WO2022108165A1 - Method for producing exosomes isolated from induced pluripotent stem cell-derived mesenchymal stem cells, and use thereof - Google Patents

Method for producing exosomes isolated from induced pluripotent stem cell-derived mesenchymal stem cells, and use thereof Download PDF

Info

Publication number
WO2022108165A1
WO2022108165A1 PCT/KR2021/015286 KR2021015286W WO2022108165A1 WO 2022108165 A1 WO2022108165 A1 WO 2022108165A1 KR 2021015286 W KR2021015286 W KR 2021015286W WO 2022108165 A1 WO2022108165 A1 WO 2022108165A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
stem cells
minutes
medium
mesenchymal stem
Prior art date
Application number
PCT/KR2021/015286
Other languages
French (fr)
Korean (ko)
Inventor
이미연
나득채
김경운
Original Assignee
주식회사 온코인사이트
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 온코인사이트 filed Critical 주식회사 온코인사이트
Publication of WO2022108165A1 publication Critical patent/WO2022108165A1/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
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources
    • 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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/24Metalloendopeptidases (3.4.24)
    • 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
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation

Definitions

  • It relates to a method for producing an exosome isolated from dedifferentiated stem cell-derived mesenchymal stem cells and a use thereof.
  • Mesenchymal stromal cells can be obtained from adult tissues such as bone marrow, adipose tissue, umbilical cord blood, and umbilical cord, and have the form of fibroblasts.
  • the stem cells can proliferate indefinitely in vitro, and unlike blood stem cells, they can be differentiated into various types of important cell lines such as fat, osteocytes, chondrocytes, cardiomyocytes, and nerve cells. Research is being done.
  • autoimmune disease is a disease in which immune cells that defend the body from external invaders such as bacteria, viruses, and foreign substances attack themselves. Since autoimmune diseases can appear in all organs and tissues of the human body, all cells in the body are attacked or only cells in specific organs are destroyed. In addition, autoimmune diseases selectively attack specific organs or the whole body, such as rheumatoid arthritis.
  • autoimmune diseases As the types and symptoms of autoimmune diseases are diverse, the treatment methods for autoimmune diseases are also diverse.
  • the goal of treatment for autoimmune diseases is to relieve symptoms, preserve function, or block the pathogenesis of the disease.
  • Drugs for this purpose include steroids, nonsteroidal anti-inflammatory drugs, immunosuppressants, and the like, but long-term administration of these drugs often causes serious side effects.
  • One aspect comprises the steps of isolating cells from a biological sample; preparing dedifferentiated stem cells from the isolated cells; Inducing mesenchymal stem cells from the prepared dedifferentiated stem cells; And to provide a method for producing an exosome comprising the step of isolating the exosome from the induced mesenchymal stem cells.
  • Another aspect is to provide a pharmaceutical composition for preventing or treating an autoimmune disease comprising the exosome prepared by the method for producing the exosome.
  • Another aspect is to provide a method for preventing or treating an autoimmune disease comprising administering the exosome prepared by the method for producing the exosome.
  • One aspect comprises the steps of isolating cells from a biological sample; preparing dedifferentiated stem cells from the isolated cells; Inducing mesenchymal stem cells from the prepared dedifferentiated stem cells; And it provides a method for producing an exo-some comprising the step of isolating the exosome from the induced mesenchymal stem cells.
  • the "biological sample” refers to a sample derived from an organism.
  • the organism may be a mammal including a human.
  • the biological sample may be derived from a tissue or body fluid of the body.
  • the body fluids include blood, plasma, serum, urine, mucus, saliva, tears, sputum, spinal fluid, pleural fluid, nipple aspirate, lymph fluid, airway fluid, intestinal fluid, genitourinary fluid, breast milk, lymphatic fluid, semen, cerebrospinal fluid, intratracheal fluid , ascites, cystic tumor body fluid, amniotic fluid, or a combination thereof.
  • the blood may be derived from peripheral blood.
  • the blood may include blood cells.
  • the blood cells may include one or more of red blood cells, white blood cells, and platelets.
  • the white blood cells may include eosinophils, basophils, neutrophils, lymphocytes, monocytes, macrophages, and the like.
  • the cells isolated from the biological cells may be mononuclear cells.
  • the “induced pluripotent stem cell (iPSC)” is also referred to as an induced pluripotent stem cell, and refers to a cell having pluripotency obtained by dedifferentiation from a differentiated cell (eg, a somatic cell). do.
  • the dedifferentiated stem cells can differentiate into various organ cells.
  • the dedifferentiated stem cells can be obtained by reprogramming the cells differentiated by the dedifferentiation inducing factors.
  • the "mesenchymal stem cell (MSC)” refers to a stem cell present in cartilage, bone tissue, adipose tissue, bone marrow stroma, etc. differentiated from mesoderm generated by division of a fertilized egg.
  • the mesenchymal stem cells may be differentiated into osteoblasts, adipocytes, chondrocytes, etc. in vitro, and may express CD73, CD90 and CD105.
  • the mesenchymal stem cells may not express c-kit, CD11b, CD19, CD14, CD34, CD45, CD14, CD79 and HLA-DR.
  • exosomes refers to endoplasmic reticulum having a size of several tens to hundreds of nanometers (about 30 to 200 nm) composed of a double phospholipid membrane identical to the structure of a cell membrane.
  • the exosome may include a protein, a nucleic acid (mRNA, miRNA, etc.) called a exosome cargo.
  • the exosome cargo may include a wide range of signaling factors, and the signaling factors may be specific to a cell type and may be differently regulated according to the environment of the secretory cell.
  • the exosomes are intercellular signaling mediators secreted by cells, and various cellular signals delivered through them regulate cell behavior, including activation, growth, migration, differentiation, dedifferentiation, apoptosis, and necrosis of target cells.
  • the exosome may include both a nano-sized vesicle secreted from mesenchymal stem cells and released into the extracellular space or a vesicle having a composition similar to that of the exosome (eg, exosome-like vesicle). have.
  • the "inducing step” may include the following steps:
  • the "inducing step” may further include washing the dedifferentiated stem cells with PBS before step (a).
  • the collagenase refers to an enzyme capable of hydrolyzing collagen or gelatin, which is a kind of hard protein.
  • the collagenase may be any one of types 1, 2, 3 and 4.
  • the collagenase solution may contain collagenase at a concentration of 0.5 to 2 mg/mL.
  • the collagenase solution may contain collagenase type 4 (type IV Collagenase) at a concentration of 1 mg/mL.
  • Step (a) may be to react for 5 to 15 minutes.
  • step (a) may be to react the prepared retrodifferentiated stem cells with a collagenase type 4 solution for 10 minutes.
  • the step (b) may be centrifugation at 1000 to 1200 rpm, preferably centrifugation at 1100 rpm.
  • Step (b) may be centrifuged for 2 to 4 minutes.
  • step (b) may be centrifuging the dedifferentiated stem cells for 3 minutes.
  • the b-FGF (basic fibroblast growth factor), also known as FGF2 or FGF- ⁇ , refers to a growth factor encoded by the FGF2 gene.
  • the b-FGF may be stimulated at 0.5 to 1.5 ng/mL.
  • step (d) may be to stimulate the suspended dedifferentiated stem cells with 1 ng/mL of b-FGF.
  • the "inducing step” may further include stimulation with recombinant human TGF- ⁇ after stimulation with b-FGF.
  • the recombinant human TGF- ⁇ is also referred to as rh TGF- ⁇ , and may be rh TGF- ⁇ 1, rh TGF- ⁇ 2 or rh TGF- ⁇ 3.
  • step of isolating exosomes may include the following steps:
  • step (c) centrifuging the supernatant of the culture solution centrifuged in step (b) at 1500 to 2500 g at 0 to 5° C. for 15 to 30 minutes;
  • step (d) centrifuging the supernatant of the culture medium centrifuged in step (c) at 8,000 to 12,000 g at 0 to 5° C. for 20 to 40 minutes;
  • step (e) filtering the supernatant of the culture medium centrifuged in step (d) with a filter paper;
  • step (f) centrifuging the culture solution filtered in step (e) at 80,000 to 120,000 g for 100 to 140 minutes.
  • Step (a) may be culturing for 48 hours.
  • the “medium” may include essential components necessary for cell growth, survival, and proliferation in vitro .
  • the medium is DMEM (Dulbecco's Modified Eagle's Medium), MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10), DMEM/F-12 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12), ⁇ -MEM ( ⁇ -Minimal essential Medium), G-MEM (Glasgow's Minimal Essential Medium), IMDM (Isocove's Modified Dulbecco's Medium), Knockout DMEM, E8 (Essential 8 Medium), SF-DMEM (serum free-Dulbecco Modified Eagle Medium), such as commercially prepared medium or artificially synthesized medium may be used, but is not limited thereto.
  • DMEM Dulbecco's
  • the medium may be ⁇ -Minimum Eagle's Medium.
  • the ⁇ -Minimum Eagle’s Medium is also referred to as ⁇ -MEM, and refers to a medium in which amino acids and vitamins are additionally added to the components of MEM.
  • the ⁇ -MEM may include non-essential amino acids, glutamine, and 2-mercaptoethanol.
  • the ⁇ -MEM may include antibiotics such as penicillin and streptomycin.
  • the non-essential amino acids may refer to one or more selected from the group consisting of isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, histidine and arginine.
  • the centrifugation can be carried out for any length of time at any speed and temperature suitable for the purpose as is known in the art.
  • the centrifugation is 100 g, 200 g, 300 g, 400 g, 500 g, 600 g, 700 g, 800 g, 900 g, 1000 g, 2000 g, 3000 g, 4000 g, 5000 g, 6000 g, 7000 g, 8000 g, 9000 g, 10000 g, 20000 g, 30000 g, 40000 g, 50000 g, 60000 g, 70000 g, 80000 g, 90000 g, 100000 g, 200 to 400 g, 250 to 350 g, 1500-2500 g, 1800-2200 g, 1900 g-2100 g, 8,000-12,000 g, 9,000-11,000 g, or 80,000-120,000 g, 90,000-110,000 g.
  • the centrifugation may be performed at 1 minute, 2 minutes, 3 minutes, 4 minutes, 5 minutes, 6 minutes, 7 minutes, 8 minutes, 9 minutes, 10 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 120 minutes, 180 minutes, 240 minutes, 1-5 minutes, 5-10 minutes, 1-10 minutes, 5-15 minutes, 10-20 minutes, 15-20 minutes, 15-30 minutes, 20-30 minutes minutes, 20 to 40 minutes, 25 to 35 minutes, 20 to 60 minutes, 20 to 80 minutes, 20 to 100 minutes, 80 to 160 minutes, 100 to 140 minutes, 110 to 130 minutes.
  • the centrifugation is 0 °C, 1 °C, 2 °C, 3 °C, 4 °C, 5 °C, 6 °C, 7 °C, 8 °C, 9 °C, 10 °C, 15 °C, 20 °C, 25 °C, 30 °C, 40 It may be carried out at °C, 50 °C, 0 to 5 °C, 0 to 10 °C, 0 to 20 °C or 0 to 30 °C, but is not limited thereto.
  • the filter paper may have a pore size of 0.22 ⁇ m, but is not limited thereto.
  • Another aspect provides a pharmaceutical composition for preventing or treating autoimmune diseases comprising the exosomes prepared by the above manufacturing method.
  • exosome and the like are the same as described above.
  • prevention means any action that suppresses or delays the onset of the obesity or autoimmune disease
  • treatment means any action that improves or beneficially changes the symptoms of the autoimmune disease.
  • the "autoimmune disease” refers to a disease in which immune function is abnormal and immune cells attack organs or tissues of the body.
  • the autoimmune disease can be divided into organ-specific autoantibody-related diseases and organ non-specific (systemic) diseases.
  • the autoimmune disease is Hemophagocytic lymphohistiocytosis, systemic lupus erythematosus, Kikuchi's disease, vasculitis, Adult onset Still's disease, rheumatoid arthritis (rheumatoid arthritis) arthritis), Inflammatory Myositis, Behcet disease, IgG4-associated disease, Sjogren syndrome, Giant cell arteritis, Temporal arteritis, Type 1 diabetes, It may be selected from the group consisting of atopic dermatitis, Crohn's disease, systemic sclerosis, psoriasis, multiple sclerosis, and Graves hyperthyroidism, but is not limited thereto.
  • the pharmaceutical composition may include a pharmaceutically acceptable carrier.
  • the carrier is used in the sense of including excipients, diluents or adjuvants.
  • the carrier may be, for example, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, polyvinyl pi It may be selected from the group consisting of rolidone, water, physiological saline, buffers such as PBS, methyl hydroxy benzoate, propyl hydroxy benzoate, talc, magnesium stearate, and mineral oil.
  • the composition may include a filler, an anti-agglomeration agent, a lubricant, a wetting agent, a flavoring agent, an emulsifying agent, a preservative, or a combination thereof
  • the pharmaceutical composition may be provided to the subject in any formulation according to a conventional method, respectively.
  • the formulation may be an oral or parenteral formulation.
  • the solid preparation for oral administration may be a tablet, pill, powder, granule, or capsule.
  • the solid formulation may further include an excipient.
  • the excipient may be starch, calcium carbonate, sucrose, lactose, or gelatin.
  • the solid formulation may further include a lubricant such as magnesium stearate or talc.
  • the oral liquid formulation may be a suspension, an oral solution, an emulsion, or a syrup.
  • the liquid formulation may contain water or liquid paraffin.
  • the liquid formulation may include a wetting agent, a sweetening agent, a flavoring agent, or a preservative.
  • Formulations for parenteral administration may be sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilizations, or suppositories.
  • the non-aqueous solvent or suspending agent may include vegetable oil or ester.
  • the vegetable oil may be, for example, propylene glycol, polyethylene glycol, or olive oil.
  • the ester is, for example, ethyl oleate.
  • the base of the suppository may be witepsol, macrogol, tween 61, cacao butter, laurin, or glycerogelatin.
  • the pharmaceutical composition may include the exosome in a pharmaceutically effective amount.
  • effective amount refers to an amount sufficient to exhibit the effect of prophylaxis or treatment when administered to a subject in need thereof.
  • the effective amount can be appropriately selected by those skilled in the art depending on the individual.
  • the effective amount of the pharmaceutical composition is that the pharmaceutical composition contains the exosomes at least about 1 ⁇ g, at least about 2 ⁇ g, at least about 5 ⁇ g, at least about 10 ⁇ g, at least about 20 ⁇ g, at least about 50 ⁇ g, at least about 100 ⁇ g.
  • the effective amount may mean the amount of the exosome contained per 0.01 mL, 0.1 mL, 1 mL, 10 mL, or 100 mL of the pharmaceutical composition.
  • the pharmaceutical composition may be administered in a conventional manner via oral, transdermal, subcutaneous, rectal, intravenous, intraarterial, intraperitoneal, intramuscular, intrasternal, topical, or intradermal routes.
  • the pharmaceutical composition may be administered in the form of an injection, for example, subcutaneously, intravenously or intramuscularly.
  • the pharmaceutical composition may be administered systemically or locally, alone or in combination with other pharmaceutically active compounds.
  • the dosage of the pharmaceutical composition is, for example, about 0.001 mg/kg to about 100 mg/kg, about 0.01 mg/kg to about 100 mg/kg, about 0.01 mg/kg to about 10 mg/kg, based on an adult.
  • about 0.1 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 5 mg/kg, about 1 mg/kg to about 5 mg/kg, or about 1 mg/kg to about 3 mg/kg may be within the range.
  • the administration is once a day, twice a day, three times a day, four times a day, once a week, once in two weeks, once in three weeks, once in four weeks, once in a month. It may be administered once, once every three months or once a year.
  • Another aspect provides a method for preventing or treating an autoimmune disease comprising administering the exosome prepared by the method for producing the exosome.
  • a pharmaceutical composition comprising an exosome prepared according to a manufacturing method according to an aspect significantly inhibits the production of inflammatory cytokines IL-2, IL-17, IFN- ⁇ in T h 0 and T h 17 cells, and , It significantly increases the production of IL-10, an anti-inflammatory cytokine, and does not affect the activity of immunoregulatory T cells (T reg ), so it shows a very good effect in the treatment of autoimmune diseases.
  • FIGS. 1A to 1C are diagrams analyzing cell shape and markers of dedifferentiated stem cell-derived mesenchymal stem cells according to an embodiment of the present invention.
  • 1a shows the expression of NANOG, SSEA-4, TRA-181, which are markers of iPSCs confirmed by immunofluorescence
  • FIG. 1b shows iPSC-derived mesenchymal stems. It shows the results of observing the cell morphology of the cells (iPSC-iMSC) for each stage of differentiation
  • FIG. 1c shows CD34, CD31, which are reported as MSC markers in iPSC-derived mesenchymal stem cells (hiPSC-MSCs).
  • hiPSC-MSCs shows the results of confirming the expression of CD19, CD11, HALDR (negative marker), CD44, CD73, CD105, and CD90 (positive marker).
  • FIGS. 2A to 2C are diagrams analyzing physical properties and markers of exosomes isolated from dedifferentiated stem cell-derived mesenchymal stem cells according to an embodiment of the present invention.
