KR20170113733A - Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof - Google Patents

Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof Download PDF

Info

Publication number
KR20170113733A
KR20170113733A KR1020160035243A KR20160035243A KR20170113733A KR 20170113733 A KR20170113733 A KR 20170113733A KR 1020160035243 A KR1020160035243 A KR 1020160035243A KR 20160035243 A KR20160035243 A KR 20160035243A KR 20170113733 A KR20170113733 A KR 20170113733A
Authority
KR
South Korea
Prior art keywords
ganglioside
stem cells
mesenchymal stem
differentiation
cell
Prior art date
Application number
KR1020160035243A
Other languages
Korean (ko)
Other versions
KR101794503B1 (en
Inventor
유재성
김용일
고용곤
Original Assignee
주식회사 티제이씨라이프
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 주식회사 티제이씨라이프 filed Critical 주식회사 티제이씨라이프
Priority to KR1020160035243A priority Critical patent/KR101794503B1/en
Publication of KR20170113733A publication Critical patent/KR20170113733A/en
Application granted granted Critical
Publication of KR101794503B1 publication Critical patent/KR101794503B1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0655Chondrocytes; Cartilage
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • 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
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2200/00Function of food ingredients
    • A23V2200/30Foods, ingredients or supplements having a functional effect on health
    • A23V2200/306Foods, ingredients or supplements having a functional effect on health having an effect on bone mass, e.g. osteoporosis prevention
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/36Lipids
    • 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
    • C12N2506/1346Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Rheumatology (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Food Science & Technology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Nutrition Science (AREA)
  • Mycology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Immunology (AREA)
  • Polymers & Plastics (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a composition for inducing differentiation of mesenchymal stem cells into chondrocytes, comprising ganglioside GM3 as an active ingredient, a method for inducing differentiation of mesenchymal stem cells into chondrocytes using the same, Or improving pharmaceutical composition and health functional food composition. The use of the composition containing ganglioside GM3 according to the present invention is excellent in the effect of inducing differentiation from mesenchymal stem cells into chondrocytes, and thus can be usefully used as a therapeutic, preventive or ameliorating agent for cartilage diseases by promoting cartilage regeneration .

Description

Composition for inducing chondrocyte differentiation containing ganglioside GM3 and its use {Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof}

The present invention relates to a composition for inducing differentiation of mesenchymal stem cells into chondrocytes, comprising ganglioside GM3 as an active ingredient, a method for inducing differentiation of mesenchymal stem cells into chondrocytes using the same, Or improving pharmaceutical composition and health functional food composition.

Mesenchymal stem cells (MSCs), also referred to as multipotent stromal progenitor cells, have been implicated in the development of mesodermal systems such as chondrocytes, osteoblasts, and adipocytes And is a resource of regenerative medicine (Non-Patent Documents 1 and 2). Specifically, synovium-derived mesenchymal stem cells (SDMSCs) have self-renewal and multipotent ability with a common surface epitope (Non-Patent Document 3). In addition, SDMSCs have cartilage cell producing ability because the cells have specific characteristics such as UDPDG (uridine diphosphoglucose dehydrogenase) activity and expression of COMP (cartilage oligomeric matrix protein) and GAG (glycosaminoglycans) 4-7). Therefore, many researchers suggest that SDMSCs provide a new strategy for clinical applications such as cell-based cartilage regeneration.

Gangliosides are complex glycosphingolipids containing sialic acids and are a major component of the cytoplasmic cell wall (non-patent document 8). Different cell types have various forms of gangliosides that accompany various biological processes such as cell death, cell proliferation, differentiation, surface interactions, and transmembrane signals (Non-Patent Documents 9-13). Many studies have confirmed that the type of ganglioside and its expression level are developmentally regulated and cell type specific (Non-Patent Literature 14, 15). In addition, recent research results of the present inventors have confirmed that the expression of ganglioside is closely related to various stem cells in vitro (Non-Patent Document 16-20). Interestingly, ganglioside GM3 regulates tyrosine phosphorylation of various growth factor receptors (Non-Patent Document 21-23). In particular, ganglioside GM3 induced by TGF-? 1 regulates the epithelial-mesenchymal transition (non-patent document 24).

Transforming growth factor-beta (TGF-β) superfamily (TGF-β1, 2, and 3) proteins play an important role in proliferation (Non-Patent Document 25). In addition, TGF-beta effectively upregulates various molecules involved in pre-cartilage cell formation and condensation and thus effectively induces chondrocyte differentiation (Non-Patent Document 26-28). Signaling associated with TGF- [beta] is initially internalized through serine / threonine phosphorylation of the TGF-beta receptor (TGF-betaR) after TGF-beta-R1 and TGF-beta-R2 are paired on the cell surface 29). Phosphorylated TGF-? R directly activates downstream mediator receptor-activated SMAD containing SMAD2 and SMAD3 (Non-Patent Documents 29-32).

Therefore, the present inventors confirmed that human SDMSC (hSDMSCs) can be differentiated into chondrocytes and continued the study on the effect of ganglioside GM3 on chondrocyte differentiation of the cells, and found that ganglioside GM3 inhibits the differentiation induction effect of hSDMSCs into chondrocytes The present invention has been completed.

