배양하였다. 오일 레드 0염색  Induction of adipocyte formation in the cellized skeletal muscle canal was performed as previously described in reversin-treated myoblasts [15, 31]. Cells were incubated for 7 days in DMEM + 10% bovine serum with 1.7 μΜ insulin, 5 μΜ dexamethasone and 0.5 mM isobutyl methyl xanthine. Oil Red 0 Dye
지방세포에서 세포 분화는 오일 레드 (XSigrna, M0, USA)염색을 통해 평가하였다。 3T3-L1 세포를 PBS 로 세척하고 4% 파라포름알데하이드로 상온에서 1 시간 동안 고정한 후, PBS 로 두 번에 걸쳐서 세척하였다. 이후, 세포를 녹여진 으 2% 오일 레드 0 를 포함하는 60% 이소프로파놀로 5분 동안 염색하였다. 인슐린—자극된 글루코오스 섭취량 (uptake) 분석 Cell differentiation in adipocytes was assessed by oil red (XSigrna, M0, USA) staining. 3T3-L1 cells were washed with PBS, fixed with 4% paraformaldehyde for 1 hour at room temperature, and then twice with PBS. Washed. Cells were then stained for 5 minutes with 60% isopropanol containing 2% oil red 0 dissolved. Insulin—Stimulated Glucose Uptake Analysis
3T3-L1 지방세포 또는 근육-유래된 지방세포를 2X105 세포 /웰의 농도로 24-웰 플레이트 (BD Bioscience, CA, USA)에 분주하였다. 48 시간 후, 세포를 혈청 -부재 저농도 글루코오스 DMEM에서 3 시간 동안 배양시켰다. 세포에 100 nM 인슐린 및 100 μΜ 6-(Ν_(7_니트로벤즈ᅳ 2-옥사 -1,3-디아졸 -4- 일)아미노 )-2-데옥시글루코오스 (6— NBDG)를 첨가한 후, 37°C에서 30 분 동안 반웅시켰다. 이후, 세포를 PBS 로 세척하고 1% 트라이톤 X-100 을 포함하는 100 μΐ 포타슘 포스페이트 완충액 (ρΗ 10)으로 용해시켰다. 용해된 용액에 50 ≠ DMS0 를 첨가한 후, 그 일부 (120 를 96-웰 마이크로타이터 플레이트 (BD Bioscience, - CA, USA)로 옮겼다 . - 형광은 형광 마이크로플레이트 판독기 (Spec traMAX GeminiXS 및 SoftMax Pro V5 software, Molecular Devices , CA, USA; ex - 466 nm, Xem = 540 nm)를 이용하여 측정하였다ᅳ 3T3-L1 adipocytes or muscle-derived adipocytes were dispensed into 24-well plates (BD Bioscience, CA, USA) at a concentration of 2 × 10 5 cells / well. After 48 hours, cells were incubated for 3 hours in serum-free low glucose DMEM. After addition of 100 nM insulin and 100 μΜ 6- (Ν_ (7_nitrobenzium 2-oxa-1,3-diazol-4-yl) amino) -2-deoxyglucose (6—NBDG) to the cells , Reacted at 37 ° C for 30 minutes. Cells were then washed with PBS and lysed with 100 μΐ potassium phosphate buffer (ρΗ 10) containing 1% Triton X-100. 50 ≠ DMS0 was added to the dissolved solution, then a portion (120) was transferred to a 96-well microtiter plate (BD Bioscience, CA, USA).-Fluorescence was measured using a fluorescence microplate reader (Spec traMAX GeminiXS and SoftMax Pro). V5 software, Molecular Devices, CA, USA; ex -466 nm, X em = 540 nm)
NBDG 섭취에 대한 현미경적 분석을 위해, 근육-유래된 지방세포를 For microscopic analysis of NBDG uptake, muscle-derived adipocytes were
2.5X105 세포 /웰의 농도로 6-웰 플레이트 (BD Bioscience, CA, USA)에 분주하여 24시간 동안 배양하였다. 이후, 세포에 목적 약물 (drug)과 함께 100 μΜ 6-NBDG 를 처리하였다. 세포를 PBS 로 세 번에 걸쳐서 세척하고 3.7%(w/v) 포름알데하이드 (in PBS)(Sigma, MO, USA)로 고정하였다. 세포를 PBS 로 세 번에 걸쳐서 세척한 후, 플루오마운트 (Fluormount; Sigma, MO, USA)로 마운팅하였다. NBDG 섭취는 MetaMorph 7.5 이미지 캡쳐 소프트웨어 (Molecular Devices, CA, USA)가 장착된 형광 현미경 (Olympus 1X81, Japan)을 이용하여 즉시 시각화하였다. 얻어진 이미지들은 포토샵 CS4 소프트웨어 (Adobe Systems Incorporated, CA, USA)로 분석하였다. 유리 지방산 방출 어세이 Aliquots were placed in 6-well plates (BD Bioscience, CA, USA) at a concentration of 2.5 × 10 5 cells / well and incubated for 24 hours. Thereafter, cells were treated with 100 μΜ 6-NBDG together with the desired drug (drug). Cells were washed three times with PBS and fixed with 3.7% (w / v) formaldehyde (in PBS) (Sigma, MO, USA). Cells were washed three times with PBS and then mounted with Fluormount (Sigma, MO, USA). NBDG uptake was immediately visualized using a fluorescence microscope (Olympus 1X81, Japan) equipped with MetaMorph 7.5 image capture software (Molecular Devices, CA, USA). The obtained images were analyzed with Photoshop CS4 software (Adobe Systems Incorporated, CA, USA). Free Fatty Acid Release Assays
이전에 기재된 방법에 따라, 지방세포로부터 에피네프린—자극된 유리 지방산 (free fatty acid, FFA) 방출을 유리 지방산 정량 키트 (Biovision, CA, USA)를 이용하여 측정하였다. 24-웰 플레이트에서 컨플루언스에 도달할 때까지 성장된 3T3-L1 지방세포를 혈청 -부재 배양 배지에서 3 시간 동안 배양시켰다. 이후, 지방세포에 염에 녹여진 10 μΜ 에피네프린 (Sigma, MO, USA)을 처리하여 FFA 방출을 유도시켰다. 에피네프린을 처리하기 10 분 전에 목적 약물을 첨가하였다. 세포 용해물을 수득하여 50 ^의 유상 (organic phase)을 발색 어세이 (VERSA Max microplate reader , Molecular Devices, CA, USA)에 이용하였다. 표준 곡선을 제작하기 위해 팔미트산을 이용하였다. 알칼린 포스파타제 어세이 According to the previously described method, epinephrine-stimulated free fatty acid (FFA) release from adipocytes was measured using the free fatty acid quantification kit (Biovision, CA, USA). 3T3-L1 adipocytes grown until reaching confluence in 24-well plates were incubated for 3 hours in serum-free culture medium. Then, 10 μΜ dissolved in salt in fat cells Epinephrine (Sigma, MO, USA) was treated to induce FFA release. The desired drug was added 10 minutes before the epinephrine treatment. Cell lysates were obtained and a 50 ^ organic phase was used for the chromogenic assay (VERSA Max microplate reader, Molecular Devices, Calif., USA). Palmitic acid was used to make a standard curve. Alkaline phosphatase assays
알칼린 포스파타제 활성은 AttoPhos® AP 형광 기질 시스템 (Promega, WI, USA)을 이용하여 측정하였다. 세포를 PBS로 두 번에 걸쳐서 세척하고 250 !Λ Cel Lytic™ M(Sigma, MO, USA)에서 용해시켰다. 어세이를 위해, 100 /g/100 ^의 세포 용해물이 이용되었으며, 이때 멀티—채널 파이펫을 이용하여 100 ^의 AttoPhos 기질이 즉시 첨가되었다. 형광은 마이크로플레이트 판독기 (SpectraMAX GeminiXS, Molecular Devices, CA, USA; λ = 420 nm; Xem = 560 nm)를 이용하여 측정하였다. 알리자린 레드 염색 Alkaline phosphatase activity was measured using the fluorogenic substrate AttoPhos ® AP system (Promega, WI, USA). Cells were washed twice with PBS and lysed at 250! Λ Cel Lytic ™ M (Sigma, MO, USA). For the assay, 100 / g / 100 ^ cell lysate was used, with 100 ^ AttoPhos substrate added immediately using a multi-channel pipette. Fluorescence was measured using a microplate reader (SpectraMAX GeminiXS, Molecular Devices, CA, USA; λ = 420 nm; X em = 560 nm). Alizarin Red Dyeing
세포를 PBS 로 한 번 세척하고 포스페이트-완층된 포르말린 (Sigma, M0, USA)으로 20 분 동안 고정시켰다. 고정된 세포를 증류수로 한 번 세척한 후, 증류수에 녹여진 1% 알리자린 레드 S(Sigma, M0, USA)로 5 분 동안 염색하였다. 나머지 다이를 증류수로 제거하고 세포를 한 번 더 세척하였다. 마지막으로, 세포를 공기 중에서 건조시킨 후, 염색된 세포의 이미지들을 광학 현미경 (CKX41, Olympus, Japan)으로 얻고 이를 소프트웨어 (Leica)로 분석하였다. 알리자린 레드를 이용한무기화 (mineralization)의 정량  Cells were washed once with PBS and fixed for 20 minutes with phosphate-completed formalin (Sigma, M0, USA). The fixed cells were washed once with distilled water and then stained with 1% alizarin red S (Sigma, M0, USA) for 5 minutes dissolved in distilled water. The remaining die was removed with distilled water and the cells washed once more. Finally, after the cells were dried in air, images of the stained cells were obtained under an optical microscope (CKX41, Olympus, Japan) and analyzed by software (Leica). Quantification of mineralization using alizarin red