  • Figure 2a shows the size distribution of the exosomes measured using the NANOSIGHT instrument after the separation of the dedifferentiated stem cell-derived mesenchymal stem cell exo (iPSC-iMSC-Exo)
  • Figure 2b is a transmission electron microscopy (Transmission electron microscopy)
  • TEM shows the morphology of dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo)
  • FIG. -Exo shows the expression of TGS101, CD9, CD63 (positive marker), which are reported as markers of exosomes confirmed by Western blotting.
  • Figures 3a to 3c show the results of confirming the immunomodulatory ability of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes to T h 0 cells according to an embodiment of the present invention.
  • Figure 3a shows IL-17, IFN-g and immunity generated in Th0 cells after co-culture of mononuclear cells cultured in Th0 cell differentiation conditions with dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo).
  • FIG. 3b is a graph showing the FACS result
  • Figure 3c is IL-17, IFN-g in the culture medium confirmed by ELISA and IL-10.
  • Figures 4a to 4c show the results of confirming the immune modulating ability of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo) to T h 17 cells according to an embodiment of the present invention.
  • Figure 4a shows IL-17, IFN-g, and immunity generated in Th17 cells after co-culture of mononuclear cells cultured in Th17 cell differentiation conditions with dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo). It is a result of FACS analysis of expression of CD25+Foxp3+, a marker generated in regulatory T cells (Treg), FIG. 4b is a graph showing the FACS result, and FIG. 4c is IL-17, IL-10 in the culture medium confirmed by ELISA. , and IL-2.
  • FIG. 5a to 5d show the results of comparing the disease control efficacy of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes and the mesenchymal stem cell exosomes in an arthritis animal model according to an embodiment of the present invention.
  • Figure 5a shows the arthritis evaluation index of the group administered with the dedifferentiated stem cell-derived mesenchymal stem cell exo (iPSC-iMSC-Exo) and the mesenchymal stem cell exo (MSC-Exo), respectively, in the induction of the rheumatoid arthritis animal model.
  • Figure 5b shows changes in CD4+T intracellular cytokines (CD4+IL-17A+, CD4+IL-10, etc.) in splenocytes of an animal model of rheumatoid arthritis confirmed by FACS
  • Figure 5c shows rheumatoid arthritis Shows the histological evaluation results by H & E, toluidine blue, and safranin O staining of the arthritis animal model
  • Figure 5d is the inflammation index (inflammation score) and cartilage erosion score (Cartilage damage score) calculated in each group through (* p ⁇ 0.05, ** p ⁇ 0.01, *** p ⁇ 0.001 ).
  • FIG. 6a to 6d show the results of confirming the disease control efficacy in the arthritis animal model according to the concentration of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes according to an embodiment of the present invention.
  • Figure 6a shows the arthritis evaluation index of the group administered with each concentration of immunized stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo) in the induction of an animal model of rheumatoid arthritis
  • Figure 6b is confirmed by FACS shows changes in cytokines (CD4+IL-17A+, CD4+IL-10, etc.) in splenocytes in an animal model of rheumatoid arthritis
  • FIG. 6c shows H&E, toluidine blue, and Shows the results of histological evaluation by safranin O staining
  • FIG. 6d shows the inflammation index and cartilage erosion score calculated in each group (* p ⁇ 0.05, ** p ⁇ 0.01, *** p ⁇ 0.001 ).
  • iPSC induced Pluripotent Stem Cells
  • Blood was obtained from a normal donor without disease with the approval of the institutional IRB and used.
  • To isolate mononuclear cells blood was centrifuged at 2,000 rpm for 30 minutes using a Ficoll Paque-PLUS (GE Healthcare).
  • the isolated mononuclear cells were cultured in StemSpan-ACF (Stem Cell Technologies) supplemented with StemSpan cc110 at 37° C. and 5% CO 2 conditions for 4 days. Thereafter, the cultured mononuclear cells were inoculated in a 24-well plate coated with vitronectin (Life Technologies, Invitrogen), and Sendai of the CytoTune®-iPS 2.0 Reprogramming Kit (Life Technologies, Invitrogen) according to the manufacturer's instructions.
  • IPSCs were prepared using a virus (Sendai virus). Specifically, from 3 to 21 days after transduction, cells were cultured at 37° C. and 5% CO 2 conditions, and on day 12, new iPSC colonies were individually picked and expanded for characterization. The medium was changed daily until iPSC colonies were formed. After manually picking iPSC colonies, they were cultured in TeSR-E8 medium (Stem Cell Technologies) in vitronectin-coated plates. Cultured cells were analyzed by flow cytometry for the expression of NANOG, SSEA-4, and TRA-181, which are markers of iPSCs, and identified as iPSCs.
  • a virus Sendai virus
  • Example 2 Preparation and identification of mesenchymal stem cells (iPSC-iMSC) derived from dedifferentiated stem cells
  • the retrodifferentiated stem cells cultured on a plate to 90% confluency (90% confluency) were washed once with PBS (1 x Phosphate buffered saline). did After that, 3 mL of a 1 mg/mL type IV collagenase solution was added to the plate and reacted for 10 minutes. After adding 6 mL of TeSR-E8 medium (Stem Cell Technologies), it was centrifuged at 1100 rpm for 3 minutes.
  • EB embryoid body
  • rh recombinant human TGF- ⁇ 1
  • 100 mm dishes coated with 1% gelatin for outgrowth recombinant human TGF- ⁇ 1
  • iPSC-MSCs were cultured until 90% confluency (90% confluency) in a gelatin-coated 100 mm dish while changing the culture medium every other day.
  • Example 2-1 it was confirmed through a marker whether dedifferentiated stem cell-derived mesenchymal stem cells (iPSC-MSC) were prepared.
  • iPSC-MSC dedifferentiated stem cell-derived mesenchymal stem cells
  • NANOG, SSEA-4 and TRA-181 which are markers of dedifferentiated stem cells, was confirmed by the immunofluorescence method, and the cell shape of dedifferentiated stem cell-derived mesenchymal stem cells was confirmed at each stage of differentiation.
  • CD34, CD31, CD19, CD11 and HALDR (negative markers), and CD44, CD73, CD105 and CD90 (positive markers), known as MSC markers were investigated for iPSC-MSCs. After culturing iPSC-MSCs for about 4 weeks, cells at passage 5 or higher were confirmed by flow cytometry.
  • Example 1C shows the expression of MSC markers by iPSC-MSCs confirmed by flow cytometry.
  • the retrodifferentiated stem cell-derived mesenchymal stem cells prepared in Example 2-1 expressed CD34, CD31, CD19, CD11 and HALDR, which are negative markers of MSC, in similar amounts as the isotype control, but CD44, a positive marker , CD73, CD105 and CD90 all expressed significantly higher amounts compared to the isotype control, confirming that dedifferentiated stem cell-derived mesenchymal stem cells were formed in Example 2-1.
  • Example 3 Isolation and identification of mesenchymal stem cell exo-some (MSC-Exo) and dedifferentiated stem cell-derived mesenchymal stem cell exo-some (iPSC-MSC-Exo)
  • Cord blood-derived MSCs and iPSC-MSCs were each cultured in a 100 mm dish to 100% confluence, and 5 mL of SF-DMEM (serum free-Dulbecco Modified Eagle Medium) was added thereto at 37°C and 5% CO 2 conditions for 48 hours. cultured. The culture medium was centrifuged for 10 minutes at 4 °C and 300 g conditions. The obtained supernatant was again centrifuged at 4°C and 2000 g conditions for 20 minutes. The obtained supernatant was centrifuged for 30 minutes at 10,000 g with a high-speed centrifuge (Hitachi KoKi #S306352A, Japan) to remove cell debris.
  • SF-DMEM serum free-Dulbecco Modified Eagle Medium
  • the supernatant was filtered through a 0.22 ⁇ m filter and then centrifuged for 120 minutes at 100,000 g in an ultra-high speed centrifuge (Thermo Scientific Fiberlite F50L-8x39 Rotor, Thermo Fisher Scientific). After taking out the tube from the centrifuge, the pellet part, which is the location of the exosomes, was marked in advance, all the rest except the pellet was removed to the opposite side, and the remaining pellet was suspended in PBS and stored at -80°C until use.
  • Thermo Scientific Fiberlite F50L-8x39 Rotor Thermo Fisher Scientific
  • the exosomes separated by Example 3-1 were diluted in PBS and measured six times consecutively with a NanoSight (Malvern instruments Ltd, Malvern, UK) instrument. .
  • the isolated exosomes were photographed with a transmission electron microscope (TEM). Then, in order to confirm whether the isolated exosome is correct, TSG101 (Santa Cruz Biotechnology), CD9 (Santa Cruz Biotechnology) and CD63 (Santa Cruz Biotechnology), known as exosome markers, were identified using a Western blot method.
  • Figure 2c shows that the exosomes (hiPSC-MSCs-Exo) isolated from dedifferentiated stem cell-derived mesenchymal stem cells are a control (Control: SF-DMEM medium) and dedifferentiated stem cell-derived mesenchymal stem cells themselves (hiPSCs).
  • -MSC it was confirmed that TSG101, CD9 and CD63, which are known as markers of exosomes, are expressed significantly more. Through this, it was confirmed that the material isolated in Example 3-1 was an exosome (iPSC-iMSC-Exo).
  • Example 4 Immune regulation ability of exosomes (iPSC-MSC-Exo) isolated from dedifferentiated stem cell-derived mesenchymal stem cells from T h 0 cells
  • Mononuclear cells obtained from normal human peripheral blood were differentiated into T h 0 cells as follows. Specifically, the mononuclear cells to be differentiated into T h 0 cells were transformed into RPMI 1640 supplemented with 10% fetal calf serum (FCS), 100 U/mL penicillin, 100 mg/mL streptomycin, and 2 mM L-glutamine.
  • iPSC-iMSC-Exo prepared in Example 3 (0, 0.1, 1) inoculated into medium, treated with anti-CD3 (1 ⁇ g/mL; BD Biosciences) and anti-CD28 (1 ⁇ g/mL; BD Biosciences) , and 10 ⁇ g) and 37° C.
  • IL-17 and IFN- ⁇ which are pro-inflammatory cytokines produced in T h 0 cells, and CD25+Foxp3+ expression, a marker of immunoregulatory T cells (T reg ).
  • T reg a marker of immunoregulatory T cells
  • the contents of the inflammation-inducing cytokine IL-17 and the anti-inflammatory cytokine IL-10 in each culture were analyzed.
  • cytokine content and marker expression were analyzed using FACS and sandwich ELISA.
  • Monoclonal antibodies anti-CD4-PE/Cy7 (RPA-T4, IgG1, BioLegend, San Diego, CA, USA) and anti-CD25-APC (M-A251, IgG1, ⁇ isotype, BD Biosciences) were administered for FACS.
  • control group was stained with the same type of fluorescence-labeled immunoglobulin of the same type without antigen specificity with an isotype antibody.
  • Intracellular cytokines were analyzed by flow cytometry (FACS Calibur; Becton Dickinson, San Diego, CA).
  • IL-17, IFN- ⁇ , IL-10 and IL-2 antibodies were first added to a 96-well plate (NUNC, Denmark) for sandwich ELISA at 4 ⁇ g/mL, respectively. 50 ⁇ l per well of the furnace was added and reacted at 4° C. overnight, and then 200 ⁇ l of blocking solution (1% (w/v) BSA/PBS (Phosphate-Buffered Saline) and 0.05% (v/v) tween 20/PBS) was added per well. and reacted at room temperature for 2 hours.
  • BSA/PBS Phosphate-Buffered Saline
  • tween 20/PBS 0.05%
  • recombinant IL-17, IFN- ⁇ , IL-10 and IL-2 were used to 78 pg/mL, 156 pg/mL, 312 pg/mL, 625 pg/mL, and 1250 pg, respectively. /mL, 2500 pg/mL, 5000 pg/mL concentrations were measured.
  • Figures 3a to 3c show the results of FACS analysis of the amounts of IL-17 and IFN- ⁇ produced in T h 0 cells and CD25+Foxp3+ expression produced in immunoregulatory T cells (Tregs), respectively, and Figure 3c is in the culture medium.
  • the results of measuring IL-17, IFN- ⁇ and IL-10 by ELISA are shown.
  • Immunomodulatory stem cell-derived mesenchymal stem cell exosomes iPSC-iMSC-Exo
  • iPSC-iMSC-Exo inhibited the production of IL-17 and IFN- ⁇ , which are pro-inflammatory cytokines, while further increasing the production of IL-10, an anti-inflammatory cytokine, in cell culture medium.
  • Example 3 The immunomodulatory ability of iPSC-iMSC-Exo and MSC-Exo obtained in Example 3 was confirmed, respectively. Specifically, in order to differentiate mononuclear cells obtained from normal human peripheral blood into T h 17 cells, anti-CD3 (1 ⁇ g/mL; BD Biosciences), anti-CD28 (1 ⁇ g/mL; BD Biosciences), IL- Neutralizing antibody against 1 ⁇ (20 ng/mL; R&D Systems, Minneapolis, MN, USA), IL-6 (20 ng/mL; R&D Systems), IL-23 (20 ng/mL; R&D Systems), IFN- ⁇ (IFN- ⁇ ; 2 ⁇ g/mL; R&D Systems) and a neutralizing antibody against IL-4 (2 ⁇ g/mL; R&D Systems), and Example 4 except that T h 17 was used instead of T h 0 cells The same experiment was performed.
  • FIG. 4a and 4b show the results of FACS analysis of the expression of IL-17 generated in Th17 cells and CD25+Foxp3+ generated in immunoregulatory T cells (Tregs), respectively, and FIG. 4c shows IL-17 and IL-10 in the culture medium. , and IL-2 measured by ELISA are shown.
  • iPSC-iMSC-Exo Inversely differentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo) inhibit the production of IL-17, an inflammation-inducing cytokine, but do not affect the activity of immunoregulatory T cells (T reg ) was able to confirm In addition, it was found that iPSC-iMSC-Exo decreased the production of IL-17 and IL-2, which are pro-inflammatory cytokines, and further increased the production of IL-10, an anti-inflammatory cytokine, in cell culture medium.
  • Example 6 Comparison of disease control effects in rheumatoid arthritis animal model of exosomes isolated from retrodifferentiated stem cell-derived mesenchymal stem cells (iPSC-iMSC-Exo) and mesenchymal stem cell exosomes (MSC-Exo)
  • Example 3 In order to confirm whether the exosomes (iPSC-iMSC-Exo) isolated from the dedifferentiated stem cell-derived mesenchymal stem cells obtained in Example 3 have an effect on rheumatoid arthritis, an animal model experiment was performed.
  • CII type II collagen
  • CFA Complete Freund's Adjuvant
  • Score 1 Mild swelling and redness localized to the foot or ankle joint.
  • Score 2 Mild swelling and redness from the ankle joint to the metatarsal.
  • Score 3 Moderate swelling and redness from the ankle joint to the tarsal bone.
  • cytokines IL-17, TNF-a, IL-1b, IL-10, IL-4, etc.
  • 5B and 6B show changes in CD4+T intracellular cytokines (CD4+IL-17A+, CD4+IL-10, etc.) in splenocytes of rheumatoid arthritis animal model mice measured by FACS.
  • CD4+IL-17A+, CD4+IL-10, etc. intracellular cytokines
  • the results in the rheumatoid arthritis animal model showed that the iPSC-iMSC-Exo administration group suppressed the production of inflammation-inducing cytokines (CD4+IL-17+), and the anti-inflammatory cytokines (CD4) compared to the MSC-Exo administration group. +IL-10+), and even when comparing the two groups iPSC-iMSC-Exo 50 ⁇ g, iPSC-iMSC-Exo100 ⁇ g, it was statistically significantly It was confirmed that it inhibits the production of IL-17+) and maintains the production of anti-inflammatory cytokines (CD4+IL-10+).
  • mice were sacrificed 5 weeks after the second boosting for further evaluation of the disease, and the leg joints of each mouse were excised and fixed in 10% formalin for 6 days, Calci -Decalcification was performed in a clear rapid solution for 7 hours. After fixation in 10% formalin for 2 days or in 10% formalin for 6 days, demineralized tissue was processed in calci-clear rapid for 7 hours by dehydration, clearing and paraffin infiltration in the following order.
  • the tissue was embedded in paraffin and manufactured as a block.
  • the tissue was sectioned into 4 ⁇ m using a microtome. Each slide was prepared so that there was one tissue per slide.
  • For hematoxylin & eosin staining paraffin-fixed tissue slides are placed in an oven at 65°C to melt paraffin for 1 hour, and then in NeoClear solution and different concentrations of ethanol for tissue deparaffinization and hydration. They were sequentially immersed for 5 minutes. After hematoxylin staining was performed for 10 seconds, the hematoxylin residue used for staining was removed by immersing the slide in tap water for 10 minutes to avoid direct contact with the slide.
  • the slide was immersed in the Eosin channel and stained for 10 minutes.
  • For dehydration sequentially immersed in ethanol and NeoClear solution of different concentrations, mounting was performed, and after drying in a cool place for more than a day, the tissue was checked through an optical microscope.
  • toluidine blue staining 0.1% toluidine blue solution was dispensed on the slides and stained for 10 minutes. For dehydration, they were sequentially immersed in different concentrations of ethanol and NeoClear solution. After mounting and drying in a cool place for more than one day, the tissue was confirmed through an optical microscope.