 Dominici, M., et al., Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8, 315, 2006.  Pittenger, M.F. , Etc., Multilineage potential of adult human mesenchymal stem cells. Science 284, 143, 1999.  Sakaguchi, Y. et al., Comparison of human stem cells derived from various mesenchymal tissues: superiority of synovium as a cell source. Arthritis Rheum 52, 2521, 2005.  Wilkinson, L. S. Comparison of surface fibroblastic cells in subcutaneous air pouch and synovial lining: differences in uridine diphosphoglucose dehydrogenase activity. Int J Exp Pathol 74, 113, 1993.  Recklies, A.D. Regulation of cartilage oligomeric matrix protein synthesis in human synovial cells and articular chondrocytes. Arthritis Rheum 41, 997, 1998.  Fife, R.S. Identification of link proteins in canine synovial cell cultures and canine articular cartilage. J Cell Biol 100, 1050, 1985.  Hamerman, D., et al., Glycosaminoglycans produced by human synovial cell cultures. Coll Relat Res 2, 313, 1982.  Hakomori, S. Bifunctional role of glycosphingolipids. Modulators for transmembrane signaling and mediators for cellular interactions. The Journal of Biological Chemistry 265, 18713, 1990.  Hakomori, S. et al., Signal transduction through glyco (sphingo) lipids. Introduction and recent studies on glyco (sphingo) lipid-enriched microdomains. Annals of the New York Academy of Sciences 845, 1, 1998.  Kwak, D.H. And regulatory roles of ganglioside GQ1b in neuronal cell differentiation of mouse embryonic stem cells. BMB reports 44, 799, 2011.  Ryu, J.S. Ganglioside GM1 influences the proliferation rate of mouse induced pluripotent stem cells. BMB reports 45, 713, 2012.  Hakomori, S. Glycosphingolipids in cellular interaction, differentiation, and oncogenesis. Annu Rev Biochem 50, 733, 1981.  Hakomori, S. Glycosylation defining cancer malignancy: new wine in an old bottle. Proceedings of the National Academy of Sciences of the United States of America 99, 10231, 2002.  Yu, R.K. Etc., Developmental changes in ganglioside composition and synthesis in embryonic rat brain. Journal of neurochemistry 50, 1825, 1988.  Yu, R.K. Development regulation of ganglioside metabolism. Progress in brain research 101, 31, 1994.  Ryu, J.S. Gangliosides are involved in neural differentiation of human dental pulp-derived stem cells. Biochemical and biophysical research communications 387, 266, 2009.  Jung, J.U. The role of glycosphingolipids in the proliferation and neural differentiation of mouse embryonic stem cells. Exp Mol Med 41, 935, 2009.  Yang, H.J. Inhibition of ganglioside GD1a synthesis suppresses the differentiation of human mesenchymal stem cells into osteoblasts. Development, growth & differentiation 53, 323, 2011.  Lee, D.H. Effects of daunorubicin on ganglioside expression and neuronal differentiation of mouse embryonic stem cells. Biochem Biophys Res Commun 362, 313, 2007.  Kwak, D.H. , Etc., and the changes of gangliosides during differentiation of embryonic and mesenchymal stem cells into neural cells. Experimental & molecular medicine 38, 668, 2006.  Chung, T.W. Ganglioside GM3 inhibits VEGF / VEGFR-2-mediated angiogenesis: direct interaction of GM3 with VEGFR-2. Glycobiology 19, 229, 2009.  Kabayama, K. et al., Dissociation of the insulin receptor and caveolin-1 complex by ganglioside GM3 in the state of insulin resistance. Proc Natl Acad Sci U SE 104, 13678, 2007.  Kawashima, N. et al., Tyrosine kinase activity of epidermal growth factor receptor is regulated by GM3 binding to carbohydrate to carbohydrate interactions. J Biol Chem 284, 6147, 2009.  Kim, S.J. , And Ganglioside GM3 participates in the TGF-beta1-induced epithelial-mesenchymal transition of human lens epithelial cells. Biochem J 449, 241, 2013.  Kim, Y.I. Overexpression of TGF-beta1 enhances chondrogenic differentiation and proliferation of human synovium-derived stem cells. Biochem Biophys Res Commun 450, 1593, 2014.  Hao, J., et al., Engineering osteogenesis and chondrogenesis with gene-enhanced therapeutic cells. Curr Opin Mol Ther 11, 404, 2009.  Park, J.S. Heparin-bound transforming growth factor-beta3 enhances neocartilage formation by rabbit mesenchymal stem cells. Transplantation 85, 589, 2008.  Chimal-Monroy, J., et al., Expression of N-cadherin, N-CAM, fibronectin and tenascin is stimulated by TGF-beta1, beta2, beta3 and beta5 during the formation of precartilage condensations. Int J Dev Biol 43, 59, 1999.  Heldin, C.H. TGF-beta signaling from cell membrane to nucleus through SMAD proteins. Nature 390, 465, 1997.  Saika, S., et al., Smad3 signaling is required for epithelial-mesenchymal transition of lens epithelium after injury. Am J Pathol 164, 651, 2004.  Derynck, R. et al., Smad-dependent and Smad-independent pathways in TGF-beta family signaling. Nature 425, 577, 2003.  Ten Dijke, P., et al., Regulation of cell proliferation by Smad proteins. J Cell Physiol 191, 1, 2002.

It is an object of the present invention to provide a composition for inducing differentiation from mesenchymal stem cells into cartilage cells, which comprises ganglioside GM3 as an active ingredient.

Another object of the present invention is to provide a method for inducing differentiation from mesenchymal stem cells into chondrocytes, comprising culturing mesenchymal stem cells in vitro in the presence of ganglioside GM3.

It is still another object of the present invention to provide a pharmaceutical composition for treating or preventing cartilage diseases containing ganglioside GM3 as an active ingredient.

Another object of the present invention is to provide a health functional food composition for improving or preventing cartilage disease containing ganglioside GM3 as an active ingredient.

It is still another object of the present invention to provide a method for treating or preventing cartilage diseases, which comprises administering a composition containing glycoside GM3 as an active ingredient to a subject in need thereof.

One embodiment of the present invention provides a composition for inducing differentiation from mesenchymal stem cells into chondrocytes, comprising ganglioside GM3 as an active ingredient.

As used herein, the term "ganglioside " is a type of glycosphingolipid, which is an amphoteric substance composed of a hydrophilic sugar moiety and a hydrophobic ceramide, N-acetylneuraminic acid (sialic acid). Gangliosides of up to 100 kinds are now known as a combination of the number of sialic acids and their binding sites, with the diversity of neutral skeletal oligosaccharides except sialic acid. M, D, and T are mono, di and trisialo, and a, b, and c are bonded to a Gal at the reducing end of GM1, and G, G, And the number of sialic acids that are being used.

The ganglioside is located in an outer cell membrane that is in direct contact with the extracellular environment and regulates cell proliferation and differentiation. Regulation of cell differentiation depends on many extracellular and intracellular factor controls. Changes in the ganglioside expression pattern are observed during cell differentiation and in response to exposure to some cytokines and growth factors. Exogenous gangliosides have been shown to respond to exposure to several growth factors and to produce platelet-derived growth factor (PDGF) receptors, epidermal growth factor (EGF) receptors, insulin-like growth factor (IGF) nerve growth factor) regulates the function of the growth factor receptor such as the receptor.

As used herein, the term "ganglioside GM3" is named II3NeuAc-LacCer according to the IUPAC-IUB nomenclature and may be represented, for example,

[Chemical Formula 1]

Figure pat00001

The ganglioside GM3 can be obtained by a method generally known in the art or commercially available.

The present invention is characterized in that ganglioside GM3 has a function of promoting the induction of differentiation from mesenchymal stem cells into cartilage cells.