이전에 기재된 바와 같이 알리자린 레드 염색의 정량을 실시하였다 [33]. 간략하게는, 알리자린 레드 염색 후 800 ^의 10%(v/v) 아세트산을 각 웰에 첨가하고 플레이트를 상온에서 30 분 동안 교반하면서 반응시켰다. 단일층이 세포 스크래퍼를 이용해 플레이트로부터 수득되어 10 (v/v) 아세트산과 함께 1.5-mL 튜브 (microcentr i fuge tube)로 옮겨서 흔합 (vortex)되었다. 슬러리에 500 μί 미네랄 오일을 로딩하고 10분 동안 85°C까지 열을 가한 후, 얼음에서 급속히 냉각시켰다. 이후, 슬러리를 20,000 g 에서 15 분 동안 원심분리하고 500 ^의 상층액을 새로운 튜브 (microcentrifuge tube)로 옮겼다. 상층액에 200 ^의 10%(v/v) 암모늄 하이드록사이드를 첨가하고 상층액의 분취액 (150 )을 405 nm 에서 96-웰 포맷의 불투명한 벽 및 투명한 바닥을 가진 플레이트에서 세 쌍으로 (triplicate) 판독하였다. 데이터의 통계 분석 Alizarin red staining was quantified as described previously [33]. Briefly, after Alizarin Red staining, 800 ^ of 10% (v / v) acetic acid was added to each well and the plates were reacted with stirring for 30 minutes at room temperature. A monolayer was obtained from the plate using a cell scraper and vortexed by transferring to a 1.5-mL microcentr i fuge tube with 10 (v / v) acetic acid. Load 500 μί mineral oil into the slurry and for 10 minutes After heating to 85 ° C, it cooled rapidly on ice. The slurry was then centrifuged at 20,000 g for 15 minutes and the 500 ^ supernatant was transferred to a new microcentrifuge tube. Add 200 ^ 10% (v / v) ammonium hydroxide to the supernatant and aliquot (150) of the supernatant in three pairs on plates with opaque walls and transparent bottoms in 96-well format at 405 nm. (triplicate) Read. Statistical analysis of the data
통계적 유의성을 결정하기 위해 Mann-Whitney U 검정 (TalkStats software; Jelsoft Enterprises Ltd. , UK)을 이용하였다. 0.05 이하의 P 값 (양쪽 -꼬리검정)을 유의한 것으로 간주하였다. 실험결과 및 추가논의사항  Mann-Whitney U test (TalkStats software; Jelsoft Enterprises Ltd., UK) was used to determine statistical significance. P values below 0.05 (both-tail test) were considered significant. Experiment result and additional discussion
근관 세포화의 유도를 위한 세포 배양 조건의 확립: AraC 처리는 근육 분화를 증진시키며 ; 미오세버린은 근육 세포화의 유도에 있어서 바람직한 제제이다 Establishment of Cell Culture Conditions for Induction of Root Canal Cellization: AraC treatment enhances muscle differentiation; Myosevery is a preferred agent in the induction of muscle cellularization
본 연구에서 이용된 작은 분자들의 리스트가표 1에 기재되어 있다. 【표 1】  The list of small molecules used in this study is shown in Table 1. Table 1
골격근 역분화 및 재-분화를 촉진시키기 위해 본 연구에서 이용된 작은 분자들의 구조, The structure of the small molecules used in this study to promote skeletal muscle dedifferentiation and re-differentiation,
Figure imgf000029_0001
Figure imgf000029_0001
Figure imgf000030_0001
근관 세포화의 과정을 효과적으로 연구하기 위해, 높은 정도의 근육 분화가 이루어져야 한다。 하지만, 비분화된 근육모세포가 포함된 배양액의 오염이 주요 문제로, 이로 인해 세포화된 근관 배양액으로부터 모순되는 결과들이 초래되었다 [14, 20]. C2C12 근육모세포 배양에서 근육형성 과정을 증가시키기 위해 항-유사분열 약물 (anti-mitotic drugs)인 AraC[26] 및 5-FU[2기가 증식하는 근육모세포를 제거할 목적으로 테스트되었다. 분화 과정 동안 AraC 의 처리는 근관에 삽입된 핵의 수를 측정하는 지표인 세포융합지수 (fusion index)를 현저하게 증가시킨다는 것을 확인하였다 (도 la). 한편, 근육형성 과정 의 다른 측정수단 (measure)은 배양에서 자발적인 '트위칭 (twitching)' 근관의 존재이다. 또한, 세포가 콜라겐-코팅된 디쉬에서 배양되는 경우 분화하는 C2C12 세포에 대한 AraC 처리는 자발적으로 수축하는 '트위칭 (twitching)' 근관의 존재를 증가시킨다는 것을 발견하였다 (도 lb). 따라서, 본 연구에서 AraC 의 처리는 C2C12 분화를 증가시키는 데 이용되었다.
Figure imgf000030_0001
In order to effectively study the process of root canal cellization, a high degree of muscle differentiation must be achieved. However, contamination of the culture medium containing undifferentiated myoblasts is a major problem, which results from the cultured root canal culture. Contradictory results have resulted [14, 20]. In order to increase the muscle formation process in C2C12 myoblast cultures, it was tested to remove the myoblasts proliferating phase 2 anti-mitotic drugs AraC [26] and 5-FU [2]. Treatment of AraC during the differentiation process was found to significantly increase the fusion index (indicator la), which is an indicator of the number of nuclei inserted into the root canal (Fig. La). On the other hand, another measure of the muscle formation process is the presence of spontaneous 'twitching' root canals in culture. In addition, AraC treatment on C2C12 cells that differentiate when cells are cultured in collagen-coated dishes increased the presence of spontaneously contracting 'twitching' root canals (FIG. Lb). Therefore, treatment of AraC in this study was used to increase C2C12 differentiation.
근관 세포화 과정을 효과적으로 연구하기 위해, 최적의 세포화 -유도 제제 (agent)가 동정되었다. 네 개의 다른 작은 분자들이 상대적으로 낮은 세포독성을 유발하면서 세포화를 유도할 수 있는 지를 테스트하였다: 콜치신, 노코다졸, 미오세버린 및 미오세버린 B. 미오세버린에 의한 세포화 유도는 가장 많은 수의 단핵세포들을 생산하는 것으로 나타났으며, 이들은 세포 생존율에 대한 마커인 0.4% 트립판 블루 다이를 배제시킬 수 있었다 (도 lc). 따라서, 미오세버린이 C2C12 세포화 및 이형분화0^313^£ ^611^ 1위 연구에 이용되었다. p21 하향-조절이 세포화된 근관에 증식, 세포주기로의 재 -진입 및 분화를 유도할 수 있다:  In order to effectively study the root canal cellularization process, an optimal cellularization-inducing agent has been identified. Four other small molecules were tested to induce cellularization with relatively low cytotoxicity: induction of cellularization by colchicine, nocodazole, myosebourne and myosebourne B. myosebourne It was shown to produce the largest number of monocytes, which could rule out 0.4% trypan blue die, a marker for cell viability (FIG. Lc). Thus, myoseburin was used for C2C12 cellization and heterodifferentiation 0 ^ 313 ^ £ ^ 611 ^ first place studies. p21 down-regulation can induce proliferation, re-entry into the cell cycle and differentiation into the cellized root canal:
C2C12 근관의 세포화에 의해 발생된 단핵세포들이 분주되고 24 시간 후, 스크램블 siRNA 및 p21 siRNA 가 처리되었다 (도 2a). 세포 핵에 대한 PKpropidium iodide) 염색 결과는 p21 하향-조절이 세포주기의 불웅성 (refractory) G0 단계에서 G2-M 단계로의 쉬프트를 유도한다는 것을 보여줬다 (도 2b). 또한, 이러한 효과는 적을 지라도 현저한 세포수의 증가를 초래하는 p21 하향-조절과 함께 40 X배율에서 관찰한 세포 수의 카운팅에서도 확인할 수 있었다 (도 2c). 상술한 결과는 p21 하향-조절이 증가된 DNA 합성의 지시자인 BrdU 가 삽입된 세포들의 부위에서의 증가를 야기한다는 관찰을 통해 다시 확인되었다 (도 2d). The scrambled siRNA and p21 siRNA were treated 24 hours after the monocytes generated by C2C12 root canal cell division were dispensed (FIG. 2A). PKpropidium iodide) staining on the cell nuclei showed that p21 down-regulation induced a shift from refractory G 0 phase to G 2 -M phase of the cell cycle (FIG. 2B). This effect was also confirmed in counting the number of cells observed at 40 × magnification with p21 down-regulation leading to a small, but significant increase in cell number (FIG. 2C). The above results indicate that p21 down-regulation It was again confirmed by the observation that BrdU, an indicator of increased DNA synthesis, caused an increase in the site of the inserted cells (FIG. 2D).
이전 연구결과는 근관의 세포화가 모근관 (parent myotube)과 비교하여 단핵세포의 분화상태에서 변화를 야기하지 않는다는 것을 나타냈다 [20]· 예를 들어 단핵세포는 후기 근조직 마커 (late myogenic markers)의 발현을 유지한다. 세포화된 근관으로부터 유래된 단핵세포의 분화된 상태에 대한 p21 하향-조절의 효과는 근관 형성의 마커인 아세틸콜린 수용체에 결합하는 FITC-컨쥬게이션된 α-붕가로록신을 이용하여 평가하였다. ρ21 하향-조절은 단핵세포의 α-붕가로특신 표지를 감소시켰는데 (도 2e 및 도 2f), 이는 근관으로부터 벗어나 ( 'away' ) 근육모세포로 향하는 ( 'towards' ) 분화 상태로의 쉬프트가 일어났음을 의미한다. p21 하향-조절의 효과는 근육세포의 발달 가소성 (developmental plasticity)을 증가시키는 것으로 알려진 리버신에 의한 효과와유사하였다. 세포화된 근관은 p21 하향 -조절 후 고유의 근전위 (myogenic potential)를 유지한다:  Previous studies have shown that myotube cellularization does not cause a change in monocyte differentiation compared to parental myotubes [20]. For example, monocytes do not express late myogenic markers. Keep it. The effect of p21 down-regulation on the differentiated state of monocytes derived from the cellized root canals was evaluated using FITC-conjugated α-bogaroroxine that binds to the acetylcholine receptor, a marker of root canal formation. ρ21 down-regulation reduced the specific markers by α-unga of monocytes (FIGS. 2E and 2F), which shifted from the root canal ('away') to myoblasts ('towards') to differentiation It means that it happened. The effect of p21 down-regulation was similar to that by reversin, which is known to increase the developmental plasticity of muscle cells. The cellized root canal maintains its inherent myogenic potential after p21 down-regulation:
유미목 양서류의 사지 재생의 홍미로운 특징은 초기에 근관의 세포화가 일어난 후 성장하는 사지에서 근육을 다시 형성하기 위해 증식 및 분화가 일어난다는 것이다 [17]. p21 하향-조절이 세포환된 근관에서 증식 반응을 유도할 수 있다는 사실에 비추어 볼 때, 근육세포 분화 (myogenic differentiation)로 재-진입하는 단핵세포의 능력이 조사되었다. 첫 테스트로서, 본 발명자들은 근육세포의 발달 가소성을 증가시키는 것 [34]으로 알려진 리버신의 근육세포 분화 상의 효과를 조사하였다. 리버신 처리는 근육형성 과정으로 재 -진입하려는 근육세포 및 세포화된 근관와 능력을 감소시키는 것으로 확인되었다 (도 3a). 흥미롭게도, 세포화된 단핵세포의 근육형성에 있어서 리버신의 억제 효과는 p21 의 넉- 다운에 의해 극복될 수 있었다. 따라서, p21 의 조절 (treatment)은 근전위의 감소 없이 세포화된 근관의 증식을 유도한다. 결과적으로, 근관은 p21 넉 -다운 후 더 높은 세포융합지수 (도 3b 및 도 3c)를 나타냈다. p21 하향-조절은 세포화된 근관에서 지방세포로의 전환을 촉진시킨다: A fascinating feature of limb regeneration in Rhesus amphibians is that early canalization occurs followed by proliferation and differentiation to rebuild muscles in growing limbs [17]. In light of the fact that p21 down-regulation can induce a proliferative response in the transplanted root canal, the ability of monocytes to re-enter myogenic differentiation has been investigated. As a first test, we investigated the effects of reversin on myocyte differentiation, known to increase the developmental plasticity of myocytes [34]. Reversin treatment was found to reduce muscle cells and cellized root canal and ability to re-enter the muscle building process (FIG. 3A). Interestingly, the inhibitory effect of reversin on the muscle formation of cellized monocytes could be overcome by knock-down of p21. Thus, the treatment of p21 induces proliferation of the cellized root canal without a decrease in myoelectric potential. As a result, the root canal showed a higher cell fusion index (p. 3b and 3c) after p21 knock-down. p21 down-regulation promotes the conversion of cellularized root canal to adipocytes:
리버신이 근육모세포에서 더 높은 가소성을 유도하고 이후 지방형성 인자들 (adipogenic factors)의 칵테일 하에서의 배양을 통해 지방세포로의 분화로 이어지는 것이 알려졌다 [15]. 분화하는 지방세포에서 축적하는 지질 물방울 (lipid droplets)에 결합하는 오일 레드 0 염색이 감소하는 결과에서 볼 수 있듯이 (도 4a), 대조군 siRNA, 리버신 및 지방형성 인자들에 의해 세포화된 근관은 지방세포로 분화할 수 없다. 하지만, 증가된 오일 레드 0 염색에서 볼 수 있듯이 (도 4a), 리버신 처리 전에 세포화된 근관에서 p21 의 하향-조절은 지방형성 세포들 (adipogenic lineage)로의 전환을 촉진시켰다. 지질의 축적은 광학 현미경으로 관찰할 수 있었다 (도 4b). 인슐린-자극된 글루코오스의 섭취는 지방형성 세포들의 특징이다 [35]. 리버신 및 지방형성 인자들에 의해 세포화된 근관은 형광—태깅된 글루코오스 유사체의 인슐린-자극된 섭취를 나타내지 않았다 (도 4c). 더욱이, 리버신 처리 전에 세포화된 근관에서 p21 의 하향-조절은 인슐린 자극 후 글루코오스를 이동시키도록 세포의 전환을 촉진시켰다 (도 4c). 하지만, 6-NBDG 섭취의 정도는 3T3-L1 지방세포주에서 관찰된 정도보다 낮았다. 현미경 분석은 지방세포 형태 (morphology)의 특징인 지질 축적을 반영하는 세포에 대한 형광-태깅된 글루코오스의 증가된 결합을 보여주었다 (도 4d). 지방세포에서 안슐린 민감성의 추가적인 테스트는 에피네프린 자극 후 인슐린-매개된 유리 지방산 방출의 억제를 테스트하는 것이다 [36, 37]. 대조군 siRNA, 리버신 및 지방형성 인자들에 의해 세포화된 근관은 에피네프린 자극 후 유리 지방산 방출의 인슐린-자극된 억제를 나타내지 않았다 (도 4e). 하지만, 리버신 처리 전에 세포화된 근관에서 p21의 하향ᅳ조절은 에피네프린에 노출 후 감소되는 유리 지방산의 양을 가지는 세포에서 세포내 유리 지방산의 증가된 양을 가지는 세포로의 전환을 촉진하였다 (도 4e). 더 나아가 인슐린 처리는 상기 세포군 (cell population)에서 에피네프린-자극된 유리 지방산 방출을 억제하였는데, 이는 리버신 및 지방형성 인자들이 처리된 근육모세포에서 관찰되는 것과 유사한 정도였다 (도 4e). 하지만, 지방상 축적 및 방출의 정도는 3T3-L1 지방세포주에서 관찰된 정도보다 낮았다. p21 하향-조절은 세포화된 근관에서 골형성을 촉진시킨다: It is known that reversin induces higher plasticity in myoblasts and then leads to differentiation into adipocytes through cultivation under a cocktail of adiogenic factors [15]. As can be seen from the decrease in oil red 0 staining that binds to lipid droplets accumulating in the adipocytes that differentiate (FIG. 4A), the root canal cellized by control siRNA, reversin and lipogenic factors Cannot differentiate into adipocytes. However, as can be seen in the increased oil red 0 staining (FIG. 4A), down-regulation of p21 in the cellized root canal prior to reversin treatment promoted the conversion to adipogenic lineage. Accumulation of lipids could be observed by light microscopy (FIG. 4B). Ingestion of insulin-stimulated glucose is characteristic of adipogenic cells [ 35 ]. Root canal cellized by reversin and lipogenic factors did not show insulin-stimulated uptake of fluorescence-tagged glucose analogs (FIG. 4C). Moreover, down-regulation of p21 in the root canal that was cellized prior to reversin treatment promoted the conversion of cells to shift glucose after insulin stimulation (FIG. 4C). However, the level of 6-NBDG intake was lower than that observed in the 3T3-L1 adipocyte line. Microscopic analysis showed increased binding of fluorescence-tagged glucose to cells reflecting lipid accumulation, which is characteristic of adipocyte morphology (FIG. 4D). An additional test of insulin sensitivity in adipocytes is to test the inhibition of insulin-mediated free fatty acid release following epinephrine stimulation [36, 37]. Root canal cellized by control siRNA, reversin and lipogenic factors did not show insulin-stimulated inhibition of free fatty acid release after epinephrine stimulation (FIG. 4E). However, downregulation of p21 in the cell root canal prior to reversin treatment promoted the conversion of cells with increased amounts of intracellular free fatty acids to cells with reduced amounts of free fatty acids after exposure to epinephrine (FIG. 4e). Furthermore insulin treatment inhibited the release of epinephrine-stimulated free fatty acids in the cell population, similar to that observed in myoblasts treated with reversin and lipogenic factors (FIG. 4E). However, the degree of fat phase accumulation and release was lower than that observed in the 3T3-L1 adipocyte line. p21 down-regulation promotes bone formation in the cellized root canal:
리버신이 처리된 근육모세포는 골형성 인자들 (osteogenic factors)의 칵테일 하에서 배양함으로써 조골세포로 분화될 수 있다 [15]. 알칼린 포스파타제 활성의 획득은 Wnt-1-유도된 분비 단백질 1 및 골형성 단백질 2 (Bone morphogenet ic protein 2) 같은 골형성 과정의 조절인자들로 트랜스펙션된 C2C12 근육모세포에서 골형성 전환 (conversion)의 지시인자로서 이용되어 왔다 [38]. 알칼린 포스파타제 활성의 부재에서 보여지듯이, 골형성 인자들 하에서 세포화된 C2C12 근관의 배양은 골형성 세포로의 전환을 유도하지 않았다 (도 5a 및 도 5b). 하지만, 세포화된 후 리버신 및 골형성 인자들의 칵테일이 처리된 근관에서 p21 하향ᅳ조절은 현저하게 증가된 알칼린 포스파타제 활성을 가지는 세포로의 분화를 촉진시켰다 (도 5a 및 도 5b). 비톡 본 연구의 실험군에서 비처리된 C2C12 세포물과 비교하여 알칼린 포스파타제 활성이 상대적인 작지만 유의하게 증가하는 것으로 나타났을 지라도, 상기 알칼린 포스파타제 활성의 획득은 대조군, 스크램블 siRNA 로 처리된 세포화된 근관에서 보여지는 활성보다 훨씬 더 뚜렷하게 증가하였다.  Reversin-treated myoblasts can be differentiated into osteoblasts by culturing under a cocktail of osteogenic factors [15]. Gaining alkaline phosphatase activity results in conversion of bone formation in C2C12 myoblasts transfected with regulators of the bone formation process such as Wnt-1-induced secreted protein 1 and Bone morphogenet ic protein 2 Has been used as an indicator [38]. As shown in the absence of alkaline phosphatase activity, culture of C2C12 root canal cellized under osteogenic factors did not induce conversion to osteogenic cells (FIGS. 5A and 5B). However, p21 downregulation in the root canal treated with a cocktail of reversin and osteogenic factors after cellularization promoted differentiation into cells with markedly increased alkaline phosphatase activity (FIGS. 5A and 5B). Beating Although alkaline phosphatase activity was shown to be relatively small but significantly increased compared to untreated C2C12 cells in the experimental group of the present study, the acquisition of the alkaline phosphatase activity was achieved by control of the cultivated root canal treated with scrambled siRNA. It is even more markedly increased than the activity seen in.
무기화 (mineralization)은 골형성 동안 일어나는 골화 (ossification)의 한 특징이다 [33]. 석회화 (calci f icat ion)는 p21 넉-다운, 리버신 처리 및 골형성 배지에서 배양 후 C2C12 세포물에서 알리자린 레드 S 염색으로 확인하였다. 내연골성 (endochondral) 골형성을 일으키도록 유도된 ATDC5 세포가 대조군으로서 이용되었다. 광학 현미경 분석은 p21 넉-다운, 리버신 처리 및 골형성 배지에서 배양 후 세포물에서 무기화가 일어났음을 나타냈다 (도 5c). 무기화는 마이크로플레이트 판독기-기반된 어세이로 정량하였다 (도 5d)[33]. P21 넉-다운, 리버신 처리 및 골형성 배지에서 배양 후 C2C12 세포물은 동일한 조건에서 배양된 C2C12 근육모세포와 유사한 정도로 무기화의 증가를 나타냈다. 무기화 레벨은 골형성 배양 후 ATDC5 세포에서 관찰되는 정도보다 더 낮았다. 더욱이, p21 siRNA 대신에 대조군 siRNA 로 처리된 C2C12 세포물은 골형성 배양 후 현저하게 감소된 레벨의 무기화를 야기하였다. Mineralization is a feature of ossification that occurs during bone formation [33]. Calcification (calci f icat ion) was confirmed by alizarin Red S staining in C2C12 cells after p21 knock-down, reversin treatment and incubation in osteogenic medium. ATDC5 cells induced to cause endochondral bone formation were used as controls. Optical microscopy analysis showed that mineralization occurred in the cell water after incubation in p21 knock-down, reversin treatment and bone formation medium (FIG. 5C). Mineralization was quantified by microplate reader-based assays (FIG. 5D) [33]. C2C12 cells after culture in P 21 knock-down, reversin treatment and bone formation medium showed an increase in mineralization to a similar extent as C2C12 myoblasts cultured under the same conditions. Mineralization levels were lower than observed in ATDC5 cells after osteogenic culture. Moreover, C2C12 cells treated with control siRNA instead of p21 siRNA resulted in a significantly reduced level of mineralization after bone formation culture.
본 연구에서 제시된 결과들은 양서류에서 사지 재생의 초기 단계를 모방한 화학물질-기반된 프로토콜의 첫 번째 기재이다. 이전에, 많은 실험적 접근방법들은 다능성 세포 또는 줄기세포로 형성시키기 위해 성체 세포 (adult cells)를 유도하도록 개발되어 왔다. 상기 예들은 체세포 핵전이 (somatic cell nuclear transfer) 및 전능성과 관련된 전사인자들의 트랜스펙션을 포함한다 [45]. 하지만, 상술한 접근방법들은 인간에서 세포군 (eel hilar population)의 대부분을 차지하는 완전히 분화된 성체세포를 이용하지 않았다 [46]. 또한, 상술한 접근방법들은 현재 낮은 효율 같은 기술적 문제에 직면하고 있다. 본 발명에서 기재하고 있는 다능성을 획득하기 위한 화학적 접근방법은 매우 높은 불웅성을 가지는 것으로 간주되고 있는 골격근 조직 [22]에서 역분화 및 세포군의 전환 (cell lineage conversion)^- 족진할 수 있다. The results presented in this study are the first description of a chemical-based protocol that mimics the early stages of limb regeneration in amphibians. Previously Experimental approaches have been developed to induce adult cells to form into pluripotent or stem cells. Examples include somatic cell nuclear transfer and transfection of transcription factors associated with omnipotence [45]. However, the above approaches did not utilize fully differentiated adult cells, which accounted for the majority of the eel hilar population in humans [46]. In addition, the above approaches currently face technical problems such as low efficiency. The chemical approach to obtaining pluripotency described in the present invention can de-differentiate and cell lineage conversion ^-in skeletal muscle tissue, which is considered to have a very high atrophy [22].
본 발명자들은 다핵 골격근 융합체의 세포화를 다능성의 유도에서 가장 중요한 첫 번째 단계로 고려하였는데, 이는 유사한 현상이 유미목 골격근 역분화 및 이후의 사지 절단에서 일어나기 때문이다 [47]. 골격근 세포화를 유도하는 다른 화합물에 대한 본 발명자들의 비교는 화학물질들의 효과가 조직에 따라 차이가 존재하기 때문에 유용하다 [22, 39, 40]. 근육모세포주의 분화에 대한 최적화된 프로토콜 (도 2a)을 이용하여 화학물질 미오세버린이 근육 융합체 (muscle syncytia)로부터 살아있는 단핵세포를 제조하는데 보다 더 효과적이라는 것을 발견하였다. 비록 본 연구가 항-유사분열 제제인 AraC 또는 5-FU 로 처리된 근육모세포로부터 유래된 근관에서 미오세버린 활성에 대한 최초 보고일 지라도, 미오세버린의 상대적으로 낮은 세포독성은 최초 보고 [14]에 기재되어 있었다- 미오세버린이 처리된 근육 융합체로부터 유래된 세포들은 여전히 불웅성을 가지는 것으로 확인되었다 (도 2b 및 도 2c). 상술한 발견은 Duckmanton 등 [2이에 의한 보고와 일치하며 미오세버린 활성에 대한 최초 보고들 [14, 16]과 대조적인데 , 이는 아마도 세포화의 유도 전에 높은 순도의 근관을 획득할 필요성이 있음을 의미하는 것이다. 사이클린- 의존성 키나제 억제제 (CDKN)인 p21 의 넉-다운은 세포화된 근관에서 증식 반웅을 유도하였으며 (도 2b-2d), 이는 p21 이 골격근에서 최종 분화의 핵심 조절자라는 최근 발견 [24]을 다시 한번 확인하는 것이다. 미오세버린 및 p21 넉-다운의 조합은 근섬유 세포화 및 세포주기 재- 진입 [21, 41] 같은 유미목 양서류에서 부속기관 재생 (appendage regeneration)의 중요한 초기 단계를 모방하도록 포유동물 근육을 유도한다. 유미목 양서류에서 부속기관 재생에 대한 최근 연구들은 근육 세포물이 이후에 신규한 근육 조직으로 재 -분화시키는 전구세포들의 일군을 형성한다는 것을 나타낸다. 즉, 재생하는 사지에서 다른 조직 형태들의 어떠한 기여도 있지 않다는 것이다 [41]. 본 연구는 작은 분자들의 처리에 의해 얻어진 근육 세포물도 근관 (muscle myotubes)으로 재-분화할 수 있다는 것을 확인하였다. 재 -분화 상에 리버신 처리의 억제 효과는 근육형성 조절인자 발현의 하향-조절과 연관될 수 있다 [42]. 따라서, 본 발명자들은 근육형성 조절인자 발현 상에 p21 넉-다운의 기능 (action)이 이후 조사될 홍미로운 분야라고 생각하는데, 이는 p21 넉-다운이 재 -분화 상에 리버신의 억제 효과를 역전시켰기 때문이다 (도 3b 및 도 3c). 더욱이, p21 넉—다운 및 리버신으로 처리된 근육 세포물은 다양한 운명을 가질 수 있는 전구세포들의 형성을 유도하였다 (도 4 및 도 5). 또한, 이와 유사한 상황이 진피-유래된 섬유아세포가. 연골, 결합조직 및 힘줄 (tendons)을 형성하는 전구세포들의 형성을 유도하는 것으로 알려진 유미목 부속기관 재생 (regenerate)에서 일어난다. 따라서, 본 연구에서 기재된 화학적 접근방법은 포유동물의 성체 골격근이 부속기관 재생의 자연적인 예에서 발견되는 것보다 더욱 우수한 정도의 다능성을 가지도록 한다. 본 발명의 화학적 접근방법의 개략적인 도면이 도 6 에 제시되어 있다. We considered the cellularization of the multinucleated skeletal muscle fusion as the first and most important step in the induction of pluripotency, since similar phenomena occur in chylotomy skeletal muscle dedifferentiation and subsequent limb cleavage [47]. Our comparisons to other compounds that induce skeletal muscle cellization are useful because the effects of chemicals vary from tissue to tissue [22, 39, 40]. An optimized protocol for differentiation of myoblasts (FIG. 2A) was used to find that the chemical myosevers were more effective at producing live mononuclear cells from muscle syncytia. Although this study was the first report of myoseberge activity in myotubes derived from myoblasts treated with anti-mitotic agents AraC or 5-FU, the relatively low cytotoxicity of myoseberge was reported [14]. ] Cells derived from myosin-treated muscle fusions were still found to have atrophy (FIGS. 2B and 2C). The above findings are consistent with the reports by Duckmanton et al. [2] and in contrast to the initial reports of myosebergine activity [14, 16], suggesting that there is a need to obtain high-purity root canals before induction of cellularization. It means. Knock-down of p21, a