  • Histological evaluation of the rheumatoid arthritis animal model was performed according to the following evaluation criteria.
  • 1 point Slight thickening of the inner layer or some infiltrating cells in the lower layer.
  • 5c and 5d show the histological evaluation results of CIA, iPSC-iMSC-Exo and MSC-Exo.
  • the average joint index in the joint tissue was significantly lower in the iPSC-iMSC-Exo group compared to the MSC-Exo group or the CIA group through the inflammation score and the cartilage damage score. confirmed that.
  • 6c and 6d show the histological evaluation results of CIA, iPSC-iMSC-Exo 50 ⁇ g and 100 ⁇ g administration groups. Even when iPSC-iMSC-Exo 50 ⁇ g and iPSC-iMSC-Exo 100 ⁇ g administration group were compared, it was confirmed that the iPSC-iMSC-Exo concentration-dependent mean inflammation index and cartilage erosion score were low. That is, it was found that iPSC-iMSC-Exo had an excellent effect in treating rheumatoid arthritis in an animal model.

Abstract

The present invention relates to a method for producing exosomes isolated from induced pluripotent stem cell-derived mesenchymal stem cells, and a use thereof, and exosomes according to one embodiment noticeably increase the generation of IL-10, which is an anti-inflammatory cytokine, and noticeably suppress the generation of inflammatory cytokines IL-2, IL-17 and IFN-γ in Th0 and Th17 cells, and thus do not have an effect on the activity of regulatory T cells (Treg), and therefore exhibit excellent benefits for the treatment of autoimmune diseases.

Description

역분화 줄기세포 유래 중간엽 줄기세포로부터 분리된 엑소좀의 제조방법 및 이의 용도Method for preparing exosomes isolated from dedifferentiated stem cell-derived mesenchymal stem cells and use thereof
역분화 줄기세포 유래 중간엽 줄기세포로부터 분리된 엑소좀의 제조방법 및 이의 용도에 관한 것이다.It relates to a method for producing an exosome isolated from dedifferentiated stem cell-derived mesenchymal stem cells and a use thereof.
중간엽 줄기세포 (mesenchymal stromal cell)는 성체조직인 골수, 흡입된 지방조직, 제대혈, 탯줄 등에서 얻을 수 있으며 섬유모세포의 형태를 갖는다. 상기 줄기세포는 시험관에서 무한정 증식이 가능하며, 혈액줄기세포와 달리 지방, 골세포, 연골세포, 심근세포, 신경세포 등 다양한 종류의 중요한 세포계열로 분화할 수 있어 조직공학과 재생의학의 영역에서 많은 연구가 이루어지고 있다.Mesenchymal stromal cells can be obtained from adult tissues such as bone marrow, adipose tissue, umbilical cord blood, and umbilical cord, and have the form of fibroblasts. The stem cells can proliferate indefinitely in vitro, and unlike blood stem cells, they can be differentiated into various types of important cell lines such as fat, osteocytes, chondrocytes, cardiomyocytes, and nerve cells. Research is being done.
한편, 자가면역 질환은 세균, 바이러스, 이물질 등 외부 침입자로부터 인체를 방어하는 면역세포가 스스로를 공격하는 질환이다. 자가면역 질환은 인체의 모든 장기와 조직에 나타날 수 있기 때문에, 전신의 모든 세포가 공격 대상이 되기도 하고, 특정 장기의 세포만 파괴하기도 한다. 또한, 자가면역 질환은 류마티스 관절염처럼 특정 장기 또는 전신을 선택적으로 공격하기도 한다.On the other hand, autoimmune disease is a disease in which immune cells that defend the body from external invaders such as bacteria, viruses, and foreign substances attack themselves. Since autoimmune diseases can appear in all organs and tissues of the human body, all cells in the body are attacked or only cells in specific organs are destroyed. In addition, autoimmune diseases selectively attack specific organs or the whole body, such as rheumatoid arthritis.
자가면역 질환의 종류와 증상이 다양한 만큼 자가면역 질환은 그 치료 방법도 다양하다. 자가면역 질환의 치료 목표는 증상 완화, 기능 보존 또는 병의 발생 기전 차단이다. 이를 위한 약물은 스테로이드, 비스테로이드성 소염제, 면역 억제제 등이 있으나, 이들 약물을 장기로 복용할 경우 심각한 부작용이 유발되는 경우가 많다.As the types and symptoms of autoimmune diseases are diverse, the treatment methods for autoimmune diseases are also diverse. The goal of treatment for autoimmune diseases is to relieve symptoms, preserve function, or block the pathogenesis of the disease. Drugs for this purpose include steroids, nonsteroidal anti-inflammatory drugs, immunosuppressants, and the like, but long-term administration of these drugs often causes serious side effects.
이러한 기술적 배경 하에서, 상술한 자가면역 질환 예방 및 치료에 관한 문제를 해결하기 위한 연구가 지속적으로 있어 왔으나 (대한민국 등록특허공보 제10-2111836호(2020.05.15)), 아직 이를 해결하기에는 미비한 실정이다.Under this technical background, research has been continuously conducted to solve the problems related to the prevention and treatment of autoimmune diseases (Korea Patent Publication No. 10-2111836 (May 15, 2020)), but it is still insufficient to solve this problem. .
일 양상은 생물학적 시료로부터 세포를 분리하는 단계; 분리된 세포로부터 역분화 줄기세포를 제조하는 단계; 제조된 역분화 줄기세포로부터 중간엽 줄기세포를 유도하는 단계; 및 유도된 중간엽 줄기세포로부터 엑소좀을 분리하는 단계를 포함하는 엑소좀의 제조방법을 제공하는 것이다.One aspect comprises the steps of isolating cells from a biological sample; preparing dedifferentiated stem cells from the isolated cells; Inducing mesenchymal stem cells from the prepared dedifferentiated stem cells; And to provide a method for producing an exosome comprising the step of isolating the exosome from the induced mesenchymal stem cells.
다른 양상은 상기 엑소좀의 제조방법에 의해 제조된 엑소좀을 포함하는 자가면역 질환의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.Another aspect is to provide a pharmaceutical composition for preventing or treating an autoimmune disease comprising the exosome prepared by the method for producing the exosome.
또 다른 양상은 상기 엑소좀의 제조방법에 의해 제조된 엑소좀을 투여하는 단계를 포함하는 자가면역 질환을 예방 또는 치료하는 방법을 제공하는 것이다.Another aspect is to provide a method for preventing or treating an autoimmune disease comprising administering the exosome prepared by the method for producing the exosome.
일 양상은 생물학적 시료로부터 세포를 분리하는 단계; 분리된 세포로부터 역분화 줄기세포를 제조하는 단계; 제조된 역분화 줄기세포로부터 중간엽 줄기세포를 유도하는 단계; 및 유도된 중간엽 줄기세포로부터 엑소좀을 분리하는 단계를 포함하는 엑소좀의 제조방법을 제공한다.One aspect comprises the steps of isolating cells from a biological sample; preparing dedifferentiated stem cells from the isolated cells; Inducing mesenchymal stem cells from the prepared dedifferentiated stem cells; And it provides a method for producing an exo-some comprising the step of isolating the exosome from the induced mesenchymal stem cells.
상기 "생물학적 시료"는 생물로부터 유래된 시료를 의미한다. 상기 생물은 인간을 포함하는 포유동물일 수 있다. 상기 생물학적 시료는 신체의 조직 또는 체액으로부터 유래된 것일 수 있다. 상기 체액은 혈액, 혈장, 혈청, 소변, 점액, 타액, 눈물, 객담, 척수액, 흉수, 유두 흡인물, 림프액, 기도액, 장액, 비뇨생식관액, 모유, 림프계 체액, 정액, 뇌척수액, 기관계내 체액, 복수, 낭성 종양 체액, 양수액 또는 이들의 조합일 수 있다. 상기 혈액은 말초 혈액으로부터 유래된 것일 수 있다. 상기 혈액은 혈구를 포함할 수 있다. 상기 혈구는 적혈구, 백혈구 및 혈소판 중 하나 이상을 포함할 수 있다. 상기 백혈구는 호산성 백혈구, 호염기성 백혈구, 호중성 백혈구, 림프구, 단핵세포, 대식세포 등을 포함할 수 있다. 상기 생물학적 세포로부터 분리된 세포는 단핵 세포일 수 있다.The "biological sample" refers to a sample derived from an organism. The organism may be a mammal including a human. The biological sample may be derived from a tissue or body fluid of the body. The body fluids include blood, plasma, serum, urine, mucus, saliva, tears, sputum, spinal fluid, pleural fluid, nipple aspirate, lymph fluid, airway fluid, intestinal fluid, genitourinary fluid, breast milk, lymphatic fluid, semen, cerebrospinal fluid, intratracheal fluid , ascites, cystic tumor body fluid, amniotic fluid, or a combination thereof. The blood may be derived from peripheral blood. The blood may include blood cells. The blood cells may include one or more of red blood cells, white blood cells, and platelets. The white blood cells may include eosinophils, basophils, neutrophils, lymphocytes, monocytes, macrophages, and the like. The cells isolated from the biological cells may be mononuclear cells.
상기 "역분화 줄기세포 (induced Pluripotent Stem Cell: iPSC)"는 유도만능 줄기세포로도 지칭되며, 분화된 세포 (예를 들어, 체세포)로부터 역분화되어 얻어진 만능분화능 (pluripotency)을 갖는 세포를 의미한다. 상기 역분화 줄기세포는 각종 장기 세포로 분화할 수 있다. 상기 역분화 줄기세포는 역분화 유도인자들에 의해 분화된 세포를 재프로그램화 (reprogramming)하여 수득될 수 있다.The “induced pluripotent stem cell (iPSC)” is also referred to as an induced pluripotent stem cell, and refers to a cell having pluripotency obtained by dedifferentiation from a differentiated cell (eg, a somatic cell). do. The dedifferentiated stem cells can differentiate into various organ cells. The dedifferentiated stem cells can be obtained by reprogramming the cells differentiated by the dedifferentiation  inducing factors.
상기 "중간엽 줄기세포(mesenchymal stem cell: MSC)"는 수정란이 분열하여 생긴 중배엽에서 분화된 연골, 골조직, 지방조직, 골수의 기질(stroma) 등에 존재하는 줄기세포를 의미한다. 상기 중간엽 줄기세포는 시험관 내에서 뼈모세포, 지방세포, 연골세포 등으로 분화할 수 있으며, CD73, CD90 및 CD105를 발현하는 것일 수 있다. 또한, 상기 중간엽 줄기세포는 c-kit, CD11b, CD19, CD14, CD34, CD45, CD14, CD79 및 HLA-DR을 발현하지 않는 것일 수 있다.The "mesenchymal stem cell (MSC)" refers to a stem cell present in cartilage, bone tissue, adipose tissue, bone marrow stroma, etc. differentiated from mesoderm generated by division of a fertilized egg. The mesenchymal stem cells may be differentiated into osteoblasts, adipocytes, chondrocytes, etc. in vitro, and may express CD73, CD90 and CD105. In addition, the mesenchymal stem cells may not express c-kit, CD11b, CD19, CD14, CD34, CD45, CD14, CD79 and HLA-DR.
상기 "엑소좀 (exosomes)"은 세포막의 구조와 동일한 이중 인지질막으로 이루어진 수십 내지 수백 나노미터 (대략 30 내지 200 nm) 크기의 소포체를 의미한다. 상기 엑소좀은 엑소좀 카고 (cargo)라고 불리는 단백질, 핵산 (mRNA, miRNA 등) 등을 포함할 수 있다. 상기 엑소좀 카고는 광범위한 신호전달 요소 (signaling factors)를 포함할 수 있으며, 상기 신호전달 요소는 세포 타입에 특이적이고 분비 세포의 환경에 따라 상이하게 조절될 수 있다. 상기 엑소좀은 세포가 분비하는 세포 간 신호전달 매개체로서 이를 통해 전달된 다양한 세포 신호는 표적 세포의 활성화, 성장, 이동, 분화, 탈분화, 아폽토시스 (apoptosis), 괴사 (necrosis)를 포함한 세포 행동을 조절할 수 있다. 상기 엑소좀은 중간엽 줄기세포로부터 분비되어 세포외 공간으로 방출된 나노 크기의 베지클이거나 또는 상기 엑소좀과 유사한 조성을 갖는 베지클 (예를 들어, 엑소좀-유사 베지클)을 모두 포함할 수 있다.The "exosomes" refers to endoplasmic reticulum having a size of several tens to hundreds of nanometers (about 30 to 200 nm) composed of a double phospholipid membrane identical to the structure of a cell membrane. The exosome may include a protein, a nucleic acid (mRNA, miRNA, etc.) called a  exosome  cargo. The exosome   cargo may include a wide range of signaling factors, and the signaling factors may be specific to a cell type and may be differently regulated according to the environment of the secretory cell. The exosomes are intercellular signaling mediators secreted by cells, and various cellular signals delivered through them regulate cell behavior, including activation, growth, migration, differentiation, dedifferentiation, apoptosis, and necrosis of target cells. can The exosome may include both a nano-sized vesicle secreted from mesenchymal stem cells and released into the extracellular space or a vesicle having a composition similar to that of the exosome (eg, exosome-like vesicle). have.
일 구체예에서, 상기 "유도하는 단계"는 하기 단계를 포함할 수 있다: In one embodiment, the "inducing step" may include the following steps:
(a) 제조된 역분화 줄기세포를 콜라게나아제 용액과 반응시키는 단계;(a) reacting the prepared retrodifferentiated stem cells with a collagenase solution;
(b) 상기 반응시킨 역분화 줄기세포를 원심분리하는 단계;(b) centrifuging the reacted dedifferentiated stem cells;
(c) 상기 원심분리한 역분화 줄기세포를 배지에서 현탁하는 단계; 및(c) suspending the centrifuged retrodifferentiated stem cells in a medium; and
(d) 상기 현탁된 역분화 줄기세포를 b-FGF로 자극하는 단계.(d) stimulating the suspended retrodifferentiated stem cells with b-FGF.
상기 "유도하는 단계" 상기 (a) 단계 전에 역분화 줄기세포를 PBS로 세척하는 단계를 추가로 포함할 수 있다.The "inducing step" may further include washing the dedifferentiated stem cells with PBS before step (a).
상기 콜라게나아제 (collagenase)는 경단백질의 일종인 콜라겐이나 젤라틴을 가수분해할 수 있는 효소를 의미한다. 상기 콜라게나아제는 타입 1, 2, 3 및 4 중 어느 하나일 수 있다. 상기 콜라게나아제 용액은 콜라게나아제를 0.5 내지 2 mg/mL의 농도로 포함할 수 있다. 예를 들어, 상기 콜라게나아제 용액은 콜라게나아제 타입 4 (type IV Collagenase)를 1 mg/mL의 농도로 포함할 수 있다. 상기 (a) 단계는 5 내지 15 분 동안 반응시키는 것일 수 있다. 바람직하게는 상기 (a) 단계는 제조된 역분화 줄기세포를 콜라게나아제 타입 4 용액과 10 분 동안 반응시키는 것일 수 있다.The collagenase (collagenase) refers to an enzyme capable of hydrolyzing collagen or gelatin, which is a kind of hard protein. The collagenase may be any one of types 1, 2, 3 and 4. The collagenase solution may contain collagenase at a concentration of 0.5 to 2 mg/mL. For example, the collagenase solution may contain collagenase type 4 (type  IV  Collagenase) at a concentration of 1 mg/mL. Step (a) may be to react for 5 to 15 minutes. Preferably, step (a) may be to react the prepared retrodifferentiated stem cells with a collagenase type 4 solution for 10 minutes.
상기 (b) 단계는 1000 내지 1200 rpm으로 원심분리하는 것일 수 있으며, 바람직하게는 1100 rpm으로 원심분리하는 것일 수 있다. 상기 (b) 단계는 2 내지 4 분 동안 원심분리되는 것일 수 있다. 예를 들어, 상기 (b) 단계는 역분화 줄기세포를 3 분 동안 원심분리하는 것일 수 있다.The step (b) may be centrifugation at 1000 to 1200 rpm, preferably centrifugation at 1100 rpm. Step (b) may be centrifuged for 2 to 4 minutes. For example, step (b) may be centrifuging the dedifferentiated stem cells for 3 minutes.
상기 b-FGF (basic fibroblast growth factor)는 FGF2 또는 FGF-β로도 알려져 있으며, FGF2 유전자로 코딩되어 있는 성장인자를 의미한다. 상기 (d) 단계에서 상기 b-FGF는 0.5 내지 1.5 ng/mL로 자극하는 것일 수 있다. 예를 들어, 상기 (d) 단계는 상기 현탁된 역분화 줄기세포를 1 ng/mL의 b-FGF로 자극하는 것일 수 있다.The b-FGF (basic fibroblast growth factor), also known as FGF2 or FGF-β, refers to a growth factor encoded by the FGF2 gene. In step (d), the b-FGF may be stimulated at 0.5 to 1.5 ng/mL. For example, step (d) may be to stimulate the suspended dedifferentiated stem cells with 1 ng/mL of b-FGF.