The ganglioside GM3 may be treated with a concentration of 0.1 to 1000 μM and may be treated with a concentration of 0.1 to 100 μM, more specifically a concentration of 0.1 to 10 μM, more specifically a concentration of 1 to 10 μM The mesenchymal stem cells can be treated with mesenchymal stem cells at a concentration of, for example, 2 to 5 [mu] M, but the present invention is not limited thereto. Concentrations having an inducing effect can be selected by an ordinary person skilled in the art.

As used herein, the term "active ingredient" refers to a component that exerts the effects of a drug or quasi-drug. In the present invention, it refers to a component that exerts a chondrocyte differentiation inducing effect.

Mesenchymal Stem Cells (MSCs) are one of stem cells collected from bone marrow and umbilical cord blood, and it is known that there are about 1 million stem cells in the body. Mesenchymal stem cells (MSCs) differentiate into mesenchymal cells such as chondrocytes, osteoblasts and adipocytes, thus providing cellular resources for cartilage regeneration. In addition, MSCs are easy to separate without significant donor-site morbidity and are easy to cultivate in vitro . MSCs are known to secrete a variety of growth factors, and some growth factors regulate immune function control activity.

The mesenchymal stem cells may be mesenchymal stem cells derived from a variety of adult tissues such as synovium-derived, bone marrow-derived, umbilical cord blood-derived, other placenta, muscle, fat and nerve tissue, Stem cells, but are not necessarily limited thereto. The term " synovium "refers to a membrane wrapping the inside of a joint pocket. When mesenchymal stem cells derived from synovium-derived mesenchymal stem cells (SDMSCs) are used as the mesenchymal stem cells, they have unique characteristics such as UDPDG activity and COMP and GAG expression as compared with other tissue-derived MSCs, The chondrocyte differentiation effect can be excellent.

The chondrocyte (cartilage cell) refers to a cell constituting cartilage tissue.

As used herein, the term "inducing differentiation from mesenchymal stem cells into chondrocytes" may mean inducing mesenchymal stem cells to differentiate into chondrocytes.

Accordingly, the composition for inducing differentiation of mesenchymal stem cells into chondrocytes according to the above embodiment can be used as a composition for regenerating cartilage tissue. The above-mentioned "cartilage tissue regeneration" may mean regenerating damaged cartilage tissue or inducing generation of insufficient cartilage tissue to regenerate cartilage tissue.

Another embodiment of the present invention provides a method for inducing differentiation from mesenchymal stem cells into chondrocytes, comprising culturing mesenchymal stem cells in vitro in the presence of ganglioside GM3.

The ganglioside GM3 may be treated with a concentration of 0.1 to 1000 μM and may be treated with a concentration of 0.1 to 100 μM, more specifically a concentration of 0.1 to 10 μM, more specifically a concentration of 1 to 10 μM The mesenchymal stem cells can be treated with mesenchymal stem cells at a concentration of, for example, 2 to 5 [mu] M, but the present invention is not limited thereto. Concentrations having an inducing effect can be selected by an ordinary person skilled in the art.

The mesenchymal stem cells may be mesenchymal stem cells derived from a variety of adult tissues such as synovium-derived, bone marrow-derived, umbilical cord blood-derived, other placenta, muscle, fat and nerve tissue, Stem cells, but are not necessarily limited thereto.

The term "in vitro" refers to cultivation in vitro , not in vivo , and may mean culturing cells isolated from a living body.

In addition to the ganglioside GM3, it may further include a chondrocyte differentiation inducer, for example, an insulin-transferrin-selenium (ITS), ascorbate-2-phosphate, dexamethasone, TGF-beta (Transforming Growth Factor), and the like, but the present invention is not limited thereto.

The chondrocyte induced differentiation from mesenchymal stem cells by the above method can be used as a cell therapy agent for treating cartilage diseases. Herein, cartilage diseases that can be treated using cartilage cells include, for example, osteoarthritis, degenerative arthropathy, cartilage dysplasia, degenerative arthritis, rheumatoid arthritis, osteomalacia, fibrous osteitis, and intractable bone disease And can be used for treatment of various diseases caused by abnormal annihilation of chondrocytes or damage of cartilage tissue.

Another embodiment of the present invention provides a pharmaceutical composition for treating or preventing cartilage disease comprising ganglioside GM3 as an active ingredient.

The cartilage disease may be cartilage disease caused by abnormal annihilation of cartilage cells or damage of cartilage tissue, and may be caused by, for example, osteoarthritis, degenerative arthropathy, cartilage dysgenesis, degenerative arthritis, rheumatoid arthritis, osteomalacia, Although bone disease is a representative example, it is not limited thereto.

As used herein, the term "treatment" refers to any action in which administration of a composition containing ganglioside GM3 improves or alleviates symptoms of cartilage disease. Thus, treatment of the cartilage disease may mean that the damage of cartilage tissue is treated by regenerating cartilage damaged by the composition.

As used herein, the term "prevention" means any action that inhibits or delays cartilage disease by administration of a composition containing the ganglioside GM3.

In the above pharmaceutical composition, naringenin and hesperetin are preferably used as such, but derivatives thereof may be prepared and used in consideration of solubility and stability characteristics.

The pharmaceutical composition according to the above embodiments may be various oral or parenteral formulations. In the case of formulation, a diluent or excipient such as a filler, an extender, a binder, a wetting agent, a disintegrant, or a surfactant is usually used. Solid formulations for oral administration include tablets, pills, powders, granules, capsules, and the like, which may contain one or more excipients such as starch, calcium carbonate, sucrose or lactose lactose, gelatin and the like. In addition to simple excipients, lubricants such as magnesium stearate, talc, and the like are also used. Liquid preparations for oral administration include suspensions, solutions, emulsions, syrups and the like. Various excipients such as wetting agents, sweeteners, fragrances, preservatives and the like may be included in addition to water and liquid paraffin, which are simple diluents commonly used. have. Formulations for parenteral administration include sterilized aqueous solutions, non-aqueous solutions, suspensions, emulsions, freeze-dried preparations, and suppositories.

Examples of the pharmaceutical composition according to the present invention include non-aqueous solutions, propylene glycol, polyethylene glycol, vegetable oils such as olive oil, injectable esters such as ethyl oleate, and the like. Examples of the suppository base include witepsol, macrogol, tween 61, cacao paper, laurin, glycerogelatin and the like.