cyclin-dependent kinase inhibitor (CDKN), induced proliferative reaction in the cellized root canal (FIGS. 2B-2D), which again reiterated the recent finding that p21 is a key regulator of final differentiation in skeletal muscle [24]. Check it once. Combination of myoserine and p21 knock-down resulted in myofiber cellular and cell cycle re- Mammalian muscles are induced to mimic the critical early stages of appendage regeneration in Rhesus amphibians such as entry [21, 41]. Recent studies on adnexal regeneration in Rhesus amphibians show that muscle cells form a group of progenitor cells that later re-differentiate into new muscle tissue. In other words, there is no contribution of other tissue types in the regenerating limb [41]. The study confirmed that muscle cells obtained by treatment of small molecules can also be re-differentiated into muscle myotubes. The inhibitory effect of reversin treatment on re-differentiation may be associated with down-regulation of myogenic regulator expression [42]. Thus, we believe that the action of p21 knock-down on the expression of anabolic regulators is a faint area to be investigated later, which p21 knock-down reverses the inhibitory effect of reversin on re-differentiation. (FIG. 3B and FIG. 3C). Moreover, muscle cells treated with p21 knock-down and reversin induced the formation of progenitor cells that could have various fates (FIGS. 4 and 5). Similar situations are also found in dermal-derived fibroblasts. It occurs in the regenerate of chylopods known to induce the formation of progenitor cells that form cartilage, connective tissue and tendons. Thus, the chemical approach described in this study allows mammalian adult skeletal muscle to have a greater degree of pluripotency than those found in the natural example of adnexal regeneration. A schematic diagram of the chemical approach of the present invention is presented in FIG. 6.
지방세포 및 골세포로 재-분화하는 리버신-처리된 근육모세포의 능력이 이전에 보고되었다 [34, 42]. 하지만, 본 발명자들의 데이터는 상술한 발견이 주의있게 해석되어야 한다는 것을 나타낸다. 리버신의 이전 연구들은 계통-특이적 마커들의 발현 또는 지방세포 형성 과정 동안 지질 축적 같은 표현형 마커들을 확인하였다. 본 연구는 보다 강력하고 기능적인 어세이를 이용하여 지방세포 및 골세포 형성을 모니터링하고 알려진 세포 형태와 비교하고자 시도하였다. 예를 들어, 본 발명자들은 리버신—처리된 근육세포가 지질 축적을 한다는 것을 확인하였다. 하지만, 그들의 기능적 '실행 (performance)' 은 계통-특이적 지방세포와 비교하여 현저하게 감소된다 (도 4c 및 도 4e). 리버신-처리된 근육세포에서 지방형성 인자들의 칵테일을 변형시킴으로써 추가적으로 지방세포형성 과정을 강제 ( 'push' )하는 것이 가능할 수 있는데, 예를 들어 핵실 지방형성 인자인 퍼록시좀 증식인자-활성화된 수용체 를 활성화시키는 티아졸리딘에디온 (triazolidinediones)를 처리하는 것이다 [48]. The ability of reversin-treated myoblasts to re-differentiate into adipocytes and osteocytes has been previously reported [34, 42]. However, our data indicate that the above findings should be interpreted with caution. Previous studies of reversin identified phenotypic markers such as expression of lineage-specific markers or lipid accumulation during adipocyte formation. This study attempted to monitor adipocyte and bone cell formation and compare them with known cell types using more powerful and functional assays. For example, we have found that reversin-treated muscle cells accumulate lipids. However, their functional 'performance' is significantly reduced compared to lineage-specific adipocytes (FIGS. 4C and 4E). In reversin-treated muscle cells It may be possible to additionally 'push' the adipocyte-forming process by modifying the cocktail of lipogenic factors, e.g. to thiazolidine, which activates the nuclear chamber adipogenic factor, peroxysome growth factor-activated receptor. Treatment of triazolidinediones [48].
분화된 근육으로부터 다능성 세포를 생산하는 본 발명의 화학적 방법 (chemically defined method)은 향후 연구에서 수많은 실행가능한 분야를 가진다. 예를 들어, 근육모세포는 뉴런 분화 배지에서 리버신 처리 및 배양함으로써 신경형성 세포를 형성할 수 있다는 것이 보고되었다 [43]. 중배엽 계통 세포 (예컨대, 지방세포 및 골세포)으로부터 신경외배엽 계통 세포로의 유사한 '점프 (jump)' 가 세포화된 근섬유에서 가능할 수 았다. 선택적으로,…환자-특어적 다능성 세포를 제조하기 위한 시도로 근육생검이 본 연구에 기재된 화학물질-기반된 프로토콜에 따라 실시될 수 있다. 예를 들어, 국소 마취 하에서 환자로부터 외측광근 골격근 스트립이 얻어질 수 있다 [44L 골격근은 본 발명의 접근방법을 위한 아주 매력적인 조직 형태인데, 그 이유는 잔여 근육 전구세포들 (muscle precursor eel Is)을 활성화시킴으로써 , 잃어버린 조질을 재생할 수 있기 때문이다 [44]. 더 나아가, 본 연구에 기재된 화학적 접근방법은 유미목 양서류의 부속기관 재생의 초기 이벤트를 모방하며, 염증의 징후 및 상처봉합 전에 기능성 전구세포들을 얻기 위한 시도에서 상처 치유의 포유동물 모델들에 본 발명의 방법을 적용하는 것이 보다 바람직할 수 있다. 따라서, 본 연구의 결과들은 포유동물에서 사지 재생을 유도할 수 있는 방법의 개발과 밀접하게 연관되어 있으며 불웅성, 완전 분화된 포유동물 조직으로부터 유도된 전능성 줄기세포의 개발을 위한 미래의 접근방법을 제시한다. 이상으로 본 발명의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서 이러한 구체적인 기술은 단지 바람직한 구현 예일 뿐이며, 이에 본 발명의 범위가 제한되는 것이 아닌 점은 명백하다. 따라서, 본 발명 의 실질적인 범위는 첨부된 청구항과 그의 등가물에 의하여 정의된다고 할 것이다. 참고문헌 The chemically defined method of the present invention for producing pluripotent cells from differentiated muscle has numerous viable fields for future research. For example, it has been reported that myoblasts can form neuronal cells by reversin treatment and culture in neuronal differentiation medium [43]. Similar 'jumps' from mesodermal lineage cells (eg, adipocytes and bone cells) to neuroectodermal lineage cells could be possible in cellized muscle fibers. Optionally… Muscle biopsies can be performed according to the chemical-based protocols described in this study in an attempt to produce patient-specific pluripotent cells. For example, lateral myoskeletal muscle strips can be obtained from a patient under local anesthesia. [44L skeletal muscle is a very attractive tissue form for the approach of the present invention because of the residual muscle precursor eel Is This is because by activating this function, the lost temper can be reproduced [44]. Furthermore, the chemical approach described in this study mimics the early events of adnexal regeneration of Rhesus amphibians, and demonstrates the present invention in mammalian models of wound healing in attempts to obtain functional progenitor cells prior to signs of inflammation and wound closure. It may be more desirable to apply the method. Thus, the results of this study are closely related to the development of a method for inducing limb regeneration in mammals and suggest a future approach for the development of omnipotent stem cells derived from afferent, fully differentiated mammalian tissue. present. Having described the specific part of the present invention in detail, it is apparent to those skilled in the art that such a specific technology is only a preferred embodiment, and the scope of the present invention is not limited thereto. Therefore, the substantial scope of the present invention will be defined by the appended claims and equivalents thereof. references
[I] Graf i G. The complexity of cellular dedifferentiation: impl i cat ions for regenerative medicine. Trends in Biotechnology 2009;27:329-332.  [I] Graf i G. The complexity of cellular dedifferentiation: impl i cat ions for regenerative medicine. Trends in Biotechnology 2009; 27: 329-332.
[2]Tsonis PA. Regeneration in Vertebrates. Developmental Biology [2] Tsonis PA. Regeneration in Vertebrates. Developmental Biology