상기 "유도하는 단계"는 b-FGF로 자극하는 단계 후 재조합 인간 (recombinant human) TGF-β로 자극하는 단계를 추가로 포함할 수 있다. 상기 재조합 인간 TGF-β는 rh TGF-β라고도 지칭하며, rh TGF-β1, rh TGF-β2 또는 rh TGF-β3일 수 있다.The "inducing step" may further include stimulation with recombinant human TGF-β after stimulation with b-FGF. The recombinant human TGF-β is also referred to as rh TGF-β, and may be rh TGF-β1, rh TGF-β2 or rh TGF-β3.
상기 "엑소좀을 분리하는 단계"는 하기 단계를 포함하는 것일 수 있다:The "step of isolating exosomes" may include the following steps:
(a) 유도된 중간엽 줄기세포를 배지에서 36 내지 60 시간 동안 배양하는 단계;(a) culturing the induced mesenchymal stem cells in a medium for 36 to 60 hours;
(b) 상기 배양된 중간엽 줄기세포 배양액을 0 내지 5℃에서 200 내지 400 g로 5 내지 15 분 동안 원심분리하는 단계;(b) centrifuging the cultured mesenchymal stem cell culture medium at 200 to 400 g at 0 to 5° C. for 5 to 15 minutes;
(c) 상기 (b) 단계에서 원심분리된 배양액의 상층액을 0 내지 5℃에서 1500 내지 2500 g로 15 내지 30 분 동안 원심분리하는 단계;(c) centrifuging the supernatant of the culture solution centrifuged in step (b) at 1500 to 2500 g at 0 to 5° C. for 15 to 30 minutes;
(d) 상기 (c) 단계에서 원심분리된 배양액의 상층액을 0 내지 5℃에서 8,000 내지 12,000 g로 20 내지 40 분 동안 원심분리하는 단계;(d) centrifuging the supernatant of the culture medium centrifuged in step (c) at 8,000 to 12,000 g at 0 to 5° C. for 20 to 40 minutes;
(e) 상기 (d) 단계에서 원심분리된 배양액의 상층액을 여과지로 필터링하는 단계; 및(e) filtering the supernatant of the culture medium centrifuged in step (d) with a filter paper; and
(f) 상기 (e) 단계에서 필터링된 배양액을 80,000 내지 120,000 g에서 100 내지 140 분 동안 원심분리하는 단계.(f) centrifuging the culture solution filtered in step (e) at 80,000 to 120,000 g for 100 to 140 minutes.
상기 (a) 단계는 48 시간 동안 배양되는 것일 수 있다. Step (a) may be culturing for 48 hours.
상기 "배지"는 시험관 내 (in vitro)에서 세포의 성장과 생존 및 증식에 필요한 필수성분을 포함하는 것일 수 있다. 예를 들어, 상기 배지는 DMEM (Dulbecco's Modified Eagle's Medium), MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10), DMEM/F-12 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12), α-MEM (α-Minimal essential Medium), G-MEM (Glasgow's Minimal Essential Medium), IMDM (Isocove's Modified Dulbecco's Medium), Knockout DMEM, E8 (Essential 8 Medium), SF-DMEM (serum free-Dulbecco Modified Eagle Medium) 등의 상업적으로 제조된 배지 또는 인위적으로 합성한 배지를 이용할 수 있으나, 이에 제한되는 것은 아니다.The "medium" may include essential components necessary for cell growth, survival, and proliferation in vitro . For example, the medium is DMEM (Dulbecco's Modified Eagle's Medium), MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10), DMEM/F-12 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12), α-MEM (α-Minimal essential Medium), G-MEM (Glasgow's Minimal Essential Medium), IMDM (Isocove's Modified Dulbecco's Medium), Knockout DMEM, E8 (Essential 8 Medium), SF-DMEM (serum free-Dulbecco Modified Eagle Medium), such as commercially prepared medium or artificially synthesized medium may be used, but is not limited thereto.
상기 배지는 α-Minimum Eagle’s Medium일 수 있다. 상기 α-Minimum Eagle’s Medium은 α-MEM이라고도 하며, MEM의 성분에 아미노산 비타민 등이 추가로 첨가되어 있는 배지를 의미한다. 구체적으로, 상기 α-MEM은 비필수 아미노산, 글루타민, 2-머캡토에탄올을 포함할 수 있다. 또한, 상기 α-MEM은 페니실린, 스트렙토마이신 등의 항생제를 포함할 수 있다. 상기 비필수 아미노산은 이소류신, 류신, 라이신, 메티오닌, 페닐알라닌, 트레오닌, 트립토판, 발린, 히스티틴 및 아르기닌으로 구성된 군으로부터 선택된 하나 이상을 의미하는 것일 수 있다.The medium may be α-Minimum Eagle's Medium. The α-Minimum Eagle’s Medium is also referred to as α-MEM, and refers to a medium in which amino acids and vitamins are additionally added to the components of MEM. Specifically, the α-MEM may include non-essential amino acids, glutamine, and 2-mercaptoethanol. In addition, the α-MEM may include antibiotics such as penicillin and streptomycin. The non-essential amino acids may refer to one or more selected from the group consisting of isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, histidine and arginine.
상기 "원심분리"는 당업계에 알려진 바에 따라 목적에 맞게 임의의 속도 및 온도로 임의의 시간 동안 수행될 수 있다. 예를 들어, 상기 원심분리는 100 g, 200 g, 300 g, 400 g, 500 g, 600 g, 700 g, 800 g, 900 g, 1000 g, 2000 g, 3000 g, 4000 g, 5000 g, 6000 g, 7000 g, 8000 g, 9000 g, 10000 g, 20000 g, 30000 g, 40000 g, 50000 g, 60000 g, 70000 g, 80000 g, 90000 g, 100000 g, 200 내지 400 g, 250 내지 350 g, 1500 내지 2500 g, 1800 내지 2200 g, 1900 g 내지 2100 g, 8,000 내지 12,000 g, 9,000 내지 11,000 g, 또는 80,000 내지 120,000 g, 90,000 내지 110,000 g의 속도로 수행될 수 있다. 예를 들어, 상기 원심분리는 1 분, 2 분, 3 분, 4 분, 5 분, 6 분, 7 분, 8 분, 9 분, 10 분, 20 분, 30 분, 40 분, 50 분, 60 분, 120 분, 180 분, 240 분, 1 내지 5 분, 5 내지 10 분, 1 내지 10 분, 5 내지 15 분, 10 내지 20 분, 15 내지 20 분, 15 내지 30 분, 20 내지 30 분, 20 내지 40 분, 25분 내지 35 분, 20 내지 60 분, 20 내지 80 분, 20 내지 100 분, 80 내지 160 분, 100 내지 140 분, 110 내지 130 분 동안 수행될 수 있다. 상기 원심분리는 0℃, 1℃, 2℃, 3℃, 4℃, 5℃, 6℃, 7℃, 8℃, 9℃, 10℃, 15℃, 20℃, 25℃, 30℃, 40℃, 50℃, 0 내지 5℃, 0 내지 10℃, 0 내지 20℃ 또는 0 내지 30℃에서 수행될 수 있으나, 이에 제한되지는 않는다.The "centrifugation" can be carried out for any length of time at any speed and temperature suitable for the purpose as is known in the art. For example, the centrifugation is 100 g, 200 g, 300 g, 400 g, 500 g, 600 g, 700 g, 800 g, 900 g, 1000 g, 2000 g, 3000 g, 4000 g, 5000 g, 6000 g, 7000 g, 8000 g, 9000 g, 10000 g, 20000 g, 30000 g, 40000 g, 50000 g, 60000 g, 70000 g, 80000 g, 90000 g, 100000 g, 200 to 400 g, 250 to 350 g, 1500-2500 g, 1800-2200 g, 1900 g-2100 g, 8,000-12,000 g, 9,000-11,000 g, or 80,000-120,000 g, 90,000-110,000 g. For example, the centrifugation may be performed at 1 minute, 2 minutes, 3 minutes, 4 minutes, 5 minutes, 6 minutes, 7 minutes, 8 minutes, 9 minutes, 10 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 120 minutes, 180 minutes, 240 minutes, 1-5 minutes, 5-10 minutes, 1-10 minutes, 5-15 minutes, 10-20 minutes, 15-20 minutes, 15-30 minutes, 20-30 minutes minutes, 20 to 40 minutes, 25 to 35 minutes, 20 to 60 minutes, 20 to 80 minutes, 20 to 100 minutes, 80 to 160 minutes, 100 to 140 minutes, 110 to 130 minutes. The centrifugation is 0 ℃, 1 ℃, 2 ℃, 3 ℃, 4 ℃, 5 ℃, 6 ℃, 7 ℃, 8 ℃, 9 ℃, 10 ℃, 15 ℃, 20 ℃, 25 ℃, 30 ℃, 40 It may be carried out at ℃, 50 ℃, 0 to 5 ℃, 0 to 10 ℃, 0 to 20 ℃ or 0 to 30 ℃, but is not limited thereto.
상기 여과지는 0.22 ㎛의 포어 사이즈를 가질 수 있으나, 이에 제한되지는 않는다.The filter paper may have a pore size of 0.22 μm, but is not limited thereto.
다른 양상은 상기 제조방법에 의해 제조된 엑소좀을 포함하는 자가면역 질환의 예방 또는 치료용 약학적 조성물을 제공한다.Another aspect provides a pharmaceutical composition for preventing or treating autoimmune diseases comprising the exosomes prepared by the above manufacturing method.
상기 용어 엑소좀 등에 대해서는 상술한 바와 같다.The term exosome and the like are the same as described above.
상기 "예방"은 상기 비만 또는 자가면역 질환의 발병을 억제 또는 지연시키는 모든 행위를 의미하고, 상기 "치료"는 상기 자가면역 질환의 증세가 호전되거나 이롭게 변경하는 모든 행위를 의미한다.The "prevention" means any action that suppresses or delays the onset of the obesity or autoimmune disease, and the "treatment" means any action that improves or beneficially changes the symptoms of the autoimmune disease.
상기 "자가면역 질환 (autoimmune disease)"은 면역 기능에 이상이 발생하여, 면역세포들이 신체의 장기나 조직을 공격하여 발생하는 질환을 말한다. 상기 자가면역 질환은 장기 특이적 자가항체와 관련된 질환과 장기 비특이적 (전신적) 질환으로 구별될 수 있다. 상기 자가면역 질환은 혈구탐식성 림프조직구증 (Hemophagocytic lymphohistiocytosis), 전신 홍반 루푸스 (systemic lupus erythematosus), 기쿠치 (Kikuchi) 병, 혈관염 (vasculitis), 성인 스틸 병 (Adult onset Still's disease), 류마티스 관절염 (rheumatoid arthritis), 염증성 근육염 (Inflammatory Myositis), 베체트 병 (Behcet disease), IgG4-연관성 질환, 쇼그렌 증후군 (Sjogren syndrome), 거대세포 동맥염 (Giant cell arteritis), 측두 동맥염 (Temporal arteritis), 제1형 당뇨병, 아토피 피부염, 크론병 (Crohn's disease), 전신성 경화증 (systemic sclerosis), 건선, 다발성 경화증 (multiple sclerosis), 및 그레이브스 갑상선 항진증 (Graves hyperthyroidism)으로 이루어진 군으로부터 선택될 수 있으나, 이에 제한되지는 않는다.The "autoimmune disease" refers to a disease in which immune function is abnormal and immune cells attack organs or tissues of the body. The autoimmune disease can be divided into organ-specific autoantibody-related diseases and organ non-specific (systemic) diseases. The autoimmune disease is Hemophagocytic lymphohistiocytosis, systemic lupus erythematosus, Kikuchi's disease, vasculitis, Adult onset Still's disease, rheumatoid arthritis (rheumatoid arthritis) arthritis), Inflammatory Myositis, Behcet disease, IgG4-associated disease, Sjogren syndrome, Giant cell arteritis, Temporal arteritis, Type 1 diabetes, It may be selected from the group consisting of atopic dermatitis, Crohn's disease, systemic sclerosis, psoriasis, multiple sclerosis, and Graves hyperthyroidism, but is not limited thereto.
상기 약학적 조성물은 약학적으로 허용가능한 담체를 포함할 수 있다. 상기 담체는 부형제, 희석제 또는 보조제를 포함하는 의미로 사용된다. 상기 담체는 예를 들면, 락토스, 덱스트로스, 수크로스, 소르비톨, 만니톨, 자일리톨, 에리트리톨, 말티톨, 전분, 아카시아 고무, 알기네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로스, 메틸 셀룰로스, 폴리비닐 피롤리돈, 물, 생리식염수, PBS와 같은 완충액, 메틸히드록시 벤조에이트, 프로필히드록시 벤조에이트, 탈크, 마그네슘 스테아레이트, 및 미네랄 오일로 이루어진 군으로부터 선택된 것일 수 있다. 상기 조성물은 충진제, 항응집제, 윤활제, 습윤제, 풍미제, 유화제, 보존제, 또는 이들의 조합을 포함할 수 있다.The pharmaceutical composition may include a pharmaceutically acceptable carrier. The carrier is used in the sense of including excipients, diluents or adjuvants. The carrier may be, for example, lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, gum acacia, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, polyvinyl pi It may be selected from the group consisting of rolidone, water, physiological saline, buffers such as PBS, methyl hydroxy benzoate, propyl hydroxy benzoate, talc, magnesium stearate, and mineral oil. The composition may include a filler, an anti-agglomeration agent, a lubricant, a wetting agent, a flavoring agent, an emulsifying agent, a preservative, or a combination thereof.
상기 약학적 조성물은 각각 통상의 방법에 따라 임의의 제제로 개체에 제공될 수 있다. 상기 제제는 경구용 또는 비경구용 제제일 수 있다. 상기 약학적 조성물에 있어서, 경구 투여를 위한 고형 제제는 정제, 환제, 산제, 과립제, 또는 캡슐제일 수 있다. 상기 고형 제제는 부형제를 더 포함할 수 있다. 상기 부형제는 전분, 칼슘카보네이트(calcium carbonate), 수크로스(sucrose), 락토오스(lactose), 또는 젤라틴일 수 있다. 또한, 상기 고형 제제는 마그네슘 스테아레이트, 또는 탈크와 같은 윤활제를 더 포함할 수 있다. 상기 약학적 조성물에 있어서, 경구를 위한 액상 제제는 현탁제, 내용액제, 유제, 또는 시럽제일 수 있다. 상기 액상 제제는 물, 또는 리퀴드 파라핀을 포함할 수 있다. 상기 액상 제제는 습윤제, 감미제, 방향제, 또는 보존제를 포함할 수 있다. 비경구 투여를 위한 제제는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조 또는 좌제일 수 있다. 비수성용제 또는 현탁제는 식물성 기름 또는 에스테르를 포함할 수 있다. 식물성 기름은 예를 들면, 프로필렌글리콜(propylene glycol), 폴리에틸렌글리콜, 또는 올리브 오일일 수 있다. 에스테르는 예를 들면 에틸올레이트이다. 좌제의 기제는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 또는 글리세로젤라틴일 수 있다.The pharmaceutical composition may be provided to the subject in any formulation according to a conventional method, respectively. The formulation may be an oral or parenteral formulation. In the pharmaceutical composition, the solid preparation for oral administration may be a tablet, pill, powder, granule, or capsule. The solid formulation may further include an excipient. The excipient may be starch, calcium carbonate, sucrose, lactose, or gelatin. In addition, the solid formulation may further include a lubricant such as magnesium stearate or talc. In the pharmaceutical composition, the oral liquid formulation may be a suspension, an oral solution, an emulsion, or a syrup. The liquid formulation may contain water or liquid paraffin. The liquid formulation may include a wetting agent, a sweetening agent, a flavoring agent, or a preservative. Formulations for parenteral administration may be sterile aqueous solutions, non-aqueous solutions, suspensions, emulsions, lyophilizations, or suppositories. The non-aqueous solvent or suspending agent may include vegetable oil or ester. The vegetable oil may be, for example, propylene glycol, polyethylene glycol, or olive oil. The ester is, for example, ethyl oleate. The base of the suppository may be witepsol, macrogol, tween 61, cacao butter, laurin, or glycerogelatin.