Dosage forms of the pharmaceutical compositions according to the above embodiments may be used in the form of their pharmaceutically acceptable salts, and may be used alone or in combination with other pharmaceutically active compounds as well as in suitable aggregates. The salt is not particularly limited as long as it is pharmaceutically acceptable so long as it is pharmaceutically acceptable and includes, for example, hydrochloric acid, sulfuric acid, nitric acid, phosphoric acid, hydrofluoric acid, hydrobromic acid, formic acid acetic acid, tartaric acid, lactic acid, citric acid, fumaric acid, , Benzenesulfonic acid, toluenesulfonic acid, and naphthalenesulfonic acid.

The pharmaceutical composition according to the above embodiments may be administered parenterally or orally according to the purpose, and may be administered in one to several divided doses in an amount of 0.1 to 500 mg and 1 to 100 mg per kg of body weight per day . The dosage for a particular patient may vary depending on the patient's body weight, age, sex, health condition, diet, time of administration, administration method, excretion rate, severity of disease, and the like.

The pharmaceutical compositions according to the above embodiments may be formulated into oral preparations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups and aerosols, external preparations such as ointments and creams, suppositories and sterilized injection solutions And may be formulated in any form suitable for pharmaceutical preparations.

The pharmaceutical compositions according to the above embodiments may be administered to mammals such as rats, mice, livestock, and humans in various routes such as parenteral, oral, etc., and all manner of administration may be expected, Or by intravenous, intramuscular, subcutaneous, intramural or intracerebroventricular injection.

Another embodiment of the present invention provides a health functional food composition for improving or preventing cartilage disease containing ganglioside GM3 as an active ingredient.

As used herein, the term "improvement" means any action that improves or alters the symptoms of cartilage disease by administering a composition containing the ganglioside GM3.

As used herein, the term "health functional food" refers to a food prepared by processing a specific ingredient as a raw material for the purpose of health assisting or by extracting, concentrating, refining, or mixing a specific ingredient contained in a food raw material , The health functional food composition can perform functions related to prevention of cartilage disease and recovery of cartilage disease.

The health functional food may contain flavoring agents such as various nutrients, vitamins, minerals (electrolytes), synthetic flavors and natural flavors, coloring agents and thickening agents (cheese, chocolate etc.), pectic acid and its salts, alginic acid and its salts, Organic acids, protective colloid thickeners, pH adjusting agents, stabilizers, preservatives, glycerin, alcohols, carbonating agents used in carbonated drinks, and the like. It can also contain natural fruit juices and pulp for the production of fruit juices and vegetable drinks. These components may be used independently or in combination. The health functional food may be any one of meat, sausage, bread, chocolate, candy, snack, confectionery, pizza, ramen, gum, ice cream, soup, beverage, tea, functional water, drink, alcohol and vitamin .

In addition, the health functional food may further include food additives, and the suitability of the food functional food as a "food additive" is not limited to the corresponding items in general rules and general test methods approved by the Food and Drug Administration Shall be determined according to the relevant standards and standards.

Examples of the products that have been used in the above-mentioned "food additives" include natural products such as ketones, chemical products such as glycine, potassium citrate, nicotinic acid and cinnamic acid, sensory coloring matter, licorice extract, crystalline cellulose, high- - Mixed preparations such as a sodium glutamate preparation, a noodle-added alkaline agent, a preservative preparation, a tar coloring agent and the like.

At this time, the content of ganglioside GM3 added to the food during the production of the health functional food can be appropriately increased or decreased as needed. The ganglioside GM3 may be added in an amount of 0.1 wt% to 50 wt% to 100 wt% of the food.

Another embodiment of the present invention provides a method for treating or preventing cartilage disease comprising administering a composition containing ganglioside GM3 as an active ingredient to a subject in need thereof.

As used herein, the term "individual" refers to all animals including humans who have already developed or are capable of developing cartilage disease, and the composition of the present invention is effective for preventing and treating the disease by administration to a subject have.

The composition is preferably a pharmaceutical composition and can be administered in a therapeutically effective amount.

As used herein, the term "therapeutically effective amount" means an amount sufficient to treat a disease at a reasonable benefit / risk ratio applicable to medical treatment and the effective dosage level will vary depending on the species and severity, The activity of the drug, the sensitivity to the drug, the time of administration, the route of administration and the rate of release, the duration of the treatment, factors including co-administered drugs, and other factors well known in the medical arts. The composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, and may be administered sequentially or simultaneously with conventional therapeutic agents. And can be administered singly or multiply. It is important to take into account all of the above factors and to administer the amount in which the maximum effect can be obtained in a minimal amount without side effects, which can be easily determined by a person skilled in the art.

Previous studies have demonstrated the role of TGF-β in many cell fates such as maturation, cell proliferation and differentiation. Specifically, in chondrocyte differentiation, the TGF-β signal is transmitted to the nucleus through the SMAD pathway, which activates the Sox-9 transcription factor for cartilage gene expression, such as type II collagen and aggrecan. In the present invention, it was confirmed that treatment of ganglioside GM3 during chondrocyte cell differentiation activates Sox-9, type II collagen, agricans and COMP. Also, according to an embodiment of the present invention, ganglioside GM3 has been shown to modulate serine phosphorylation of TGF-s-R1 and -R2 and SMAD2 / 3 during chondrocyte cell differentiation.

In conclusion, it was confirmed that exogenous ganglioside GM3 did not affect cytotoxicity of hSDMSCs. However, exogenous ganglioside GM3 significantly increased the tissue weight of chondrocyte cell groups differentiated from the chondrocyte cell group differentiation, especially in the 5 [mu] M treated group, and the amount of toluidine blue was high, and aggrecan, Sox-9, 2 expression of cartilage cell specific marker genes such as collagen and COMP was increased and GAG accumulation was increased. In addition, exogenous ganglioside GM3 enhanced the TGF-beta signaling pathway through SMAD 2/3 during chondrocyte cell differentiation. Therefore, ganglioside GM3 is expected to be useful as a therapeutic agent for promoting cell-based articular cartilage regeneration in articular cartilage diseases.

The use of the composition containing ganglioside GM3 according to the present invention is excellent in the effect of inducing differentiation from mesenchymal stem cells into chondrocytes, and thus can be usefully used as a therapeutic, preventive or ameliorating agent for cartilage diseases by promoting cartilage regeneration .