2000;221:273-284. 2000; 221: 273-284.
[3]Luc B. Transdi f f erent i at ion versus stem cell hypothesis for the regeneration of islet betaᅳ cells in the pancreas . Microscopy Research and Technique 1998 ;43 :332ᅳ 336.  [3] Luc B. Transdi f f erent i at ion versus stem cell hypothesis for the regeneration of islet beta ᅳ cells in the pancreas. Microscopy Research and Technique 1998; 43: 332 # 336.
[4] Christina M0H, Margaret WE, Ellen AGC. Reorganization of the ependyma during axolot 1 spinal cord regeneration: Changes in intermediate filament and f ibronect in expression. Developmental Dynamics 1992 ;193 :103-115.  [4] Christina M0H, Margaret WE, Ellen AGC. Reorganization of the ependyma during axolot 1 spinal cord regeneration: Changes in intermediate filament and f ibronect in expression. Developmental Dynamics 1992; 193: 103-115.
[5]Tsonis PA. Regeneration of the vertebrate lens and other eye structures . London.: Nature Pub 1. [www.els.net] , 1999.  [5] Tsonis PA. Regeneration of the vertebrate lens and other eye structures. London .: Nature Pub 1. [www.els.net], 1999.
[6]Tsonis PA. Regeneration of the lens in amphibia. . Heidelberg-' Springerᅳ Ver lag, 2000. [6] Tsonis PA. Regeneration of the lens in amphibia. . Heidelberg- ' Springer ' Ver lag, 2000.
[7]Tsonis PA, . Limb Regeneration, Cambridge, UK: Cambridge Univ. Press, 1996.  [7] Tsonis PA,. Limb Regeneration, Cambridge, UK: Cambridge Univ. Press, 1996.
[8]Takebe I. Regeneration of whole plants from isolated mesophyl 1 protoplasts of tobacco. Naturwissenschaf ten 1971 ;58 :318一 320.  [8] Takebe I. Regeneration of whole plants from isolated mesophyl 1 protoplasts of tobacco. Naturwissenschaf ten 1971; 58: 318 I 320.
[9]Mart in AB, Schultz PG. Opportunities at the interface of chemistry and biology. Trends in Cell Biology 1999; 9: M24-M28.  [9] Mart in AB, Schultz PG. Opportunities at the interface of chemistry and biology. Trends in Cell Biology 1999; 9: M24-M28.
[10]Williams D, Jung D-W, Khersonsky SM, Heidary N, Chang Y_T, Or 1 ow SJ . Identification of Compounds that Bind Mitochondrial FIFO ATPase by Screening a Triazine Library for Correction of Albinism. Chemistry & Biology 2004; 11: 1251-1259.  [10] Williams D, Jung D-W, Khersonsky SM, Heidary N, Chang Y_T, Or 1 ow SJ. Identification of Compounds that Bind Mitochondrial FIFO ATPase by Screening a Triazine Library for Correction of Albinism. Chemistry & Biology 2004; 11: 1251-1259.
[II] Darren RW, Sung-Kyun K, Sungj in P, Myung-Ryul L, Injae S. An Apopt os is- Inducing Small Molecule That Binds to Heat Shock Protein 7013. Angewandte Chemie International Edition 2008; 47: 7466-7469. 30 [II] Darren RW, Sung-Kyun K, Sungj in P, Myung-Ryul L, Injae S. An Apopt os is- Inducing Small Molecule That Binds to Heat Shock Protein 7013. Angewandte Chemie International Edition 2008; 47 : 7466-7469 . 30
2^  2 ^
20  20
Figure imgf000039_0001
Figure imgf000039_0001
1010
,¾gpg,y,,, Dn Ja sfim Ki CaG Kan Khesosa.2m Yirini P Mrnk confined to GO but can enter Gl upon growth factor stimulation. Cell Growth Differ 1996;7: 1039-1050. , ¾gpg , y ,,, Dn Ja s fi m K i CaG Kan Khesosa.2m Y i r i n i P Mrnk confined to GO but can enter Gl upon growth factor stimulation. Cell Growth Differ 1996; 7: 1039-1050.
[23] Myers TK, Andreuzza SE, Franklin DS. pl8INK4c and p27KIPl are required for cell cycle arrest of differentiated myotubes . Experimental Cell Research 2004; 300: 365-378.  [23] Myers TK, Andreuzza SE, Franklin DS. pl8INK4c and p27KIPl are required for cell cycle arrest of differentiated myotubes. Experimental Cell Research 2004; 300: 365-378.
[24]Pajalunga D, Mazzol A, Salzano AM, Biferi MG, De Luc a G, Crescenzi M. Critical requirement for cell cycle inhibitors in sustaining nonproliferative states. J Cell Biol 2007; 176 :80그 818.  [24] Pajalunga D, Mazzol A, Salzano AM, Biferi MG, De Luc a G, Crescenzi M. Critical requirement for cell cycle inhibitors in sustaining nonproliferative states. J Cell Biol 2007; 176: 80 He 818.
[25]Halevy 0, Nov itch BG, Spicer DB, Skapek SX, Rhee J, Hannon GJ , et al . Correlation of terminal cell cycle arrest of skeletal muscle with induction of p21 by MyoD. Science 1995 ;267: 1018-1021.  [25] Halevy 0, Nov itch BG, Spicer DB, Skapek SX, Rhee J, Hannon GJ, et al. Correlation of terminal cell cycle arrest of skeletal muscle with induction of p21 by MyoD. Science 1995; 267: 1018-1021.
[26]Thelen MH, Simonides WS, van Hardeveld C. Electrical stimulation of C2C12 myotubes induces contract ions and represses thyroid-hormone—dependent transcript ion of the fast-type sarco lasmic- reticulum Ca2+-ATPase gene. Biochem J 1997 ;321 ( Pt 3) :845-848.  [26] Thelen MH, Simonides WS, van Hardeveld C. Electrical stimulation of C2C12 myotubes induces contract ions and represses thyroid-hormone—dependent transcript ion of the fast-type sarco lasmic- reticulum Ca2 + -ATPase gene. Biochem J 1997; 321 (Pt 3): 845-848.
[27] Im YS, Shin HK, Kim HR, Jeong SH, Kim SR, Kim YM, et al. Enhanced cytotoxicity of 5-FU by bFGF through up-regulation of uridine phosphorylase 1. Mol Cells 2009 ;28: 119-124.  [27] Im YS, Shin HK, Kim HR, Jeong SH, Kim SR, Kim YM, et al. Enhanced cytotoxicity of 5-FU by bFGF through up-regulation of uridine phosphorylase 1.Mol Cells 2009; 28 : 119-124.
[28] Jones-Vi 1 leneuve EM , McBurney 丽, Rogers KA, Kalnins VI. Ret inoic acid induces embryonal carcinoma eel Is to differentiate into neurons and glial cells. J Cell Biol 1982;94:253-262.  [28] Jones-Vi 1 leneuve EM, McBurney Li, Rogers KA, Kalnins VI. Ret inoic acid induces embryonal carcinoma eel Is to differentiate into neurons and glial cells. J Cell Biol 1982; 94: 253-262.
[29] van der Put ten HH, Joosten BJ, Klaren PH, Everts ME. Uptake of tri-iodothyronine and thyro ine in myoblasts and myotubes of the embryonic heart cell line H9c2(2-1). J Endocrinol 2002; 175: 587-596.  [29] van der Put ten HH, Joosten BJ, Klaren PH, Everts ME. Uptake of tri-iodothyronine and thyro ine in myoblasts and myotubes of the embryonic heart cell line H9c2 (2-1). J Endocrinol 2002; 175: 587-596.
[30]Arias E, Ales E, Gab i lan NH, Cano-Abad MF, Villarroya M, [30] Arias E, Ales E, Gab i lan NH, Cano-Abad MF, Villarroya M,
Garcia AG, et al. Gal ant amine prevents apoptosis induced by beta- amyloid and thapsigargin: involvement of nicotinic acetylcholine receptors . Neuropharmacology 2004 ;46: 103-114. Garcia AG, et al. Gal ant amine prevents apoptosis induced by beta- amyloid and thapsigargin: involvement of nicotinic acetylcholine receptors. Neuropharmacology 2004; 46: 103-114.