상기 약학적 조성물은 상기 엑소좀을 약학적으로 유효한 양으로 포함할 수 있다. 용어 "유효한 양"은 예방 또는 치료를 필요로 하는 개체에게 투여되는 경우 예방 또는 치료의 효과를 나타내기에 충분한 양을 말한다. 상기 유효한 양은 당업자가 개체에 따라 적절하게 선택할 수 있다. 질환의 중증도, 환자의 연령, 체중, 건강, 성별, 환자의 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 사용된 조성물과 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 상기 약학적 조성물의 유효한 양은 상기 약학적 조성물이 상기 엑소좀을 약 1 ㎍ 이상, 약 2 ㎍ 이상, 약 5 ㎍ 이상, 약 10 ㎍ 이상, 약 20 ㎍ 이상, 약 50 ㎍ 이상, 약 100 ㎍ 이상, 약 200 ㎍ 이상, 약 400 ㎍ 이상, 약 1 mg 이상, 약 2 mg 이상, 약 4 mg 이상, 약 10 mg 이상, 약 20 mg 이상, 약 40 mg 이상, 약 100 mg 이상, 약 1 ㎍ 내지 1 mg, 약 10 μg 내지 1 mg, 약 1 내지 100 ㎍, 약 10 내지 100 ㎍, 약 10 내지 200 ㎍, 또는 약 10 내지 500 ㎍으로 포함하는 것일 수 있으나, 이에 한정되는 것은 아니다. 상기 유효한 양은 상기 엑소좀이 상기 약학적 조성물 0.01 mL, 0.1 mL, 1 mL, 10 mL 또는 100 mL 당 포함되어 있는 양을 의미하는 것일 수 있다.The pharmaceutical composition may include the exosome in a pharmaceutically effective amount. The term "effective amount" refers to an amount sufficient to exhibit the effect of prophylaxis or treatment when administered to a subject in need thereof. The effective amount can be appropriately selected by those skilled in the art depending on the individual. disease severity, patient's age, weight, health, sex, patient's sensitivity to drug, administration time, administration route and excretion rate, duration of treatment, factors including drugs used in combination with or concurrently with the composition used, and other medical fields can be determined according to well-known factors in The effective amount of the pharmaceutical composition is that the pharmaceutical composition contains the exosomes at least about 1 μg, at least about 2 μg, at least about 5 μg, at least about 10 μg, at least about 20 μg, at least about 50 μg, at least about 100 μg. , about 200 μg or more, about 400 μg or more, about 1 mg or more, about 2 mg or more, about 4 mg or more, about 10 mg or more, about 20 mg or more, about 40 mg or more, about 100 mg or more, about 1 μg to 1 mg, about 10 μg to 1 mg, about 1 to 100 μg, about 10 to 100 μg, about 10 to 200 μg, or about 10 to 500 μg may be included, but is not limited thereto. The effective amount may mean the amount of the exosome contained per 0.01 mL, 0.1 mL, 1 mL, 10 mL, or 100 mL of the pharmaceutical composition.
상기 약학적 조성물은 경구, 경피, 피하, 직장, 정맥내, 동맥내, 복강내, 근육내, 흉골내, 국소, 또는 피내 경로를 통해 통상적인 방식으로 투여될 수 있다. 상기 약학적 조성물은 예를 들어 피하, 정맥내 또는 근육내로 주사제 제형으로 투여될 수 있다. 또한 상기 약학적 조성물은 전신적으로 또는 국부적으로 투여될 수 있고, 단독으로 또는 다른 약학적 활성 화합물과 함께 투여될 수 있다. 상기 약학적 조성물의 투여량은 예를 들어, 성인 기준으로 약 0.001 ㎎/kg 내지 약 100 ㎎/kg, 약 0.01 ㎎/kg 내지 약 100 ㎎/kg, 약 0.01 ㎎/kg 내지 약 10 ㎎/kg, 약 0.1 ㎎/kg 내지 약 10 ㎎/kg, 약 0.1 ㎎/kg 내지 약 5 ㎎/kg, 약 1 ㎎/kg 내지 약 5 ㎎/kg, 또는 약 1 ㎎/kg 내지 약 3 ㎎/kg의 범위내일 수 있다. 상기 투여는 1 일 1 회, 1 일 2 회, 1 일 3 회, 1 일 4 회, 1 주일에 1 회, 2 주일에 1 회, 3 주일에 1 회, 4 주일에 1 회, 1 달에 1 회, 3 달에 1회 또는 1 년에 1 회 투여될 수 있다.The pharmaceutical composition may be administered in a conventional manner via oral, transdermal, subcutaneous, rectal, intravenous, intraarterial, intraperitoneal, intramuscular, intrasternal, topical, or intradermal routes. The pharmaceutical composition may be administered in the form of an injection, for example, subcutaneously, intravenously or intramuscularly. In addition, the pharmaceutical composition may be administered systemically or locally, alone or in combination with other pharmaceutically active compounds. The dosage of the pharmaceutical composition is, for example, about 0.001 mg/kg to about 100 mg/kg, about 0.01 mg/kg to about 100 mg/kg, about 0.01 mg/kg to about 10 mg/kg, based on an adult. , about 0.1 mg/kg to about 10 mg/kg, about 0.1 mg/kg to about 5 mg/kg, about 1 mg/kg to about 5 mg/kg, or about 1 mg/kg to about 3 mg/kg may be within the range. The administration is once a day, twice a day, three times a day, four times a day, once a week, once in two weeks, once in three weeks, once in four weeks, once in a month. It may be administered once, once every three months or once a year.
또 다른 양상은 상기 엑소좀의 제조방법에 의해 제조된 엑소좀을 투여하는 단계를 포함하는 자가면역 질환을 예방 또는 치료하는 방법을 제공한다.Another aspect provides a method for preventing or treating an autoimmune disease comprising administering the exosome prepared by the method for producing the exosome.
일 양상에 따른 제조방법에 따라 제조된 엑소좀을 포함하는 약학적 조성물은 Th0 및 Th17 세포에서 염증성 사이토카인인 IL-2, IL-17, IFN-γ의 생성을 유의하게 억제시키고, 염증억제성 사이토카인인 IL-10의 생성을 유의하게 증가시키며, 면역조절 T세포 (Treg)의 활성에는 영향을 미치지 않기 때문에 자가면역 질환의 치료에 매우 우수한 효과를 나타낸다.A pharmaceutical composition comprising an exosome prepared according to a manufacturing method according to an aspect significantly inhibits the production of inflammatory cytokines IL-2, IL-17, IFN-γ in T h 0 and T h 17 cells, and , It significantly increases the production of IL-10, an anti-inflammatory cytokine, and does not affect the activity of immunoregulatory T cells (T reg ), so it shows a very good effect in the treatment of autoimmune diseases.
도 1a 내지 1c는 본 발명의 일 구체예에 따른 역분화 줄기세포 유래 중간엽 줄기세포의 세포 모양 및 마커를 분석한 도면이다. 도 1a는 면역형광(Immunofluorescence)에 의해 확인된 역분화줄기세포 (iPSC)의 마커인 NANOG, SSEA-4, TRA-181발현을 보여주고, 도 1b는 역분화줄기세포 (iPSC) 유래 중간엽 줄기세포 (iPSC-iMSC)의 분화단계별 세포 형태를 관찰한 결과를 보여주며, 도 1c는 역분화줄기세포 (iPSC)유래 중간엽 줄기세포 (hiPSC-MSCs)에서 MSC의 마커로 보고되어 있는 CD34, CD31, CD19, CD11, HALDR (Negative marker), CD44, CD73, CD105, CD90 (Positive marker)의 발현을 확인한 결과를 보여준다. 1A to 1C are diagrams analyzing cell shape and markers of dedifferentiated stem cell-derived mesenchymal stem cells according to an embodiment of the present invention. 1a shows the expression of NANOG, SSEA-4, TRA-181, which are markers of iPSCs confirmed by immunofluorescence, and FIG. 1b shows iPSC-derived mesenchymal stems. It shows the results of observing the cell morphology of the cells (iPSC-iMSC) for each stage of differentiation, and FIG. 1c shows CD34, CD31, which are reported as MSC markers in iPSC-derived mesenchymal stem cells (hiPSC-MSCs). , shows the results of confirming the expression of CD19, CD11, HALDR (negative marker), CD44, CD73, CD105, and CD90 (positive marker).
도 2a 내지 2c는 본 발명의 일 구체예에 따른 역분화 줄기세포 유래 중간엽 줄기세포으로부터 분리된 엑소좀의 물리적 특성 및 마커를 분석한 도면이다. 도 2a는 역분화줄기세포 유래 중간엽 줄기세포 엑소좀 (iPSC-iMSC-Exo)의 분리후 NANOSIGHT 기기를 이용하여 측정된 엑소좀의 크기 분포를 보여주고, 도 2b는 투과전자현미경 (Transmission electron microscopy, TEM)을 이용하여 확인된 역분화줄기세포 유래 중간엽 줄기세포 엑소좀(iPSC-iMSC-Exo)의 형태를 보여주며, 도 2c는 역분화줄기세포 유래 중간엽 줄기세포 엑소좀(iPSC-iMSC-Exo)에서 웨스턴 블롯팅에 의해 확인된 엑소좀의 마커로 보고되어 있는 TGS101, CD9, CD63 (Positive marker)의 발현을 보여준다. 2A to 2C are diagrams analyzing physical properties and markers of exosomes isolated from dedifferentiated stem cell-derived mesenchymal stem cells according to an embodiment of the present invention. Figure 2a shows the size distribution of the exosomes measured using the NANOSIGHT instrument after the separation of the dedifferentiated stem cell-derived mesenchymal stem cell exo (iPSC-iMSC-Exo), Figure 2b is a transmission electron microscopy (Transmission electron microscopy) , TEM) shows the morphology of dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo), and FIG. -Exo) shows the expression of TGS101, CD9, CD63 (positive marker), which are reported as markers of exosomes confirmed by Western blotting.
도 3a 내지 3c는 본 발명의 일 구체예에 따른 역분화 줄기세포 유래 중간엽 줄기세포 엑소좀의 Th0 세포에 대한 면역 조절능을 확인한 결과를 보여준다. 도 3a는 Th0 세포 분화 조건에서 배양된 단핵구 세포를 역분화줄기세포 유래 중간엽 줄기세포 엑소좀 (iPSC-iMSC-Exo)과 공배양한 후 Th0 세포에서 생성되는 IL-17, IFN-g 및 면역조절T세포 (Treg)에서 생성되는 마커인 CD25+Foxp3+ 발현을 FACS로 분석한 결과이고, 도 3b는 FACS 결과를 도시한 그래프이며, 도 3c는 ELISA로 확인된 배양액 중 IL-17, IFN-g 및 IL-10을 보여준다. Figures 3a to 3c show the results of confirming the immunomodulatory ability of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes to T h 0 cells according to an embodiment of the present invention. Figure 3a shows IL-17, IFN-g and immunity generated in Th0 cells after co-culture of mononuclear cells cultured in Th0 cell differentiation conditions with dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo). It is a result of FACS analysis of the expression of CD25+Foxp3+, a marker generated in regulatory T cells (Treg), Figure 3b is a graph showing the FACS result, Figure 3c is IL-17, IFN-g in the culture medium confirmed by ELISA and IL-10.
도 4a 내지 4c는 본 발명의 일 구체예에 따른 역분화 줄기세포 유래 중간엽 줄기세포 엑소좀(iPSC-iMSC-Exo)의 Th17 세포에 대한 면역 조절능을 확인한 결과를 보여준다. 도 4a는 Th17 세포 분화 조건에서 배양된 단핵구 세포를 역분화줄기세포 유래 중간엽 줄기세포 엑소좀 (iPSC-iMSC-Exo)과 공배양한 후 Th17세포에서 생성되는 IL-17, IFN-g 및 면역조절T세포 (Treg)에서 생성되는 마커인 CD25+Foxp3+ 발현을 FACS로 분석한 결과이고, 도 4b는 FACS 결과를 도시한 그래프이며, 도 4c는 ELISA로 확인된 배양액 중 IL-17, IL-10, 및 IL-2를 보여준다.Figures 4a to 4c show the results of confirming the immune modulating ability of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo) to T h 17 cells according to an embodiment of the present invention. Figure 4a shows IL-17, IFN-g, and immunity generated in Th17 cells after co-culture of mononuclear cells cultured in Th17 cell differentiation conditions with dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo). It is a result of FACS analysis of expression of CD25+Foxp3+, a marker generated in regulatory T cells (Treg), FIG. 4b is a graph showing the FACS result, and FIG. 4c is IL-17, IL-10 in the culture medium confirmed by ELISA. , and IL-2.
도 5a 내지 5d는 본 발명의 일 구체예에 따른 역분화 줄기세포 유래 중간엽 줄기세포 엑소좀과 중간엽 줄기세포 엑소좀의 관절염 동물 모델에서 질환 제어 효능을 비교한 결과를 보여준다. 도 5a는 류마티스 관절염 동물 모델의 유도에서 각각 역분화줄기세포 유래 중간엽 줄기세포 엑소좀(iPSC-iMSC-Exo) 및 중간엽 줄기세포 엑소좀(MSC-Exo)을 투여한 그룹의 관절염 평가 지수를 보여주고, 도 5b는 FACS에 의해 확인된 류마티스 관절염 동물 모델의 비장세포 내 CD4+T 세포내 사이토카인 (CD4+IL-17A+, CD4+IL-10 등)의 변화를 보여주며, 도 5c는 류마티스 관절염 동물 모델의 H&E, 톨루이딘 블루, 및 사프라닌 O 염색에 의한 조직학적 평가 결과를 보여주고, 도 5d는 각 그룹에서 계산된 염증지수 (inflammation score) 및 연골 미란 점수 (Cartilage damage score)를 통해 보여준다(*p<0.05, **p<0.01, ***p<0.001).5a to 5d show the results of comparing the disease control efficacy of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes and the mesenchymal stem cell exosomes in an arthritis animal model according to an embodiment of the present invention. Figure 5a shows the arthritis evaluation index of the group administered with the dedifferentiated stem cell-derived mesenchymal stem cell exo (iPSC-iMSC-Exo) and the mesenchymal stem cell exo (MSC-Exo), respectively, in the induction of the rheumatoid arthritis animal model. Figure 5b shows changes in CD4+T intracellular cytokines (CD4+IL-17A+, CD4+IL-10, etc.) in splenocytes of an animal model of rheumatoid arthritis confirmed by FACS, and Figure 5c shows rheumatoid arthritis Shows the histological evaluation results by H & E, toluidine blue, and safranin O staining of the arthritis animal model, Figure 5d is the inflammation index (inflammation score) and cartilage erosion score (Cartilage damage score) calculated in each group through (* p<0.05, ** p<0.01, *** p<0.001 ).
도 6a 내지 6d는 본 발명의 일 구체예에 따른 역분화 줄기세포 유래 중간엽 줄기세포 엑소좀의 농도에 따른 관절염 동물모델에서 질환 제어 효능을 확인한 결과를 보여준다. 도 6a는 류마티스 관절염 동물 모델의 유도에서 각각 역분화줄기세포 유래 중간엽 줄기세포 엑소좀(iPSC-iMSC-Exo)을 농도별로 투여한 그룹의 관절염 평가 지수를 보여주고, 도 6b는 FACS에 의해 확인된 류마티스 관절염 동물 모델의 비장세포 내 CD4+T 세포내 사이토카인 (CD4+IL-17A+, CD4+IL-10 등)의 변화를 보여주며, 도 6c는 류마티스 관절염 동물 모델의 H&E, 톨루이딘 블루, 및 사프라닌 O 염색에 의한 조직학적 평가 결과를 보여주고, 도 6d는 각 그룹에서 계산된 염증지수 및 연골 미란 점수를 통해 보여준다(*p<0.05, **p<0.01, ***p<0.001).6a to 6d show the results of confirming the disease control efficacy in the arthritis animal model according to the concentration of the dedifferentiated stem cell-derived mesenchymal stem cell exosomes according to an embodiment of the present invention. Figure 6a shows the arthritis evaluation index of the group administered with each concentration of immunized stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo) in the induction of an animal model of rheumatoid arthritis, Figure 6b is confirmed by FACS shows changes in cytokines (CD4+IL-17A+, CD4+IL-10, etc.) in splenocytes in an animal model of rheumatoid arthritis, FIG. 6c shows H&E, toluidine blue, and Shows the results of histological evaluation by safranin O staining, and FIG. 6d shows the inflammation index and cartilage erosion score calculated in each group (* p<0.05, ** p<0.01, *** p<0.001 ). ).
이하 본 발명을 실시예를 통하여 보다 상세하게 설명한다. 그러나, 이들 실시예는 본 발명을 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail through examples. However, these examples are for illustrative purposes only, and the scope of the present invention is not limited to these examples.