Figure 1 shows the characteristics of human synovium-derived mesenchymal stem cells (hSDMSCs): (A) MSC negative surface proteins, CD4, CD34 and CD45 (upper), and positive surface proteins, CD44, CD73, CD90 and CD105 The results of FACS analysis of the expression of the expression of And (B) staining results of osteoblast differentiated from hSDMSCs (left: Alizarin red S staining) and adipocyte differentiated from hSDMSCs (right: Oil red O staining).
Figure 2 shows the effect of ganglioside GM3 on cell viability in hSDMSCs: hSDMSCs were treated with ganglioside GM3 (0, 1, 2, 5 and 10 [mu] M) for 7 days. Cell viability was calculated according to the formula [(OD value after treatment with ganglioside GM3) / OD value in the group treated with 0 [mu] M ganglioside GM3) x 100]. Values are expressed as mean ± SEM from 6 independent experiments.
Figure 3 shows the effect of ganglioside GM3 on cartilage cell population differentiation: hSDMSCs treated ganglioside GM3 (0, 1, 2, 5 and 10 [mu] M) for 21 days during chondrocyte cell population changes. (A) Analysis of the tissue weight of differentiated chondrocytes at day 21 after chondrocyte differentiation. Values are expressed as mean ± SEM from 6 independent experiments. ** p < 0.01 and *** p < 0.001 compared to GM3 treated group at 0 μM; And (B) Toluidine blue and H & E staining results to confirm the histological characteristics of the cell groups of all groups.
FIG. 4 is a graph showing the effect of ganglioside GM3 on the tissue weight of differentiation of chondrocytic cell group: (A) the result of analysis of the weight of the differentiated tissue of the cartilage cell group on the 7th day and (B) 14th day. Values are expressed as mean ± SEM from 6 independent experiments. * P < 0.05 and ** p < 0.01 compared to 0 μM GM3 treated group.
5 shows the effect of ganglioside GM3 on the histological analysis of differentiation of chondrocyte cell group. For histological analysis of differentiated chondrocytic cell groups on days 7 and 14, differentiated chondrocyte cell groups of all groups were treated with toluidine Blue (TB) and HE stained.
FIG. 6 is a graph showing a comparison of chondrocyte-specific marker expression in chondrocyte cell group differentiation: (A) Aggrecan, (B) Sox-9, (C ) Type 2 collagen and (D) cartilage oligomeric matrix protein (COMP) expression were compared by qPCR. mRNA expression levels were normalized to the always expressed gene GAPDH. Values are expressed as mean ± SEM from 3 independent experiments. * P < 0.05 and *** p < 0.001 as compared to 0 μM GM3 treated group.
FIG. 7 shows a comparison of chondrocyte-specific marker expression in differentiated chondrocyte cell groups: (A) aggrecan, (B) Sox- 9, (C) type 2 collagen and (D) cartilage oligomeric matrix protein (COMP) expression was compared by qPCR. mRNA expression levels were normalized to the always expressed gene GAPDH. Values are expressed as mean ± SEM from 3 independent experiments per week. * P <0.05, ** p <0.01 and *** p <0.001 compared with 0 μM GM3 treatment group.
FIG. 8 is a graph showing the results of biochemical analysis for evaluating the differentiation of chondrocyte cell group. After 21 days of chondrocyte cell differentiation, the cell group was digested with papain and S-GAG (sulfated glycosaminoglycans) Respectively. GAG content was normalized to the total DNA content of each sample. Values are expressed as mean ± SEM from 3 independent experiments. * P < 0.05 and *** p < 0.001 as compared to 0 μM GM3 treated group.
Fig. 9 is a diagram showing the results of biological analysis for evaluation of chondrocyte cell differentiation. After 7 and 14 days of chondrocyte cell differentiation, the cells were digested with papain and S-GAG (sulfated glycosaminoglycans) And the content thereof was measured. GAG content was normalized to the total DNA content of each sample. Values are expressed as mean ± SEM from 3 independent experiments per week. * P < 0.05, and *** p < 0.001 compared to GM3 treated group at 0 μM.
Figure 10 shows the effect of ganglioside GM3 on chondrocyte cell differentiation: hSDMSCs that underwent chondrocyte cell differentiation were treated with various concentrations of GM3 (0, 1, 2, 5, and 10 [mu] M) for 21 days. After SDS-PGE and Western blotting, serine (TGF-β-R1 and -R2) and phosphorylation of SMAD 2/3 were detected by immunoblotting using specific antibodies. β-actin was used as a control.
Figure 11 shows the effect of differentiating chondrocyte cell cultures of ganglioside GM3 on differentiation: hSDMSCs that underwent differentiation into chondrocyte cell groups were cultured at various concentrations of GM3 (0, 1, 2, 5, and 10 [mu] M) 14 days. After SDS-PGE and Western blotting, phosphorylation of serine (TGF-β-R1 and -R2), SMAD 2, and SMAD3 was detected by immunoblotting using specific antibodies. β-actin was used as a control.

Hereinafter, the present invention will be described in more detail in the following Examples. It should be noted, however, that the following examples are illustrative only and do not limit or limit the scope of the present invention. It will be understood by those of ordinary skill in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention as defined by the appended claims.

Example  One. hSDMSCs  Culture and on hSDMSCs  About Ganglioside  Evaluation of GM3 cytotoxic effect

1-1. hSDMSCs  Isolation and Culture

Primary cell lines were obtained from primary synovial membrane tissues isolated from human knees.

Specifically, human synovium-derived mesenchymal stem cells (hSDMSCs) derived from human synovium were obtained from the synovium of osteoarthritis patients according to the regulations of the Ethics Committee of Yonsei Sarang Hospital. Synovial tissue was chopped, digested with 0.25% trypsin EDTA (Hyclone), and incubated at 37 ° C for 30 minutes. After 0.25% trypsin EDTA was removed, the cells were treated with 0.3% collagenase (Worthington) in DMEM (high glucose Dulbecco's modified Eagle's medium; Hyclone) and cultured at 37 ° C for 90 minutes. Single cell suspensions were centrifuged and supernatant removed. Cells were resuspended in pre-warmed alpha-modified Eagle's medium (Hyclone) supplemented with 20% fetal bovine serum (HBS) and 1 x penicillin / streptomycin (Hyclone) I moved. When adherent cells proliferated and reached confluence, cells were trypsinized and transferred back to the dish for expansion, which was used as hSDMSCs.

1-2. Intermediate lobe  Identification of stem cells

In order to identify the hSDMSCs cells obtained in Example 1-1, FACS analysis and the ability to differentiate (osteocyte and adipocyte differentiation) were performed.