[31]Sze Wan S, Mei Kuen T, Pak Ham C, Miguel M, Dong Qing C, Kenneth KHL. Induct ion of growth arrest and poly comb gene expression by reversine al lows C2C12 eel Is to be reprogrammed to various differentiated cell types . PROTEOMICS 2007 ;7:4303-4316. [31] Sze Wan S, Mei Kuen T, Pak Ham C, Miguel M, Dong Qing C, Kenneth KHL. Induct ion of growth arrest and poly comb gene expression by reversine al lows C2C12 eel Is to be reprogrammed to various differentiated cell types. PROTEOMICS 2007; 7: 4303-4316.
[32]Nakai M, Sekiguchi F, Obata M, Ohtsiiki C, Adachi Y, Sakurai H, et al. Synthesis and insul in-mimet ic activities of metal com lexes with 3-hydroxypyr idine-2-carboxyl ic acid. J Inorg Biochem 2005;99:1275-1282.  [32] Nakai M, Sekiguchi F, Obata M, Ohtsiiki C, Adachi Y, Sakurai H, et al. Synthesis and insul in-mimet ic activities of metal com lexes with 3-hydroxypyr idine-2-carboxyl ic acid. J Inorg Biochem 2005; 99: 1275-1282.
[33]Gregory CA, Gunn WG, Peister A, Prockop DJ. An Alizarin red- based assay of mineralization by adherent cells in culture: compar i son with cetylpyr idinium chloride extract ion. Anal Biochem 2004; 329: 77-84.  [33] Gregory CA, Gunn WG, Peister A, Prockop DJ. An Alizarin red- based assay of mineralization by adherent cells in culture: compar i son with cetylpyr idinium chloride extract ion. Anal Biochem 2004; 329: 77-84.
[34] Chen S, Takanashi S, Zhang Q, Xiong W, Zhu S, Peters EC, et al. Reversine increases the plasticity of 1 ineage-committed mammal i an cells. Proceedings of the Nat ional Academy of Sciences 2007; 104: 10482- 10487.  [34] Chen S, Takanashi S, Zhang Q, Xiong W, Zhu S, Peters EC, et al. Reversine increases the plasticity of 1 ineage-committed mammal i an cells. Proceedings of the Nat ional Academy of Sciences 2007; 104: 10482-10487.
[35]Garcia de Herreros A, Birnbaum MJ . The acquisition of increased i nsulirr responsive hexose transport in 3T3—L1 adipocytes correlates with expression of a novel transporter gene. J Biol Chem 1989;264:19994-19999.  [35] Garcia de Herreros A, Birnbaum MJ. The acquisition of increased i nsulirr responsive hexose transport in 3T3—L1 adipocytes correlates with expression of a novel transporter gene. J Biol Chem 1989; 264: 19994-19999.
[36]Nishide M, Yoshikawa Y, Yoshikawa EU, Matsumoto K, Sakurai H Kaj iwara NM. Insul inomimet ic Zn( 11 ) complexes as evaluated by both glucose-uptake activity and inhibition of free fatty acids release ' in isolated rat adipocytes. Chem Pharm Bull (Tokyo) 2008;56:1181-1183. [36] Nishide M, Yoshikawa Y, Yoshikawa EU, Matsumoto K, Sakurai H Kaj iwara NM. Insul inomimet ic Zn (11) complexes as evaluated by both glucose-uptake activity and inhibition of free fatty acids release ' in isolated rat adipocytes. Chem Pharm Bull (Tokyo) 2008; 56: 1181-1183.
[37]Kawabe K, Yoshikawa Y, Adachi Y, Sakurai H. Possible mode of act ion for insul inomimet ic activity of vanadyl (IV) compounds in adipocytes. Life Sci 2006 ;78 :2860-2866.  [37] Kawabe K, Yoshikawa Y, Adachi Y, Sakurai H. Possible mode of act ion for insul inomimet ic activity of vanadyl (IV) compounds in adipocytes. Life Sci 2006; 78: 2860-2866.
[38]French DM, Kaul RJ, D'Souza AL, Crowley CW, Bao M, Frantz GD: et al . WISP-1 Is an Osteoblast ic Regulator Expressed During Skeletal Development and Fracture Repair. Am J Pathol 2004; 165: 855-867. [38] French DM, Kaul RJ, D'Souza AL, Crowley CW, Bao M, Frantz GD : et al. WISP-1 Is an Osteoblast ic Regulator Expressed During Skeletal Development and Fracture Repair. Am J Pathol 2004; 165: 855-867.
[39]Huby RDJ, Weiss A, Ley SC. Nocodazole Inhibits Signal Transduction by the T Cell Antigen Receptor . Journal of Biological Chemistry 1998 ;273: 12024-12031. [40]Ai H, Ralston' E, Lauritzen HP, Gal bo H, Ploug T. Disruption of microtubules in rat skeletal muscle does not inhibit insulinᅳ or contract ion-st imulated glucose transport . Am J Physiol Endocrinol Metab 2003; 285 :E836-844. [39] Hubby RDJ, Weiss A, Ley SC. Nocodazole Inhibits Signal Transduction by the T Cell Antigen Receptor. Journal of Biological Chemistry 1998; 273: 12024-12031. [40] Ai H, Ralston ' E, Lauritzen HP, Gal bo H, Ploug T. Disruption of microtubules in rat skeletal muscle does not inhibit insulin ᅳ or contract ion-st imulated glucose transport. Am J Physiol Endocrinol Metab 2003; 285: E836-844.
[41]Tweedel 1 KS. The urodele limb regeneration blastema: the cell potential . Sc i ent i f i cWor 1 dJourna 1; 10: 954-971.  [41] Tweedel 1 KS. The urodele limb regeneration blastema: the cell potential. Sc i ent i f i cWor 1 dJourna 1; 10: 954-971.
[42]Amabile G, D'Alise AM, Iovino M, Jones P, Santaguida S, Musacchio A, et al . The Aurora B kinase activity is required for the maintenance of the differentiated state of murine myoblasts. Cell Death Differ 2009;16:321-330.  [42] Amabile G, D'Alise AM, Iovino M, Jones P, Santaguida S, Musacchio A, et al. The Aurora B kinase activity is required for the maintenance of the differentiated state of murine myoblasts. Cell Death Differ 2009; 16: 321-330.
[43] Lee EK, Bae GU, You JS, Lee JC, Jeon YJ, Park JW, et al. Reversine increases the plasticity of lineage—committed eel Is toward neuroectodermal lineage. J Biol Chem 2009;284:2891-2901.  [43] Lee EK, Bae GU, You JS, Lee JC, Jeon YJ, Park JW, et al. Reversine increases the plasticity of lineage—committed eel Is toward neuroectodermal lineage. J Biol Chem 2009; 284: 2891-2901.
[44]Zierath JR, Krook A, Wal lberg-Henr iksson H. Insulin act ion and insulin resistance in human skeletal muscle. Diabetologia 2000 ;43 :821-835.  [44] Zierath JR, Krook A, Wal lberg-Henr iksson H. Insulin act ion and insulin resistance in human skeletal muscle. Diabetologia 2000; 43: 821-835.
[45] Hochedl inger , K. ; Plath, K. , Ep i genet ic reprogramming and induced plur i otency . Deve 1 opment 2009, 136, (4), 509-23.  [45] Hochedl inger, K.; Plath, K., Ep i genet ic reprogramming and induced plur i otency. Deve 1 opment 2009, 136, (4), 509-23.
[46] Mar i eb , E. K. Ή. , Human Anatomy & Physiology. 7th ed.; Pearson Benjamin Cummings: 2007.  [46] Mar i eb, E. K. VIII. , Human Anatomy & Physiology. 7th ed.'S Pearson Benjamin Cummings: 2007.
[47] Brockes , J. P.; Kumar , A. , Comparative aspects of animal regeneration. Annu Rev Cell Dev Biol 2008, 24, 525-49.  [47] Brockes, J. P .; Kumar, A., Comparative aspects of animal regeneration. Annu Rev Cell Dev Biol 2008, 24, 525-49.
[48] Braissant, 0.; Foufelle, F.; Scotto, C.; Dauca , M.; Wahli, W. , Differential expression of peroxisome prol i f erator-act i vated receptors (PPARs): tissue distribution of PPARᅳ alpha, -beta, and ᅳ gamma in the adult rat . Endocrinology 1996, 137, (1), 354-66.  [48] Braissant, 0 .; Foufelle, F .; Scotto, C .; Dauca, M .; Wahli, W., Differential expression of peroxisome prol if erator-act i vated receptors (PPARs): tissue distribution of PPAR 'alpha , -beta, and ᅳ gamma in the adult rat. Endocrinology 1996, 137, (1), 354-66.