실시예 1. 정상인의 말초혈액 단핵세포로부터 역분화 줄기세포 (induced Pluripotent Stem Cells: iPSC) 제조Example 1. Preparation of induced Pluripotent Stem Cells (iPSC) from normal human peripheral blood mononuclear cells
기관 IRB를 승인받아 질환이 없는 정상인 기부자로부터 혈액을 수득하여 사용하였다. 단핵 세포를 분리하기 위하여 혈액을 2,000 rpm으로 30 분간 Ficoll Paque-PLUS (GE Healthcare)를 사용하여 원심분리하였다. 분리된 단핵 세포를 StemSpan cc110이 보충된 StemSpan-ACF (Stem Cell Technologies)에서 4일 동안 37℃ 및 5% CO2 조건에서 배양시켰다. 그 후, 비트로넥틴 (vitronectin) (Life Technologies, Invitrogen)이 코팅된 24-웰 플레이트에 배양된 단핵 세포를 접종하고, 제조사의 설명서에 따라 CytoTune®-iPS 2.0 Reprogramming 키트(Life Technologies, Invitrogen)의 센다이 바이러스 (Sendai virus)를 이용하여 역분화 줄기세포 (iPSC)를 제조하였다. 구체적으로, 형질 도입 후 3 일부터 21 일까지 세포를 37℃ 및 5% CO2 조건에서 배양하고, 12일 차에 신생 iPSC 콜로니를 개별적으로 골라 특성화(characterization)를 위해 확장시켰다. iPSC 콜로니가 형성될 때까지 배지를 매일 교체하였다. 수동으로 iPSC 콜로니를 피킹한 후, 비트로넥틴으로 코팅된 플레이트 중 TeSR-E8 배지 (Stem Cell Technologies)에서 배양하였다. 배양된 세포를 역분화줄기세포(iPSC)의 마커인 NANOG, SSEA-4, 및 TRA-181 발현에 대해 유동세포분석법(flow cytometry)으로 분석하여 역분화줄기세포로 확인하였다. Blood was obtained from a normal donor without disease with the approval of the institutional IRB and used. To isolate mononuclear cells, blood was centrifuged at 2,000 rpm for 30 minutes using a Ficoll Paque-PLUS (GE Healthcare). The isolated mononuclear cells were cultured in StemSpan-ACF (Stem Cell Technologies) supplemented with StemSpan cc110 at 37° C. and 5% CO 2 conditions for 4 days. Thereafter, the cultured mononuclear cells were inoculated in a 24-well plate coated with vitronectin (Life Technologies, Invitrogen), and Sendai of the CytoTune®-iPS 2.0 Reprogramming Kit (Life Technologies, Invitrogen) according to the manufacturer's instructions. IPSCs were prepared using a virus (Sendai virus). Specifically, from 3 to 21 days after transduction, cells were cultured at 37° C. and 5% CO 2 conditions, and on day 12, new iPSC colonies were individually picked and expanded for characterization. The medium was changed daily until iPSC colonies were formed. After manually picking iPSC colonies, they were cultured in TeSR-E8 medium (Stem Cell Technologies) in vitronectin-coated plates. Cultured cells were analyzed by flow cytometry for the expression of NANOG, SSEA-4, and TRA-181, which are markers of iPSCs, and identified as iPSCs.
실시예 2. 역분화 줄기세포 유래 중간엽 줄기세포 (mesenchymal stem cell) (iPSC-iMSC)의 제조 및 확인Example 2. Preparation and identification of mesenchymal stem cells (iPSC-iMSC) derived from dedifferentiated stem cells
2-1. 역분화 줄기세포 유래 중간엽 줄기세포의 제조2-1. Preparation of mesenchymal stem cells derived from dedifferentiated stem cells
실시예 1에서 제조된 역분화 줄기세포로부터 중간엽 줄기세포를 유도하기 위해, 플레이트에서 90% 합류(90% confluency)까지 배양된 역분화 줄기세포를 PBS(1 x Phosphate buffered saline)로 1회 세척하였다. 그 후, 플레이트에 1 mg/mL type IV Collagenase 용액을 3 mL 넣고 10분 동안 반응시켰다. TeSR-E8 배지 (Stem Cell Technologies)를 6 mL 첨가한 후 1100 rpm으로 3분 동안 원심분리하였다. 6-웰 ultra low attachment 디쉬(직경 100 mm)에 1 디쉬 당 2 웰 3 mL 배양액 20 % α-Minimum Eagle’s Medium (0.1 mM 비필수 아미노산 (100x) gibco, 1 mM L-글루타민, 0.1 mM 2-머캡토에탄올, 1% 페니실린, 1% 스트렙토마이신)으로 세포를 현탁한 후 b-FGF 1 ng/mL의 존재 하에 4일간 37℃ 및 5% CO2 조건에서 배양하여 분화를 유도하였다. In order to induce mesenchymal stem cells from the retrodifferentiated stem cells prepared in Example 1, the retrodifferentiated stem cells cultured on a plate to 90% confluency (90% confluency) were washed once with PBS (1 x Phosphate buffered saline). did After that, 3 mL of a 1 mg/mL type IV collagenase solution was added to the plate and reacted for 10 minutes. After adding 6 mL of TeSR-E8 medium (Stem Cell Technologies), it was centrifuged at 1100 rpm for 3 minutes. 2 wells 3 mL culture medium per dish in 6-well ultra low attachment dishes (100 mm diameter) 20% α-Minimum Eagle's Medium (0.1 mM non-essential amino acids (100x) gibco, 1 mM L-glutamine, 0.1 mM 2-mer After suspending the cells with captoethanol, 1% penicillin, 1% streptomycin), differentiation was induced by culturing at 37° C. and 5% CO 2 conditions for 4 days in the presence of 1 ng/mL of b-FGF.
분화 4 일 후, 배아체 (embryoid body: EB) 증식 (outgrowth)을 위해 1 % 젤라틴이 코팅된 100 mm 디쉬에 재조합 인간 (recombinant human: rh) TGF-β1로 자극하였다. 동일한 방법으로 격일마다 배양액을 교체하면서 젤라틴이 코팅된 100 mm 디쉬에서 90% 합류(90% confluency)까지 iPSC-MSC를 배양하였다.After 4 days of differentiation, embryoid body (EB) was stimulated with recombinant human (rh) TGF-β1 in 100 mm dishes coated with 1% gelatin for outgrowth. In the same manner, iPSC-MSCs were cultured until 90% confluency (90% confluency) in a gelatin-coated 100 mm dish while changing the culture medium every other day.
2-2. 역분화 줄기세포 유래 중간엽 줄기세포 마커 확인2-2. Identification of mesenchymal stem cell markers derived from dedifferentiated stem cells
실시예 2-1에서 역분화 줄기세포 유래 중간엽 줄기세포(iPSC-MSC)가 제조되었는지 마커를 통해 확인하였다.In Example 2-1, it was confirmed through a marker whether dedifferentiated stem cell-derived mesenchymal stem cells (iPSC-MSC) were prepared.
구체적으로, 역분화 줄기세포의 마커인 NANOG, SSEA-4 및 TRA-181 발현을 면역형광 방법으로 확인하고, 역분화 줄기세포 유래 중간엽 줄기세포를 분화 단계별로 세포 모양을 확인하였다. 또한, iPSC-MSC에 대해 MSC 마커로 알려져 있는 CD34, CD31, CD19, CD11 및 HALDR (음성 마커), 및 CD44, CD73, CD105 및 CD90 (양성 마커)의 발현 여부를 조사하였다. iPSC-MSC를 4주 정도 배양한 후 passage 5 이상인 세포에서 유세포 분석기로 확인하였다. Specifically, the expression of NANOG, SSEA-4 and TRA-181, which are markers of dedifferentiated stem cells, was confirmed by the immunofluorescence method, and the cell shape of dedifferentiated stem cell-derived mesenchymal stem cells was confirmed at each stage of differentiation. In addition, the expression of CD34, CD31, CD19, CD11 and HALDR (negative markers), and CD44, CD73, CD105 and CD90 (positive markers), known as MSC markers, were investigated for iPSC-MSCs. After culturing iPSC-MSCs for about 4 weeks, cells at passage 5 or higher were confirmed by flow cytometry.
그 결과, 도 1a에 나타낸 바와 같이, 실시예 2-1에서 사용된 역분화 줄기세포는 NANOG, SSEA-4 및 TRA-181을 발현함을 확인하였고, 도 1b에 표시된 바와 같이, 역분화 줄기세포로부터 중간엽 줄기세포로 분화되는 것을 세포 모양을 통해 확인하였다. As a result, as shown in Figure 1a, it was confirmed that the dedifferentiated stem cells used in Example 2-1 express NANOG, SSEA-4 and TRA-181, and as shown in Figure 1b, the dedifferentiated stem cells Differentiation into mesenchymal stem cells was confirmed through cell shape.
도 1c는 유세포 분석기로 확인된 iPSC-MSC에 의한 MSC 마커의 발현을 보여준다. 실시예 2-1에서 제조된 역분화 줄기세포 유래 중간엽 줄기세포는 MSC의 음성 마커인 CD34, CD31, CD19, CD11 및 HALDR을 동형 대조군 (isotype control)과 유사한 양으로 발현하였으나, 양성 마커인 CD44, CD73, CD105 및 CD90는 모두 동형 대조군 대비 유의하게 많은 양을 발현하여, 실시예 2-1에서 역분화 줄기세포 유래 중간엽 줄기세포가 형성되었음을 확인할 수 있었다.1C shows the expression of MSC markers by iPSC-MSCs confirmed by flow cytometry. The retrodifferentiated stem cell-derived mesenchymal stem cells prepared in Example 2-1 expressed CD34, CD31, CD19, CD11 and HALDR, which are negative markers of MSC, in similar amounts as the isotype control, but CD44, a positive marker , CD73, CD105 and CD90 all expressed significantly higher amounts compared to the isotype control, confirming that dedifferentiated stem cell-derived mesenchymal stem cells were formed in Example 2-1.
실시예 3. 중간엽 줄기세포 엑소좀 (MSC-Exo) 및 역분화 줄기세포 유래 중간엽 줄기세포로부터 엑소좀 (iPSC-MSC-Exo) 분리 및 확인Example 3. Isolation and identification of mesenchymal stem cell exo-some (MSC-Exo) and dedifferentiated stem cell-derived mesenchymal stem cell exo-some (iPSC-MSC-Exo)
3-1. 중간엽 줄기세포 엑소좀 및 역분화 줄기세포 유래 중간엽 줄기세포으부터 엑소좀 분리3-1. Isolation of exosomes from mesenchymal stem cell exosomes and dedifferentiated stem cell - derived mesenchymal stem cells
제대혈 유래 MSC 및 iPSC-MSC를 각각 100 mm 디쉬에서 100% 합류까지 배양한 상태로 SF-DMEM (serum free-Dulbecco Modified Eagle Medium) 5 mL를 첨가하여 37℃ 및 5% CO2 조건에서 48 시간 동안 배양하였다. 배양액을 4℃ 및 300 g 조건으로 10분 동안 원심분리하였다. 수득된 상층액을 다시 4℃ 및 2000 g 조건으로 20분 동안 원심분리하였다. 수득된 상층액을 고속 원심분리기 (Hitachi KoKi #S306352A, Japan)로 10,000 g 조건에서 30 분 동안 원심분리시켜 세포 파쇄물 (cell debris)을 제거하였다. 상층액을 0.22 ㎛ 필터를 통해 여과시킨 후 초고속 원심분리기 (Thermo Scientific Fiberlite F50L-8x39 Rotor, Thermo Fisher Scientific)에서 100,000 g 조건으로 120분 동안 원심분리하였다. 원심분리기로부터 튜브를 꺼내면서 엑소좀이 있는 위치인 펠렛 부분을 미리 표시한 후, 펠렛을 제외한 나머지를 반대편으로 모두 제거하고, 남은 펠렛을 PBS로 현탁하여 사용하기 전까지 -80℃로 보관하였다.Cord blood-derived MSCs and iPSC-MSCs were each cultured in a 100 mm dish to 100% confluence, and 5 mL of SF-DMEM (serum free-Dulbecco Modified Eagle Medium) was added thereto at 37°C and 5% CO 2 conditions for 48 hours. cultured. The culture medium was centrifuged for 10 minutes at 4 °C and 300 g conditions. The obtained supernatant was again centrifuged at 4°C and 2000 g conditions for 20 minutes. The obtained supernatant was centrifuged for 30 minutes at 10,000 g with a high-speed centrifuge (Hitachi KoKi #S306352A, Japan) to remove cell debris. The supernatant was filtered through a 0.22 μm filter and then centrifuged for 120 minutes at 100,000 g in an ultra-high speed centrifuge (Thermo Scientific Fiberlite F50L-8x39 Rotor, Thermo Fisher Scientific). After taking out the tube from the centrifuge, the pellet part, which is the location of the exosomes, was marked in advance, all the rest except the pellet was removed to the opposite side, and the remaining pellet was suspended in PBS and stored at -80°C until use.
3-2. 역분화 줄기세포 유래 중간엽 줄기세포 엑소좀 (iPSC-iMSC-Exo)의 특성 및 마커 확인 3-2. Identification of characteristics and markers of dedifferentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo)
분리된 엑소좀의 크기 및 균일한 입자 분포 여부를 확인하기 위해, 실시예 3-1에 의해 분리된 엑소좀을 PBS에 희석하여 NanoSight (Malvern instruments Ltd, Malvern, UK) 기기로 6 회 연속 측정하였다. 또한, 분리된 엑소좀을 투과 전자 현미경 (TEM)으로 촬영하였다. 이어서, 분리된 것이 엑소좀이 맞는지 확인하기 위해, 웨스턴 블롯 방법을 이용하여 엑소좀 마커로 알려진 TSG101 (Santa Cruz Biotechnology), CD9 (Santa Cruz Biotechnology) 및 CD63 (Santa Cruz Biotechnology)를 확인하였다.In order to confirm the size and uniform particle distribution of the separated exosomes, the exosomes separated by Example 3-1 were diluted in PBS and measured six times consecutively with a NanoSight (Malvern instruments Ltd, Malvern, UK) instrument. . In addition, the isolated exosomes were photographed with a transmission electron microscope (TEM). Then, in order to confirm whether the isolated exosome is correct, TSG101 (Santa Cruz Biotechnology), CD9 (Santa Cruz Biotechnology) and CD63 (Santa Cruz Biotechnology), known as exosome markers, were identified using a Western blot method.
그 결과, 도 2a에 나타낸 바와 같이, 역분화 줄기세포 유래 중간엽 줄기세포로부터 분리된 물질의 크기가 NanoSight에 의해 엑소좀의 크기로 알려진 100 내지 200 nm임을 확인하여, 상기 물질이 엑소좀 (iPSC-iMSC-Exo)임을 알 수 있었다. 도 2b는 투과 전자 현미경을 이용하여 엑소좀을 확인한 결과를 보여준다.As a result, as shown in Figure 2a, it was confirmed that the size of the material isolated from the dedifferentiated stem cell-derived mesenchymal stem cells is 100 to 200 nm, which is known as the size of the exosome by NanoSight, and the material is the exosome (iPSC). -iMSC-Exo). Figure 2b shows the result of confirming the exosomes using a transmission electron microscope.
또한, 도 2c는 역분화 줄기세포 유래 중간엽 줄기세포에서 분리된 엑소좀 (hiPSC-MSCs-Exo)이 대조군(Control: SF-DMEM 배지) 및 역분화 줄기세포 유래 중간엽 줄기세포 그 자체 (hiPSC-MSC)보다 엑소좀의 마커로 알려진 TSG101, CD9 및 CD63를 유의하게 많이 발현한다는 것을 확인한 것이다. 이를 통해 실시예 3-1에서 분리된 물질이 엑소좀 (iPSC-iMSC-Exo)임을 알 수 있었다. In addition, Figure 2c shows that the exosomes (hiPSC-MSCs-Exo) isolated from dedifferentiated stem cell-derived mesenchymal stem cells are a control (Control: SF-DMEM medium) and dedifferentiated stem cell-derived mesenchymal stem cells themselves (hiPSCs). -MSC), it was confirmed that TSG101, CD9 and CD63, which are known as markers of exosomes, are expressed significantly more. Through this, it was confirmed that the material isolated in Example 3-1 was an exosome (iPSC-iMSC-Exo).
실시예 4. Th0 세포에서 역분화 줄기세포 유래 중간엽 줄기세포로부터 분리된 엑소좀 (iPSC-MSC-Exo)의 면역 조절능 확인Example 4. Immune regulation ability of exosomes (iPSC-MSC-Exo) isolated from dedifferentiated stem cell-derived mesenchymal stem cells from T h 0 cells
실시예 3에서 수득된 iPSC-MSC-Exo의 면역 조절능을 각각 확인하였다. The immunomodulatory ability of iPSC-MSC-Exo obtained in Example 3 was confirmed, respectively.
정상인의 말초혈액으로부터 수득된 단핵 세포를 하기와 같이 Th0 세포로 분화시켰다. 구체적으로, Th0 세포로 분화시킬 단핵 세포를 10% 소 태아 혈청 (fetal calf serum: FCS), 100 U/mL 페니실린, 100 mg/mL 스트렙토마이신, 및 2 mM L-글루타민이 보충된 RPMI 1640 배지에 접종하고, 항-CD3 (1 ㎍/mL; BD Biosciences) 및 항-CD28 (1 ㎍/mL; BD Biosciences)로 처리하고 실시예 3에서 제조된 iPSC-iMSC-Exo(0, 0.1, 1, 및 10 ㎍)와 37℃ 및 5% CO2 조건에서 48 시간 동안 공배양시켰다. 그 후, FACS를 이용하여 Th0 세포에서 생성되는 염증유도성 사이토카인인 IL-17 및 IFN-γ, 및 면역조절 T 세포 (Treg)의 마커인 CD25+Foxp3+ 발현을 조사하였다. 또한, 각 배양액에서 염증유도성 사이토카인 IL-17 및 염증억제성 사이토카인 IL-10의 함량을 분석하였다. 구체적으로, FACS 및 샌드위치 (sandwich) ELISA를 이용하여 사이토카인 함량 및 마커 발현을 분석하였다. Mononuclear cells obtained from normal human peripheral blood were differentiated into T h 0 cells as follows. Specifically, the mononuclear cells to be differentiated into T h 0 cells were transformed into RPMI 1640 supplemented with 10% fetal calf serum (FCS), 100 U/mL penicillin, 100 mg/mL streptomycin, and 2 mM L-glutamine. iPSC-iMSC-Exo prepared in Example 3 (0, 0.1, 1) inoculated into medium, treated with anti-CD3 (1 μg/mL; BD Biosciences) and anti-CD28 (1 μg/mL; BD Biosciences) , and 10 μg) and 37° C. and 5% CO 2 conditions were co-cultured for 48 hours. Then, using FACS, IL-17 and IFN-γ, which are pro-inflammatory cytokines produced in T h 0 cells, and CD25+Foxp3+ expression, a marker of immunoregulatory T cells (T reg ), were investigated. In addition, the contents of the inflammation-inducing cytokine IL-17 and the anti-inflammatory cytokine IL-10 in each culture were analyzed. Specifically, cytokine content and marker expression were analyzed using FACS and sandwich ELISA.