FACS analysis was performed as follows. After trypinization, the hSDMSCs of Example 1-1 above were resuspended at 1.0 x 10 cells / ml in ice cold PBS containing 0.5% BSA (bovine serum albumin). The cells were then incubated in ice for 20 minutes with PerCP-conjugated anti-human CD4, PE-conjugated anti-human CD34, APC-conjugated anti-human CD45, PE- CD40, FITC-conjugated anti-human CD73, PerCP-conjugated anti-human CD90, or PerCP-conjugated anti-human CD105 (BD Biosciences). Analysis was performed using CellQuest software (BD Biosciences) by flow cell counting (BD Biosciences).

The ability to differentiate into osteoblasts was confirmed as follows. the hSDMSCs in 24-well tissue culture plates 5 x 10 4 cells / well were seeded and grown for one day in a standard growth medium. The medium was replaced with an osteoblast-producing medium composed of 1-penicillin / streptomycin, 10% FBS, 100 nM dexamethasone, 10 mM? -Glycerophosphate (Sigma-aldrich) and 50 μM ascorbic acid in high- Lt; / RTI &gt; The medium was changed every 2 days. After differentiation, the cultured cells were fixed with 4% PFA (paraformaldehyde) and stained with 2% Alizarin red S pH 7.2 (Sigma-aldrich). Samples were observed through a light microscope (Leica, DMI 3000B).

The ability to differentiate into adipocytes was confirmed as follows. the hSDMSCs in 12-well tissue culture plates 5 x 10 3 cells / well were seeded and grown for one day in a standard growth medium. The medium was supplemented with 1 × penicillin / streptomycin, 10% FBS, 1 μM dexamethasone, 200 μM indomethacin (Sigma-aldrich), 0.5 mM IBMX (3-butyl-1-methylxanthine) mL insulin (GIBCO) and replaced with an adipocyte-derived medium consisting of complete high-glucose DMEM for 21 days. The medium was changed every 2 days. After differentiation, the cultured cells were fixed with 4% PFA (paraformaldehyde) and stained with 2% Oil O Red solution (Sigma-aldrich). Samples were observed through a light microscope (Leica, DMI 3000B).

As a result, it was confirmed that the cells obtained in Example 1-1 were mesenchymal stem cells (MSCs) (FIG. 1).

1-3. Ganglioside  Cytotoxicity assay of GM3

To assess ganglioside GM3 toxicity, hSDMSCs cells were incubated with various concentrations of ganglioside GM3.

Specifically, the cell viability assay was performed as follows. HSDMSCs cells of Example 1-1 were inoculated on a 24-well plate at a density of 1 x 10 4 cells / well. Cells were treated with GM3 (1, 2, 5, and 10 μM) (Matreya) and then cell proliferation was assessed by MTT (3- (4,5-dimethylthiazol-2-yl) -2,5-diphenyltetrazolium bromide) Based tests. Each well was incubated with MTT solution (Sigma-Aldrich) for 4 hours, and the absorbance of each well was measured at 590 nm using a spectrophotometer.

Experimental The statistical analysis was carried out in the following manner in all experiments related to the present invention. All data are expressed as mean ± S.E.M. Comparisons of multiple groups were performed by one-way ANOVA followed by Bonferroni post-test and compared in pairs. Differences were considered statistically significant at P <0.05. All data was analyzed using GraphPad Prism software, version 5.00 (Graph-Pad software).

As a result, the GM3 treatment group had no significant difference in the cell survival rate at the respective concentrations for 7 days from the GM3 treatment group of 0 μM. Therefore, it was found that ganglioside GM3 did not affect cytotoxicity of hSDMSCs (Fig. 2).

Example  2. Ganglioside  The induction effect of GM3 on differentiation of chondrocytic cell group

2-1. Tissue weighing of chondrocytes

An aggregates culture system was used to confirm the effect of ganglioside GM3 on chondrocyte differentiation of hSDMSCs. During chondrocyte cell differentiation, cell populations of all groups were weighed.

Specifically, chondrocyte cell population differentiation was carried out by the following method. The hSDMSCs in Example 1-1 in polypropylene tubes were inoculated with 1.0 x 10 6 cells / ml. The cells were centrifuged at 1,500 rpm for 5 minutes for use in an aggregate culture system. Cells were prepared and transfected with 1xITS (Insulin-Transferrin-Selenium; GIBCO), 50 mM ascorbate-2-phosphate, 100 nM dexamethasone (Sigma), 5 ng / And cultured at 37 占 폚 in chondrocyte-producing medium consisting of growth factor-beta (Prospec) and GM3 (or without GM3). Cells were formed as free-floating masses in the first 24-hour culture. The medium was replaced every 2 days for 21 days.

As a result, the tissue weights did not differ in the respective groups during the 7 days of chondrocyte cell differentiation. However, especially in the GM3 treated group of 2, 5 and 10 μM, the tissue weight increased on the 14th day after the differentiation of chondrocyte cell group. On the 21st day after chondrocyte cell differentiation, the mean values of the pellet weights in the 2 μM and 5 μM GM3-treated groups were 10.13 ± 0.37 mg and 11.37 ± 0.59 mg, respectively, which were significantly increased compared with the 0 μM GM3-treated group (Figs. 3A and 4).

Therefore, the induction of chondrocyte differentiation was promoted by treatment with ganglioside GM3, and in particular, it was confirmed that the effect of inducing chondrocyte differentiation in the GM3 treatment group of 2 and 5 μM was excellent.

2-2. Histological analysis of chondrocytes

Histological analysis was performed by staining with toluidine blue and H & E for each of the experimental groups of Example 2-1.

Specifically, histological analysis was performed as follows. Cells were fixed with 10% formaldehyde for 10 min and washed twice with PBS (phosphate buffered saline). Cells were placed in paraffin and cut into 5-μm sections. The sections were stained according to conventional protocols with toluidine blue (for matrix proteoglycan staining), H & E (haematoxylin and eosin) staining. Samples were observed through a light microscope (Leica, DMI 3000BI).

As a result, the GM3-treated group of 5 [mu] M had a considerable amount of toluidine blue (Fig. 3B and Fig. 5). Since the content of glycosaminoglycan in the cartilage tissue can be visually confirmed according to the toluidine blue staining intensity, it was confirmed that the GM3-treated group, especially 5 μM of the ganglioside GM3-treated group, Respectively.

2-3. Cartilage cell specific Marker  Gene expression measurement

Expression of chondrocyte-specific marker genes such as aggrecan, Sox-9, type 2 collagen and COMP was measured for each of the experimental groups of Example 2-1.