Claims

【특허청구범위】 [Patent Claims]
【청구항 1】 [Claim 1]
다음의 단계를 포함하는 포유동물 분화세포로부터 다능성 세포 (mult i potent or pluri otent eel Is)의 화학적 제조방법 :  Chemical preparation of pluripotent cells (mult i potent or pluri otent eel Is) from mammalian differentiated cells comprising the following steps:
(a) 포유동물부터 분화세포를 수득하는 단계; 및  (a) obtaining differentiated cells from a mammal; And
(b) 상기 분화세포에 p21 발현의 하향 -조절 유도 후 역분화제 (dedifferentiation agent)를 처리하여 다능성 세포를 수득하는 단계.  (b) treating the differentiated cells with a dedifferentiation agent after down-regulation induction of p21 expression to obtain pluripotent cells.
【청구항 2】 [Claim 2]
제 1 항에 있어서, 상기 분화세포는 말기 분화세포 (terminally differentiated cell)인 것을 특징으로 하는 방법.  The method of claim 1, wherein the differentiated cells are terminally differentiated cells.
【청구항 3】 [Claim 3]
제 1 항에 있어서, 상기 분화세포는 골격근 세포, 심근 세포, 평활근 세포, 피부 세포, 연골 세포, 지방 세포 및 골세포로 구성된 군으로부터 선택되는 하나의 세포인 것을 특징으로 하는 방법.  The method of claim 1, wherein the differentiated cells are one cell selected from the group consisting of skeletal muscle cells, cardiomyocytes, smooth muscle cells, skin cells, chondrocytes, adipocytes and osteocytes.
【청구항 4】 [Claim 4]
제 1 항에 있어서, 상기 분화세포는 결합 조직, 신경 세포, 림프 세포, 맥관 세포, 신장 세포, 췌장 세포, 폐 세포, 요도 세포, 방광 세포, 위 세포, 간 세포, 소장 세포, 대장 세포 및 식도 세포로 구성된 군으로부터 선택되는 하나의 세포인 것을 특징으로 하는 방법.  The method of claim 1, wherein the differentiated cells are connective tissue, nerve cells, lymph cells, vasculature cells, kidney cells, pancreas cells, lung cells, urethral cells, bladder cells, gastric cells, liver cells, small intestine cells, colon cells and esophagus And one cell selected from the group consisting of cells.
【청구항 5】 [Claim 5]
제 1 항에 있어서, 상기 단계 (a)는 포유동물의 근육세포로부터 유래된 융합체 (syncytia)를 분절시키고 단핵세포 또는 세포물 (eel lulate)을 얻기 위하여 상기 분화된 융합체에 세포화 -유도 제제 (cellularization- inducing agent)를 처리하여 실시하는 것을 특징으로 하는 방법。 The method according to claim 1, wherein step (a) comprises the step of a cell-derived agent in the differentiated fusion to segment the syncytia derived from the muscle cells of the mammal and to obtain mononuclear cells or eel lulates. Method characterized in that carried out by processing the cellularization-inducing agent.
【청구항 6】 [Claim 6]
제 5 항에 있어서, 상기 세포화—유도 제제는 미오세버린 (myoseverin), 미오세버린 B, 콜치신 또는 노코다졸인 것을 특징으로 하는 방법.  6. The method of claim 5, wherein said cellularization-inducing agent is myoseverin, myoseverin B, colchicine or nocodazole.
【청구항 7】 [Claim 7]
제 6 항에 있어서, 상기 세포화 -유도 제제는 미오세버린인 것을 특징으로 하는 방법ᅳ  7. The method of claim 6, wherein said cellularization-inducing agent is myoseborn.
【청구항 8] [Claim 8]
제 1 항에 있어서, 상기 p21 발현의 하향-조절은 안티센스 을리고뉴클레오타이드, siRNA 또는 miRNA 를 이용하여 실시되는 것을 특징으로 하는 방법 . The method of claim 1, wherein down-regulation of p21 expression is performed using antisense oligonucleotides, siRNAs or miRNAs.
【청구항 9] [Claim 9]
제 1 항의 단계 (b)에 있어서, 상기 역분화제는 리버신 (reversine; The process of claim 1 wherein step (b), said derivatizer comprises reversine;
2- ( 4-mor pho 1 i noan i 1 i no ) -6~cyc 1 ohexy 1 am i nopur i ne ) ¾ 것을 특징으로 하는 방법. 2- (4-mor pho 1 i noan i 1 i no) -6 to cyc 1 ohexy 1 am i nopur i ne) ¾.
【청구항 10】 [Claim 10]
제 1 항에 있어서, 상기 p21 발현의 하향-조절은 세포화된 근관의 증식, 세포주기로의 재 -진입 (re-entry) 및 분화를 유도하는 것을 특징으로 하는 방법 .  The method of claim 1, wherein down-regulation of p21 expression induces proliferation of cellularized root canals, re-entry and differentiation into the cell cycle.
【청구항 11】 [Claim 11]
제 1 항에 있어서, 상기 다능성 세포는 근육형성 리니지 (myogenic lineage), 지방형성 리니지 (adipogenic lineage) 또는 골형성 리니지 (osteogenic lineage)으로 분화할 수 있는 것을 특징으로 하는 방법.  The method of claim 1, wherein the pluripotent cells are capable of differentiating into myogenic lineage, adipogenic lineage, or osteogenic lineage.
【청구항 12】 [Claim 12]
제 1 항에 있어서, 상기 방법은 단계 (b)의 역분화제 처리이후 분화 유도제를 단계 (b)에서 얻은 세포에 처리하여 단계 (a)의 분화세포와 동일 3 배엽 계통의 다른 세포 리니지 (cell lineage)로 분화시키는 단계 추가적으로 포함하는 것을 특징으로 하는 방법 . The method according to claim 1, wherein the method is followed by treatment of a differentiation inducing agent to the cells obtained in step (b) after treatment with the reverse differentiation agent of step (b), to be identical to the differentiated cells of step (a). 3 differentiating into other cell lineages of the germ line.
【청구항 13】 [Claim 13]
제 3 항에 있어서, 상기 분화세포는 골격근 세포인 것을 특징으로 하는 방법 .  The method of claim 3, wherein the differentiated cells are skeletal muscle cells.
【청구항 14] [Claim 14]
제 1 항의 방법에 따라 제조된 다능성 세포.  Pluripotent cells prepared according to the method of claim 1.
【청구항 15】 [Claim 15]
다음의 단계를 포함하는 포유동물 조직 또는 기관의 재생 (regeneration)방법:  Regeneration of a mammalian tissue or organ comprising the following steps:
(a) 포유동물부터 분화세포를 수득하는 단계 ;  (a) obtaining differentiated cells from a mammal;
(b) 상기 분화세포에 P21 하향—조절 유도 후 역분화제^6(^£^^11 1011 agent)를 처리하여 다능성 세포를 수득하는 단계; 및 (b) treating the differentiated cells with P 21 down-regulation induction and then treating with a differentiation agent ^ 6 (^ £ ^^ 11 1011 agent) to obtain pluripotent cells; And
(c) 상기 다능성 세포를 원하는 조직 또는 기관으로 분화시키는 단계.  (c) differentiating said pluripotent cells into a desired tissue or organ.
PCT/KR2011/008473 2010-11-08 2011-11-08 Chemical preparation method of skeletal muscle-derived multipotent cells and use thereof WO2012064090A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2010-0110219 2010-11-08
KR20100110219 2010-11-08

Publications (3)

Publication Number Publication Date
WO2012064090A2 true WO2012064090A2 (en) 2012-05-18
WO2012064090A3 WO2012064090A3 (en) 2012-07-19
WO2012064090A9 WO2012064090A9 (en) 2012-08-23

Family

ID=46051410

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2011/008473 WO2012064090A2 (en) 2010-11-08 2011-11-08 Chemical preparation method of skeletal muscle-derived multipotent cells and use thereof

Country Status (2)

Country Link
KR (1) KR101352641B1 (en)
WO (1) WO2012064090A2 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9517779D0 (en) 1995-08-31 1995-11-01 Roslin Inst Edinburgh Biological manipulation

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHEN ET AL.: 'Dedifferentiation of lineage-committed cells by a small molecule' JOURNAL OF THE AMERICAN CHEMICAL SOCIETY vol. 126, 19 December 2003, pages 410 - 411 *
JUNG ET AL.: 'Novel chemically defined approach to produce multipotent cells from terminally differentiated tissue syncytia' ACS CHEMICAL BIOLOGY vol. 6, 15 February 2011, pages 553 - 562 *
LEE ET AL.: 'Reversine increases the plasticity of lineage-committed cells toward neuroectodermal lineage' THE JOURNAL OF BIOLOGICAL CHEMISTRY vol. 284, no. 5, 30 January 2009, pages 2891 - 2901 *
SHAN ET AL.: 'Induction of growth arrest and polycomb gene expression by reversine allows C2C12 cells to be reprogrammed to various differentiated cell types' PROTEOMICS vol. 7, 2007, pages 4303 - 4316 *

Also Published As

Publication number Publication date
WO2012064090A9 (en) 2012-08-23
KR101352641B1 (en) 2014-01-17
KR20120049155A (en) 2012-05-16
WO2012064090A3 (en) 2012-07-19

Similar Documents

Publication Publication Date Title
Guo et al. Cardiomyocyte differentiation of mesenchymal stem cells from bone marrow: new regulators and its implications
Anastasia et al. Reversine-treated fibroblasts acquire myogenic competence in vitro and in regenerating skeletal muscle
Hattori et al. Nongenetic method for purifying stem cell–derived cardiomyocytes
CA2540135C (en) Method of inducing the differentiation of stem cells into cardiomyocytes
JP2016127849A (en) Method and composition for preparing cardiac muscle cell from stem cell, and use of the same
Sisakhtnezhad et al. Transdifferentiation: a cell and molecular reprogramming process
Bhagavati et al. Generation of skeletal muscle from transplanted embryonic stem cells in dystrophic mice
US10752884B2 (en) Method of inducing beta cells from urine-derived cells using small molecules
EP2925860B1 (en) Methods of differentiating stem cells by modulating mir-124
KR102150244B1 (en) CD49f promoting proliferation, multipotency and reprogramming of Adult Stem Cells via PI3K/AKT/GSK3 pathway
Müller et al. Ca2+ activated K channels-new tools to induce cardiac commitment from pluripotent stem cells in mice and men
Fang et al. Skeletal muscle regeneration via the chemical induction and expansion of myogenic stem cells in situ or in vitro
Mezentseva et al. The histone methyltransferase inhibitor BIX01294 enhances the cardiac potential of bone marrow cells
JP2018531597A (en) Method for differentiating induced pluripotent stem cells produced from adult endocardial stem cells into cardiovascular cells and uses thereof
Quattrocelli et al. Synthetic sulfonyl‐hydrazone‐1 positively regulates cardiomyogenic microRNA expression and cardiomyocyte differentiation of induced pluripotent stem cells
Alder et al. Cell cycles and in vitro transdifferentiation and regeneration of isolated, striated muscle of jellyfish
Abdel-Latif et al. TGF-β1 enhances cardiomyogenic differentiation of skeletal muscle-derived adult primitive cells
Tchao et al. Combined biophysical and soluble factor modulation induces cardiomyocyte differentiation from human muscle derived stem cells
KR101352641B1 (en) Chemically-Defined Methods for Preparing Multipotent Cells Derived from Terminally Differentiated Skeletal Muscle and Uses Thereof
Wegener et al. How to mend a broken heart: adult and induced pluripotent stem cell therapy for heart repair and regeneration
Asashima et al. Elucidation of the role of activin in organogenesis using a multiple organ induction system with amphibian and mouse undifferentiated cells in vitro
EP3478824A1 (en) Amplifying beta cell differentiation with small molecules bet (bromodomain and extraterminal family of bromodomain-containing proteins) inhibitors
US20200407687A1 (en) Mature Cardiomyocyte Compositions
CA3139072A1 (en) Cardiomyocyte compositions and use thereof
JP2013511275A (en) How to induce tissue regeneration

Legal Events

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

Ref document number: 11840302

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11840302

Country of ref document: EP

Kind code of ref document: A2