FACS를 위해 단일 클론 항체 항-CD4-PE/Cy7 (RPA-T4, IgG1, BioLegend, San Diego, CA, USA) 및 항-CD25-APC (M-A251, IgG1, κ 이소형, BD Biosciences)를 이용하고, 세포 내 염색법으로 세포를 세척한 다음, 투과성으로 만들고 단일 클론 항체 항-IL-17-PE (eBio64dec17, IgG1, κ; eBioscience, San Diego, CA, USA), 항-IFN-γ-FITC (4S.B3, IgG1, κ; eBioscience), 항-IL-10-APC (JES3-19F1, IgG2a, κ; Biolegend), 및 항-Foxp3-FITC (PCH101, IgG2a, κ; eBioscience) 항체와 함께 암실 상태로 4℃에서 30분간 인큐베이션시켰다. 또한, 대조군은 같은 종류의 형광이 표지되고 항원 특이성이 없는 동종의 면역글로불린을 동형 항체로 염색하였다. 유세포 분석기(FACS Calibur; Becton Dickinson, San Diego, CA)로 세포 내 사이토카인을 분석하였다. Monoclonal antibodies anti-CD4-PE/Cy7 (RPA-T4, IgG1, BioLegend, San Diego, CA, USA) and anti-CD25-APC (M-A251, IgG1, κ isotype, BD Biosciences) were administered for FACS. cells were used, washed by intracellular staining, then permeabilized and monoclonal antibody anti-IL-17-PE (eBio64dec17, IgG1, κ; eBioscience, San Diego, CA, USA), anti-IFN-γ-FITC (4S.B3, IgG1, κ; eBioscience), anti-IL-10-APC (JES3-19F1, IgG2a, κ; Biolegend), and anti-Foxp3-FITC (PCH101, IgG2a, κ; eBioscience) antibodies in the dark. It was incubated for 30 minutes at 4°C. In addition, the control group was stained with the same type of fluorescence-labeled immunoglobulin of the same type without antigen specificity with an isotype antibody. Intracellular cytokines were analyzed by flow cytometry (FACS Calibur; Becton Dickinson, San Diego, CA).
또한, 샌드위치 ELISA를 위해 먼저 샌드위치 ELISA용 96 웰 플레이트 (NUNC, Denmark)에 단일 클론성 IL-17, IFN-γ, IL-10 및 IL-2 항체(R&D systems, USA)를 각각 4 ㎍/mL로 웰당 50 ㎕ 넣고 4℃에서 밤새 반응시킨 다음, 차단 용액 (1% (w/v) BSA/PBS (Phosphate-Buffered Saline) 및 0.05% (v/v) tween 20/PBS)을 웰당 200 ㎕ 첨가하여 실온에서 2 시간 동안 반응시켰다. 대조군으로는 재조합 IL-17, IFN-γ, IL-10 및 IL-2 (R&D, USA)를 이용하여 각각 78 pg/mL, 156 pg/mL, 312 pg/mL, 625 pg/mL, 1250 pg/mL, 2500 pg/mL, 5000 pg/mL 농도를 측정하였다. In addition, for sandwich ELISA, monoclonal IL-17, IFN-γ, IL-10 and IL-2 antibodies (R&D systems, USA) were first added to a 96-well plate (NUNC, Denmark) for sandwich ELISA at 4 μg/mL, respectively. 50 μl per well of the furnace was added and reacted at 4° C. overnight, and then 200 μl of blocking solution (1% (w/v) BSA/PBS (Phosphate-Buffered Saline) and 0.05% (v/v) tween 20/PBS) was added per well. and reacted at room temperature for 2 hours. As a control group, recombinant IL-17, IFN-γ, IL-10 and IL-2 (R&D, USA) were used to 78 pg/mL, 156 pg/mL, 312 pg/mL, 625 pg/mL, and 1250 pg, respectively. /mL, 2500 pg/mL, 5000 pg/mL concentrations were measured.
표준 시료와 함께 측정할 엑소좀과 공배양하여 수득된 Th0 세포 배양액을 웰당 50 ㎕ 첨가하고 실온에서 2 시간 동안 반응시켰다. 반응 용기를 세척용액 (0.05% (v/v) Tween 20/PBS)으로 4 회 세척하고 비오티닐화된 (biotinylated) IL-17, IFN-γ, IL-10 및 IL-2 항체를 200 ng/mL로 희석하여 웰당 50 ㎕ 첨가한 후, 실온에서 2 시간 동안 반응시키고 세척 용액으로 4 회 세척하였다. 50 μl of a T h 0 cell culture solution obtained by co-culture with an exosome to be measured together with a standard sample per well was added and reacted at room temperature for 2 hours. The reaction vessel was washed 4 times with a washing solution (0.05% (v/v) Tween 20/PBS) and biotinylated IL-17, IFN-γ, IL-10 and IL-2 antibodies were added at 200 ng/ After dilution to mL, 50 μl per well was added, followed by reaction at room temperature for 2 hours, and washing with a washing solution 4 times.
마지막으로, 엑스트라비딘-알칼라인 포스파타아제 컨쥬게이트 (Extravidin-Alkaline phosphatase conjugate) (SIGMA, USA, #E2636)를 1:2,000으로 희석하여 웰당 50 ㎕ 첨가하고 실온에서 2 시간 반응시킨 후 세척하고, 포스페이트 디소듐염 헥사하이드레이트 (phosphate disodium salt hexahydrate) (PNPP, Fluka)/디에탄올아민 (Diethanolamine) 용액 (DEA, 97 mL, NaN3 0.2 g, MgCl2ㆍH2O 0.1 g, 1차 증류수 800 mL)을 1 mg/mL 농도로 용해하여 웰당 50 ㎕ 첨가하고 30 분 후, 0.2 M NaOH로 반응을 멈춘 뒤 405 nm 파장에서 흡광도를 측정하였다.Finally, 50 μl per well of an extravidin-alkaline phosphatase conjugate (SIGMA, USA, #E2636) was diluted 1:2,000, reacted at room temperature for 2 hours, washed, and phosphate phosphate disodium salt hexahydrate (PNPP, Fluka)/Diethanolamine solution (DEA, 97 mL, NaN 3 0.2 g, MgCl 2 H 2 O 0.1 g, primary distilled water 800 mL) was dissolved at a concentration of 1 mg/mL, 50 μl per well was added, and after 30 minutes, the reaction was stopped with 0.2 M NaOH, and absorbance was measured at a wavelength of 405 nm.
그 결과가 도 3a 내지 3c에 도시된다. 도 3a 및 3b는 각각 Th0 세포에서 생성된 IL-17 및 IFN-γ의 양과 면역조절 T 세포(Treg)에서 생성되는 CD25+Foxp3+ 발현을 FACS로 분석한 결과를 나타내고, 도 3c는 배양액 중 IL-17, IFN-γ 및 IL-10을 ELISA로 측정한 결과를 나타낸다. 역분화 줄기세포 유래 중간엽 줄기세포 엑소좀 (iPSC-iMSC-Exo)은 염증성 사이토카인인 IL-17 및 IFN-γ의 생성을 억제하는 반면 면역조절 T세포 (Treg)의 활성에는 영향을 미치지 않음을 확인하였다. 또한, iPSC-iMSC-Exo는 세포 배양액에서 염증유도성 사이토카인인 IL-17 및 IFN-γ의 생성을 억제시키는 반면 염증억제성 사이토카인인 IL-10의 생성을 더욱 증가시킴을 확인하였다. The results are shown in Figures 3a to 3c. Figures 3a and 3b show the results of FACS analysis of the amounts of IL-17 and IFN-γ produced in T h 0 cells and CD25+Foxp3+ expression produced in immunoregulatory T cells (Tregs), respectively, and Figure 3c is in the culture medium. The results of measuring IL-17, IFN-γ and IL-10 by ELISA are shown. Immunomodulatory stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo) inhibit the production of inflammatory cytokines IL-17 and IFN-γ, while not affecting the activity of immunoregulatory T cells (T reg ). confirmed that it is not. In addition, it was confirmed that iPSC-iMSC-Exo inhibited the production of IL-17 and IFN-γ, which are pro-inflammatory cytokines, while further increasing the production of IL-10, an anti-inflammatory cytokine, in cell culture medium.
실시예 5. Th17 세포에서 역분화 줄기세포 유래 중간엽 줄기세포로부터 분리된 엑소좀 (iPSC-iMSC-Exo)의 면역 조절능 확인Example 5. Confirmation of immune modulatory ability of exosomes (iPSC-iMSC-Exo) isolated from dedifferentiated stem cell-derived mesenchymal stem cells in T h 17 cells
실시예 3에서 수득된 iPSC-iMSC-Exo 및 MSC-Exo의 면역 조절능을 각각 확인하였다. 구체적으로, 정상인의 말초혈액으로부터 수득된 단핵 세포를 Th17 세포로 분화시키기 위해, 항-CD3 (1 ㎍/mL; BD Biosciences), 항-CD28 (1 ㎍/mL; BD Biosciences), IL-1β (20 ng/mL; R&D Systems, Minneapolis, MN, USA), IL-6 (20 ng/mL; R&D Systems), IL-23 (20 ng/mL; R&D Systems), IFN-γ에 대한 중화 항체 (IFN-γ; 2 ㎍/mL; R&D Systems) 및 IL-4에 대한 중화항체 (2 ㎍/mL; R&D Systems)로 처리하고, Th0 세포 대신 Th17를 이용한 것 외에는 실시예 4와 동일하게 실험을 수행하였다.The immunomodulatory ability of iPSC-iMSC-Exo and MSC-Exo obtained in Example 3 was confirmed, respectively. Specifically, in order to differentiate mononuclear cells obtained from normal human peripheral blood into T h 17 cells, anti-CD3 (1 μg/mL; BD Biosciences), anti-CD28 (1 μg/mL; BD Biosciences), IL- Neutralizing antibody against 1β (20 ng/mL; R&D Systems, Minneapolis, MN, USA), IL-6 (20 ng/mL; R&D Systems), IL-23 (20 ng/mL; R&D Systems), IFN-γ (IFN-γ; 2 μg/mL; R&D Systems) and a neutralizing antibody against IL-4 (2 μg/mL; R&D Systems), and Example 4 except that T h 17 was used instead of T h 0 cells The same experiment was performed.
그 결과가 도 4a 내지 4c에 도시된다. 도 4a 및 4b는 각각 Th17 세포에서 생성된 IL-17 및 면역조절 T 세포(Treg)에서 생성되는 CD25+Foxp3+ 발현을 FACS로 분석한 결과를 나타내고, 도 4c는 배양액 중 IL-17, IL-10, 및 IL-2를 ELISA로 측정한 결과를 나타낸다. 역분화줄기세포 유래 중간엽 줄기세포 엑소좀 (iPSC-iMSC-Exo)은 염증유도성 사이토카인인 IL-17의 생성을 억제하는 반면, 면역조절 T세포 (Treg)의 활성에는 영향을 미치지 않음을 확인할 수 있었다. 또한, iPSC-iMSC-Exo는 세포 배양액에서 염증유도성 사이토카인인 IL-17 및 IL-2의 생성을 감소시키고 염증억제성 사이토카인인 IL-10의 생성을 더욱 증가시킴을 알 수 있었다.The results are shown in Figures 4a to 4c. 4a and 4b show the results of FACS analysis of the expression of IL-17 generated in Th17 cells and CD25+Foxp3+ generated in immunoregulatory T cells (Tregs), respectively, and FIG. 4c shows IL-17 and IL-10 in the culture medium. , and IL-2 measured by ELISA are shown. Inversely differentiated stem cell-derived mesenchymal stem cell exosomes (iPSC-iMSC-Exo) inhibit the production of IL-17, an inflammation-inducing cytokine, but do not affect the activity of immunoregulatory T cells (T reg ) was able to confirm In addition, it was found that iPSC-iMSC-Exo decreased the production of IL-17 and IL-2, which are pro-inflammatory cytokines, and further increased the production of IL-10, an anti-inflammatory cytokine, in cell culture medium.
실시예 6. 역분화 줄기세포 유래 중간엽 줄기세포로부터 분리된 엑소좀 (iPSC-iMSC-Exo)과 중간엽 줄기세포 엑소좀 (MSC-Exo)의 류마티스 관절염 동물 모델 질환 제어 효과 비교 Example 6. Comparison of disease control effects in rheumatoid arthritis animal model of exosomes isolated from retrodifferentiated stem cell-derived mesenchymal stem cells (iPSC-iMSC-Exo) and mesenchymal stem cell exosomes (MSC-Exo)
6-1. 평균 관절 지수를 통한 류마티스 관절염 치료 효과 확인6-1. Confirmation of treatment effect for rheumatoid arthritis through mean joint index
실시예 3에서 수득된 역분화 줄기세포 유래 중간엽 줄기세포로부터 분리된 엑소좀(iPSC-iMSC-Exo)이 류마티스 관절염에 효과를 나타내는지 확인하기 위해 동물 모델 실험을 수행하였다.In order to confirm whether the exosomes (iPSC-iMSC-Exo) isolated from the dedifferentiated stem cell-derived mesenchymal stem cells obtained in Example 3 have an effect on rheumatoid arthritis, an animal model experiment was performed.
구체적으로, 류마티스 관절염 동물 모델(Collagen induced arthritis: CIA)을 제조하기 위해, 6 내지 7 주령인 수컷 DBA/1J 마우스에 CII(type II collagen) 100 ㎍을 CFA(Complete Freund's Adjuvant)와 1:1 비율로 혼합하여 꼬리에 피하주사하였다(1차 부스팅). 2주 후 CII 100 ㎍을 IFA(Incomplete Freund's Adjuvant)와 1:1 비율로 혼합하여 뒷다리 족근골에 피하주사하여(2차 부스팅) 류마티스 관절염을 유도하였다. 류마티스 관절염 동물 모델을 4개의 그룹, CIA 대조군(n=4), iPSC-iMSC-Exo(50 ㎍)(n=4), iPSC-iMSC-Exo(100 ㎍)(n=4) 및 MSC-Exo(100 ㎍)(n=4)로 나누고, 그룹별로 iPSC-iMSC-Exo 또는 MSC-Exo를 1차 부스팅 1일 전 및 2차 부스팅 일에 투여하여 총 2회 투여하고, 그룹별 질환 발달 차이를 확인하였다. 질환 발달의 정도는 하기의 류마티스 관절염 평가를 통해 확인하였다. Specifically, in order to prepare an animal model of rheumatoid arthritis (CIA), 100 μg of type II collagen (CII) was mixed with Complete Freund's Adjuvant (CFA) at a 1:1 ratio in 6 to 7-week-old male DBA/1J mice. was mixed and injected subcutaneously into the tail (1st boosting). After 2 weeks, 100 μg of CII was mixed with IFA (Incomplete Freund's Adjuvant) in a 1:1 ratio and injected subcutaneously into the tarsal bone of the hind leg (secondary boosting) to induce rheumatoid arthritis. Rheumatoid arthritis animal models were analyzed in 4 groups, CIA control (n=4), iPSC-iMSC-Exo (50 μg) (n=4), iPSC-iMSC-Exo (100 μg) (n=4) and MSC-Exo (100 μg) (n=4), iPSC-iMSC-Exo or MSC-Exo for each group was administered 1 day before the 1st boosting and on the 2nd boosting day, a total of 2 doses were administered, and the difference in disease development by group Confirmed. The degree of disease development was confirmed through the following rheumatoid arthritis evaluation.
류마티스 관절염 평가는 Rosoliniec 등에 의한 평균 관절 점수 (mean arthritic score) (Coligan JE, Kruisbeek AM, Matqulies DH, Shevach EM, Strober W: Collagen-induced arthritis. Current protocols in immunology vol 3;15.5.1-19, 1996)에 기초하였다. 구체적으로, 각 마우스에 대해 아래의 척도에 따라 매긴 점수(Arthritis score)를 합하여 3으로 나눈 평균치를 얻고, 다시 각 마우스에서 3 명의 관찰자가 얻은 수치를 합산하여 나눈 평균치를 사용한다.The evaluation of rheumatoid arthritis was performed using the mean arthritic score by Rosoliniec et al. (Coligan JE, Kruisbeek AM, Matqulies DH, Shevach EM, Strober W: Collagen-induced arthritis. Current protocols in immunology vol 3;15.5.1-19, 1996 ) was based on Specifically, for each mouse, the average value divided by 3 is obtained by summing the scores (Arthritis score) according to the scale below, and the average value obtained by adding up the values obtained by three observers in each mouse is used.
<평가 기준> <Evaluation criteria>
0점: 부종이나 종창이 없음. 0 points: No edema or swelling.
1점: 발 또는 발목관절에 국한된 경미한 부종과 발적이 있음Score 1: Mild swelling and redness localized to the foot or ankle joint.