Specifically, the analysis of mRNA expression was performed in the following manner. Total RNA was prepared from samples using the Trizol Kit according to the manufacturer's instructions (Invitrogen). 2 μg of total RNA was reverse transcribed (RT) according to the manufacturer's instructions (Qiagen) using Omniscript RT kit and oligo dT. Real-time PCR was performed using an ABI PRISM 7500 sequence detection system (Applied Biosystems). The reaction was carried out in three independent experiments. GAPDH (glyceraldehyde 3-phosphate dehydrogenase) was used as a control. The primer sequences are shown in Table 1 below. Expression data were normalized to the geometric mean of the expression gene GAPDH at all time to control for changes in expression levels and analyzed using the 2 - ΔΔCT method.

gene Accession No. SEQ ID NO: primer  direction order Aggrecan
NM-001135.3
One Forward 5`- CTG AGT GAA ACC ACC TCT GCA TT -3`
2 Reverse 5`- GAC GCC TCG CCT TCT TGA - 3` SOX-9
NM_000346.3
3 Forward 5`- CCC CAA CAG ATC GCC TAC AG -3`
4 Reverse 5`- TCT GGT GGT CGG TGT AGT CGTA -3 ` Collagen type 2 NM_001844.4
5 Forward 5`- GGC AAT AGC AGG TTC ACG TACA -3`
6 Reverse 5`- CGA TAA CAG TCT TGC CCC ACT T -3` COMP
NM_000095.2
7 Forward 5`- CAA GAA GTC CTA TCG TTG GTT CCT -3
8 Reverse 5`- CTC AGG GCC CTC ATA GAA TCG -3` GAPHD
NM_002046.5
9 Forward 5`- GGC ATC CTG GGC TAC ACT GA -3 `
10 Reverse 5`- GAG GAG TGG GTG TCG CTG TT -3`

As a result, at day 21 after chondrocyte cell differentiation, chondrocyte-specific markers were highly expressed in 5 μM ganglioside GM3 (3.00 ± 0.02 fold compared to 0 μM GM3 treated group, 3.00 ± 0.02 fold, Sox-9, 2.85 占 .55 times; type 2 collagen, 2.55 占 .9 times; COMP, 5.92 占 .26 times) (Figs. 6 and 7).

2-4. Of GAGs  Accumulation analysis

In Example 2-1, the accumulation of GAGs (glycosaminoglycans) normalized to whole DNA in all groups of cells during chondrocyte differentiation was analyzed.

Specifically, the measurement of the GAG (glycosaminoglycan) content was carried out in the following manner. After 21 days of chondrocyte induction, all cell populations were stimulated with 125 μg / mL papain dissolved in PBE buffer (10 mM EDTA, 100 mM sodium phosphate, pH 6.5) containing 5 mM L-cysteine- Gt; 65 C &lt; / RTI &gt; per sample. The chondrocyte productivity was determined by measuring the production of S-GAG (sulfated glycosaminoglycans) according to the manufacturer's recommended protocol (Blyscan ™ Glycosaminoglycan Assay Kit) using the 1,9-dimethylmethylene blue (DMMB) test. Absorbance was measured at 656 nm using a microplate immunoreader (Sunrise ™, TECAN). To quantify the cell density, the amount of DNA in the papain digest was analyzed using the Quant-iT PicoGreens dsDNA Assay kit (Invitrogen) according to the manufacturer's recommended protocol. (Excitation at 485 nm; emission at 538 nm) using a CytoFluors Series 4000 (Applied Biosystems). The GAG content was normalized to the total DNA content.

As a result, the GAG / DNA ratio was 16.48 ± 1.45 μg / μg in the group treated with 5 μM ganglioside GM3 (0 μM, 3.69 ± 0.12 μg / μg; 1 μM, 5.05 ± 1.08 μg / μg; 2 μM, 4.93 ± 0.21 μg / μg; 10 μM, 6.28 ± 0.41 μg / μg) (FIGS. 8 and 9).

Therefore, it was confirmed that GAG (glycosaminoglycan), which is important in articular cartilage formation, was highly effective in inducing chondrocyte differentiation because of its high concentration in GM3 treatment group of 5 μM, especially in ganglioside GM3 treatment group.

Example  3. Ganglioside  In the differentiation of GM3 chondrocytes TGF Signal path  Upward adjustment ( UpRegulation ) effect

Ganglioside GM3 involves the phosphorylation of SMAD2 / 3, which is serine, TGF-beta receptor-1 (TGF-beta-R1) and TGF- beta-R2 and downstream of the TGF- Lt; RTI ID = 0.0 &gt; TGF-beta &lt; / RTI &gt; signaling.

Specifically, Western blot analysis was performed as follows. Cells were homogenized with RIPA buffer (Sigma-Aldrich) and centrifuged at 15,000 x g for 30 minutes. Protein concentrations were measured using the Bradford method. The same amount of protein (30 μg) was separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to a Hybond ECL nitrocellulose membrane (Amersham Pharmacia). Blot was stopped at room temperature for 2 hours with 5% (w / v) skim milk powder dissolved in a Tris-buffered saline containing 0.05% Tween 20. Each membrane was incubated with a specific antibody for p-serine, TGF-? -1, TGF-? -2, p-SMAD 2/3 or? -Actin (1: 500; Santa Cruz Biotechnology, And incubated for 16 hours. Each blot was incubated with the corresponding horseradish peroxidase-conjugated anti-mouse, rabbit secondary antibody (Santa Cruz Biotechnology) and the protein visualized by an enhanced chemiluminescence system (Pierce).

As a result, phosphorylation of TGF-βRs, particularly TGF-β-R2, and SMAD2 / 3 was increased in the ganglioside GM3 treated group at 5 μM (FIGS. 10 and 11). These results indicate that GM3 and TGF-β are closely related to the chondrocyte cell differentiation of hSDMSCs in TGF-β receptor activation.

<110> TJC Life <120> Composition containing ganglioside GM3 for inducing chondrogenic          differentiation, and uses thereof <130> P16E30D0461 <160> 10 <170> KoPatentin 3.0 <210> 1 <211> 23 <212> DNA <213> Artificial Sequence <220> <223> Aggrecan forward primer <400> 1 ctgagtgaaa ccacctctgc att 23 <210> 2 <211> 18 <212> DNA <213> Artificial Sequence <220> <223> Aggrecan reverse primer <400> 2 gacgcctcgc cttcttga 18 <210> 3 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> SOX-9 forward primer <400> 3 ccccaacaga tcgcctacag 20 <210> 4 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> SOX-9 reverse primer <400> 4 tctggtggtc ggtgtagtcg ta 22 <210> 5 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Collagen type 2 forward primer <400> 5 ggcaatagca ggttcacgta ca 22 <210> 6 <211> 22 <212> DNA <213> Artificial Sequence <220> <223> Collagen type 2 reverse primer <400> 6 cgataacagt cttgccccac tt 22 <210> 7 <211> 24 <212> DNA <213> Artificial Sequence <220> <223> COMP forward primer <400> 7 caagaagtcc tatcgttggt tcct 24 <210> 8 <211> 21 <212> DNA <213> Artificial Sequence <220> <223> COMP reverse primer <400> 8 ctcagggccc tcatagaatc g 21 <210> 9 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> GAPHD forward primer <400> 9 ggcatcctgg gctacactga 20 <210> 10 <211> 20 <212> DNA <213> Artificial Sequence <220> <223> GAPHD reverse primer <400> 10 gaggagtggg tgtcgctgtt 20

Claims (8)

A composition for inducing differentiation of mesenchymal stem cells into cartilage cells, comprising GM3 as an active ingredient. 2. The composition of claim 1, wherein the ganglioside GM3 is treated to a mesenchymal stem cell at a concentration of 1 to 10 [mu] M. The composition according to claim 1, wherein the mesenchymal stem cells are synovium-derived mesenchymal stem cells. A method for inducing differentiation from mesenchymal stem cells into chondrocytes, comprising culturing the mesenchymal stem cells in vitro in the presence of ganglioside GM3. 5. The method according to claim 4, wherein the ganglioside GM3 is treated to a mesenchymal stem cell at a concentration of 1 to 10 [mu] M. 5. The method according to claim 4, wherein the mesenchymal stem cells are synovium-derived mesenchymal stem cells. A pharmaceutical composition for the treatment or prevention of cartilage diseases containing ganglioside GM3 as an active ingredient. A health functional food composition for improving or preventing cartilage disease containing ganglioside GM3 as an active ingredient.
KR1020160035243A 2016-03-24 2016-03-24 Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof KR101794503B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
KR1020160035243A KR101794503B1 (en) 2016-03-24 2016-03-24 Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020160035243A KR101794503B1 (en) 2016-03-24 2016-03-24 Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof

Publications (2)

Publication Number Publication Date
KR20170113733A true KR20170113733A (en) 2017-10-13
KR101794503B1 KR101794503B1 (en) 2017-11-08

Family

ID=60139675

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020160035243A KR101794503B1 (en) 2016-03-24 2016-03-24 Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof

Country Status (1)

Country Link
KR (1) KR101794503B1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114206330A (en) * 2019-08-02 2022-03-18 三得利控股株式会社 Composition for promoting cartilage regeneration
WO2022114845A1 (en) * 2020-11-27 2022-06-02 주식회사 강스템바이오텍 Method for preparing injectable injection composition derived from animal cartilage, and use thereof

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114206330A (en) * 2019-08-02 2022-03-18 三得利控股株式会社 Composition for promoting cartilage regeneration
EP4008401A4 (en) * 2019-08-02 2023-05-03 Suntory Holdings Limited Composition for cartilage regeneration promotion
WO2022114845A1 (en) * 2020-11-27 2022-06-02 주식회사 강스템바이오텍 Method for preparing injectable injection composition derived from animal cartilage, and use thereof

Also Published As

Publication number Publication date
KR101794503B1 (en) 2017-11-08

Similar Documents

Publication Publication Date Title
Nimura et al. Increased proliferation of human synovial mesenchymal stem cells with autologous human serum: comparisons with bone marrow mesenchymal stem cells and with fetal bovine serum
US20080233195A1 (en) Synthetic triterpenoids and tricyclic-bis-enones for use in stimulating bone and cartilage growth
KR102035273B1 (en) Composition for preventing or treating osteoporosis comprising stem cell-derived exosomes
US11213572B2 (en) Cartilage regeneration composition containing HAPLN1 as active ingredient
KR20180127355A (en) Colony-forming medium and uses thereof
KR101980453B1 (en) Composition For Promoting Production of Stem Cell-derived Exosomes
JP2022533277A (en) Composition for promoting stem cell-derived exosome production and enhancing stem cell ability
KR101794503B1 (en) Composition containing ganglioside GM3 for inducing chondrogenic differentiation, and uses thereof
KR20230109119A (en) Compositions for enhancing stemness and uses thereof
EP3822343A1 (en) Production method for neuron-like cells
Zhang et al. Regulation of human mesenchymal stem cell differentiation by TREM-2
CA3188486A1 (en) A composition comprising mesenchymal precursor or stem cells and their use
WO2007136157A1 (en) Compositions and methods for treating motor neuron diseases
US20210147482A1 (en) Peptide for inhibiting bone resorption
Zhang et al. Neuritin Promotes Bone Marrow-Derived Mesenchymal Stem Cell Migration to Treat Diabetic Peripheral Neuropathy
US11466254B2 (en) Composition containing, as active ingredient, culture of chicken bone marrow-derived osteochondral progenitor cells for promoting osteogenesis or inducing chondrogenic differentiation
WO2019045451A1 (en) Cartilage regeneration composition containing hapln1 as active ingredient
KR20130085863A (en) Composition for inducing migration of mesenchymal stem cells
KR102580518B1 (en) Method for selecting mesenchymal stem cell with improved self-maintenance and mesenchymal stem cells selected by thereof
KR101998257B1 (en) Composition for preventing or treating cardiofaciocutaneous syndrome
JP5854492B2 (en) Chondrocyte differentiation inducer
KR102308505B1 (en) COMPOSITION FOR TREATMENT OF SARCOPENIA OR OSTEOPOROSIS THROUGH THE MECHANISM OF PROMOTING MYOBLAST DIFFERENTIATION OR INHIBITING THE DIFFERENTIATION OF OSTEOCLAST COMPRISING Cyclo (L-phenylalanyl-L-proline)
US20110223138A1 (en) Mesenchymal stem cells that express increased amounts of anti-apoptotic proteins
KR101805771B1 (en) A pharmaceutical composition for preventing or treating blood dyscrasia comprising TMEM100 protein
KR20240054468A (en) Medium composition for inducing differentiation from pluripotent stem cells to macrophages and a differentiation method using the same

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
E701 Decision to grant or registration of patent right
GRNT Written decision to grant