2점: 발목관절에서 족근골(metatarsal)에 걸친 경미한 부종과 발적이 있음Score 2: Mild swelling and redness from the ankle joint to the metatarsal.
3점: 발목관절에서 족근골에 걸친 중등도의 부종과 발적이 있음Score 3: Moderate swelling and redness from the ankle joint to the tarsal bone.
4점: 발목에서 다리 전체에 걸쳐 부종과 발적이 있음4 points: swelling and redness from the ankle to the entire leg
도 5a 및 도 6a에 도시된 바와 같이, 엑소좀을 투여하지 않은 대조군(CIA control) 및 iPSC-iMSC-Exo 투여군에서 MSC-Exo 투여군과 비교하여 마우스 관절염 평가 지수가 모두 감소되었고, iPSC-iMSC-Exo (50㎍)과 iPSC-iMSC-Exo (100㎍) 투여군을 비교하여 농도의존적으로 평균 관절지수가 유의하게 낮아짐을 확인하였다. 즉, iPSC-MSC-Exo는 류마티스 관절염 동물 모델에서 류마티스 관절염을 치료하는 데 우수한 효과가 있음을 알 수 있었다.As shown in FIGS. 5A and 6A , in the control group (CIA control) and the iPSC-iMSC-Exo administration group not administered with exosomes, compared with the MSC-Exo administration group, all of the mouse arthritis evaluation indices were reduced, and iPSC-iMSC- Exo (50㎍) and iPSC-iMSC-Exo (100㎍) administration group were compared, and it was confirmed that the mean joint index significantly decreased in a concentration-dependent manner. That is, it was found that iPSC-MSC-Exo had an excellent effect in treating rheumatoid arthritis in an animal model of rheumatoid arthritis.
6-2. 면역학적 평가를 통한 류마티스 관절염 치료 효과 확인6-2. Confirmation of treatment effect for rheumatoid arthritis through immunological evaluation
실시예 6-1의 류마티스 관절염 동물 모델을 이용하여, 질환의 추가적인 평가를 위해 마우스의 비장세포 내 사이토카인 (IL-17, TNF-a, IL-1b, IL-10, IL-4 등)의 변화를 실시예 4에 기재된 바와 같이 ELISA를 이용해 확인하였다. Using the rheumatoid arthritis animal model of Example 6-1, cytokines (IL-17, TNF-a, IL-1b, IL-10, IL-4, etc.) in mouse splenocytes for further evaluation of disease Changes were confirmed using ELISA as described in Example 4.
도 5b 및 도 6b는 FACS를 통해 측정된 류마티스 관절염 동물 모델 마우스의 비장세포 내 CD4+T 세포내 사이토카인 (CD4+IL-17A+, CD4+IL-10 등)의 변화를 보여준다. CIA 그룹과 iPSC-iMSC-Exo, MSC-Exo 투여 그룹 간에는 유의성 있는 차이가 없었으나, iPSC-iMSC-Exo 100㎍ 투여 그룹과 iPSC-iMSC-Exo 50㎍ 투여 그룹 간에는 염증성 T세포인 CD4+IL-17A+ (%) 세포가 iPSC-iMSC-Exo 100㎍ 투여 그룹에서 의미있게 감소하였다. 5B and 6B show changes in CD4+T intracellular cytokines (CD4+IL-17A+, CD4+IL-10, etc.) in splenocytes of rheumatoid arthritis animal model mice measured by FACS. There was no significant difference between the CIA group and the iPSC-iMSC-Exo and MSC-Exo groups, but between the iPSC-iMSC-Exo 100㎍ group and the iPSC-iMSC-Exo 50㎍ group, the inflammatory T cells, CD4+IL- 17A+ (%) cells were significantly reduced in the iPSC-iMSC-Exo 100 μg administration group.
류마티스 관절염 동물 모델에서의 결과는 iPSC-iMSC-Exo 투여 그룹이 MSC-Exo 투여 그룹과 비교하여 염증유도성 사이토카인(CD4+IL-17+)의 생성을 억제하고, 염증억제성 사이토카인(CD4+IL-10+)의 생성을 유지한다는 것을 보여주었고, iPSC-iMSC-Exo 50㎍, iPSC-iMSC-Exo 100㎍ 두 그룹을 비교한 경우에도, 통계적으로 유의하게 염증유도성 사이토카인 (CD4+IL-17+)의 생성을 억제하고, 염증억제성 사이토카인 (CD4+IL-10+)의 생성을 유지한다는 것을 확인할 수 있었다. The results in the rheumatoid arthritis animal model showed that the iPSC-iMSC-Exo administration group suppressed the production of inflammation-inducing cytokines (CD4+IL-17+), and the anti-inflammatory cytokines (CD4) compared to the MSC-Exo administration group. +IL-10+), and even when comparing the two groups iPSC-iMSC-Exo 50㎍, iPSC-iMSC-Exo100㎍, it was statistically significantly It was confirmed that it inhibits the production of IL-17+) and maintains the production of anti-inflammatory cytokines (CD4+IL-10+).
6-3. 관절 조직 슬라이드를 이용한 류마티스 관절염 동물모델에 대한 효능 평가6-3. Efficacy evaluation for rheumatoid arthritis animal model using joint tissue slides
실시예 6-1의 류마티스 관절염 동물 모델을 이용하여, 질환의 추가적인 평가를 위해 마우스를 2차 부스팅 5주 후에 희생시키고 각각의 마우스의 다리 관절을 적출하여 10% 포르말린에 6일 동안 고정하고, Calci-clear rapid 용액에서 7시간 동안 탈회를 진행하였다. 10% 포르말린에서 2일 동안 또는 10% 포르말린에 6일 고정 후 Calci-clear rapid에 7시간 탈회된 조직은 아래와 같은 순서로 탈수, 투명화 및 파라핀을 침투시켜 조직 처리하였다.Using the rheumatoid arthritis animal model of Example 6-1, mice were sacrificed 5 weeks after the second boosting for further evaluation of the disease, and the leg joints of each mouse were excised and fixed in 10% formalin for 6 days, Calci -Decalcification was performed in a clear rapid solution for 7 hours. After fixation in 10% formalin for 2 days or in 10% formalin for 6 days, demineralized tissue was processed in calci-clear rapid for 7 hours by dehydration, clearing and paraffin infiltration in the following order.
No.No. 용기(Jar)Jar 시간hour
1One 흐르는 물(tap water) tap water 1 시간1 hours
22 70% EtOH70% EtOH 1 시간1 hours
33 80% EtOH80% EtOH 1 시간1 hours
44 90% EtOH90% EtOH 1 시간1 hours
55 95% EtOH # 195% EtOH #1 1 시간1 hours
66 95% EtOH # 295% EtOH #2 1 시간1 hours
77 100% EtOH # 1100% EtOH #1 1 시간1 hours
88 100% EtOH # 2100% EtOH #2 1 시간1 hours
99 크실렌 # 1 Xylene #1 1 시간1 hours
1010 크실렌 # 2 Xylene #2 1 시간1 hours
1111 파라핀 # 1 Paraffin #1 2 시간2 hours
1212 파라핀 # 2 Paraffin #2 2 시간2 hours
전술된 조직 처리가 끝난 조직은 파라핀에 포매하여 블록으로 제작하였다. 관절조직 염색을 수행하기 위하여 조직을 마이크로톰(microtome)을 이용하여 4㎛으로 절편하였다. 각 슬라이드는 1장의 슬라이드 당 한 마리의 조직 1개가 되도록 제작하였다. 헤마톡실린 & 에오신(Hematoxylin & Eosin) 염색을 위해 파라핀으로 고정된 조직 슬라이드를 65℃ 오븐에 넣어 1시간 동안 파라핀을 녹인 후 조직의 탈파라핀화와 수화를 위해 NeoClear 용액과 각기 다른 농도의 에탄올에 5분씩 순차적으로 침지하였다. 헤마톡실린 염색을 10초 진행 후, 슬라이드에 직접 닿지 않도록 흐르는 물(tap water)에 10분간 담궈 염색에 사용한 헤마톡실린 잔여물을 제거했다. Eosin 채널에 슬라이드를 침지하여 10분간 염색을 진행했다. 탈수를 위해 각기 다른 농도의 에탄올과 NeoClear 용액에 순차적으로 침지 후 마운팅(mounting)을 수행하고 하루 이상 서늘한 곳에서 건조시킨 후, 광학현미경을 통하여 조직을 확인하였다.After the above-described tissue treatment, the tissue was embedded in paraffin and manufactured as a block. In order to perform joint tissue staining, the tissue was sectioned into 4 μm using a microtome. Each slide was prepared so that there was one tissue per slide. For hematoxylin & eosin staining, paraffin-fixed tissue slides are placed in an oven at 65°C to melt paraffin for 1 hour, and then in NeoClear solution and different concentrations of ethanol for tissue deparaffinization and hydration. They were sequentially immersed for 5 minutes. After hematoxylin staining was performed for 10 seconds, the hematoxylin residue used for staining was removed by immersing the slide in tap water for 10 minutes to avoid direct contact with the slide. The slide was immersed in the Eosin channel and stained for 10 minutes. For dehydration, sequentially immersed in ethanol and NeoClear solution of different concentrations, mounting was performed, and after drying in a cool place for more than a day, the tissue was checked through an optical microscope.
사프라닌 O(Safranin O) 염색을 위해 바이게르트 철 헤마톡실린 용액(Weigert's iron hematoxylin working solution)에 10분 염색 후, 슬라이드에 직접 닿지 않도록 흐르는 물에 10분간 담궈 염색에 사용한 헤마톡실린 잔여물을 제거하였다. 0.05% Fast green (FCF) 용액 채널에 슬라이드를 침지하여 10분간 염색을 진행했다. 1% 아세트산 용액 채널에 슬라이드를 10초간 침지하였다. 1% 사프라닌 O 용액을 분주하여, 슬라이드를 10분간 염색시켰다. 탈수를 위해 각기 다른 농도의 에탄올과 NeoClear 용액에 순차적으로 침지하였다. 마운팅을 수행하고 하루 이상 서늘한 곳에서 건조시킨 후, 광학현미경을 통하여 조직을 확인하였다.For Safranin O staining After staining in Weigert's iron hematoxylin working solution for 10 minutes, the slide was immersed in running water for 10 minutes to avoid direct contact with the slide to remove the hematoxylin residue used for staining. The slide was immersed in a channel of 0.05% Fast green (FCF) solution and stained for 10 minutes. The slides were immersed in a 1% acetic acid solution channel for 10 seconds. A 1% safranin O solution was dispensed, and the slides were stained for 10 minutes. For dehydration, they were sequentially immersed in different concentrations of ethanol and NeoClear solution. After mounting and drying in a cool place for at least one day, the tissue was confirmed through an optical microscope.
톨루이딘 블루(Toluidine blue) 염색을 위해 0.1% 톨루이딘 블루 용액을 슬라이드에 분주하고 10분간 염색하였다. 탈수를 위해 각기 다른 농도의 에탄올과 NeoClear 용액에 순차적으로 침지하였다. 마운팅을 수행하고 하루 이상 서늘한 곳에서 건조시킨 후, 광학현미경을 통하여 조직을 확인하였다.For toluidine blue staining, 0.1% toluidine blue solution was dispensed on the slides and stained for 10 minutes. For dehydration, they were sequentially immersed in different concentrations of ethanol and NeoClear solution. After mounting and drying in a cool place for more than one day, the tissue was confirmed through an optical microscope.
류마티스 관절염 동물 모델의 조직학적 평가는 아래 평가 기준에 따라 실시하였다.Histological evaluation of the rheumatoid arthritis animal model was performed according to the following evaluation criteria.
<염증 평가 기준 점수; Inflammation score><Inflammation Assessment Criteria Score; Inflammation score>
0점: 염증 없음 0 points: no inflammation
1점: 내층의 약간의 두꺼워짐 또는 하층의 일부 침윤성 세포가 있음.1 point: Slight thickening of the inner layer or some infiltrating cells in the lower layer.
2점: 내층의 약간의 두꺼워짐과 하층의 일부 침윤성 세포 있음.2 points: Slight thickening of the inner layer and some infiltrating cells in the lower layer.
3점: 안감층(sublininig layer)이 두꺼워짐, 안감층으로 세포의 유입 및 활막강에 세포가 존재함.3 points: thickening of the sublininig layer, the influx of cells into the lining layer and the presence of cells in the synovial cavity.
4점: 많은 염증 세포가 침윤된 활막에 존재함Score 4: Many inflammatory cells are present in the infiltrated synovial membrane
<연골 미란 점수; Cartilage erosion score><Cartilage erosion score; Cartilage erosion score>
0점: 파괴 없음0 points: no destruction
1점: 제한되는 최소 침식이 있음.1 point: Limited minimal erosion.
2점: 제한된 지역에서 약간에서 중간 정도의 침식이 있음. 2 points: Slight to moderate erosion in a limited area.
3점: 더 확장된 침식이 있음.Score 3: There is more extensive erosion.
4점: 대부분의 연골 파괴가 보임.4 points: Most cartilage destruction is seen.
도 5c 및 5d는 CIA, iPSC-iMSC-Exo 및 MSC-Exo의 조직학적 평가 결과를 나타낸다. 관절 조직에서 염증 지수(inflammation score)와 연골 미란 점수 (Cartilage damage score)를 통해 iPSC-iMSC-Exo 투여 그룹이 MSC-Exo 투여 그룹이나 CIA 그룹과 비교하여 관절조직에서 평균 관절지수가 유의하게 낮다는 것을 확인하였다. 5c and 5d show the histological evaluation results of CIA, iPSC-iMSC-Exo and MSC-Exo. In the joint tissue, the average joint index in the joint tissue was significantly lower in the iPSC-iMSC-Exo group compared to the MSC-Exo group or the CIA group through the inflammation score and the cartilage damage score. confirmed that.
도 6c 및 6d는 CIA, iPSC-iMSC-Exo 50㎍ 및 100㎍ 투여 그룹의 조직학적 평가 결과를 나타낸다. iPSC-iMSC-Exo 50㎍와 iPSC-iMSC-Exo 100 ㎍ 투여 그룹을 비교한 경우에도, iPSC-iMSC-Exo 농도 의존적으로 염증 평균 지수와 연골 미란 점수가 낮음을 확인하였다. 즉, iPSC-iMSC-Exo는 동물 모델에서 류마티스 관절염을 치료하는 데 우수한 효과가 있음을 알 수 있었다.6c and 6d show the histological evaluation results of CIA, iPSC-iMSC-Exo 50 μg and 100 μg administration groups. Even when iPSC-iMSC-Exo 50 μg and iPSC-iMSC-Exo 100 μg administration group were compared, it was confirmed that the iPSC-iMSC-Exo concentration-dependent mean inflammation index and cartilage erosion score were low. That is, it was found that iPSC-iMSC-Exo had an excellent effect in treating rheumatoid arthritis in an animal model.

Claims (10)

  1. 생물학적 시료로부터 세포를 분리하는 단계;isolating cells from the biological sample;
    분리된 세포로부터 역분화 줄기세포를 제조하는 단계;preparing dedifferentiated stem cells from the isolated cells;
    제조된 역분화 줄기세포를 b-FGF의 존재 하에 배양하여 중간엽 줄기세포를 유도하는 단계; 및Inducing mesenchymal stem cells by culturing the prepared dedifferentiated stem cells in the presence of b-FGF; and
    유도된 중간엽 줄기세포로부터 엑소좀을 분리하는 단계를 포함하는 엑소좀의 제조방법.A method for producing an exosome comprising the step of isolating the exosome from the induced mesenchymal stem cells.
  2. 청구항 1에 있어서, 상기 분리된 세포는 단핵 세포인 것인 엑소좀의 제조방법.The method according to claim 1, wherein the isolated cells are mononuclear cells.
  3. 청구항 1에 있어서, 상기 유도하는 단계는 하기를 포함하는 것인 엑소좀의 제조방법:The method according to claim 1, wherein the inducing step comprises the following:
    (a) 제조된 역분화 줄기세포를 콜라게나아제 용액과 반응시키는 단계;(a) reacting the prepared retrodifferentiated stem cells with a collagenase solution;
    (b) 상기 반응시킨 역분화 줄기세포를 원심분리하는 단계;(b) centrifuging the reacted dedifferentiated stem cells;
    (c) 상기 원심분리한 역분화 줄기세포를 배지에서 현탁하는 단계; 및(c) suspending the centrifuged retrodifferentiated stem cells in a medium; and
    (d) 상기 현탁된 역분화 줄기세포를 b-FGF로 자극하는 단계.(d) stimulating the suspended retrodifferentiated stem cells with b-FGF.
  4. 청구항 3에 있어서, 상기 단계 (a)의 콜라게나아제는 콜라게나아제 타입 4 (type IV Collagenase)인 것인 엑소좀의 제조방법.The method according to claim 3, wherein the collagenase in step (a) is collagenase type 4 (type  IV  Collagenase).
  5. 청구항 3에 있어서, 상기 단계 (c)의 배지는 αMEM(α-Minimum Eagle’s Medium)인 것인 엑소좀의 제조방법.The method of claim 3, wherein the medium in step (c) is αMEM (α-Minimum Eagle's Medium).
  6. 청구항 3에 있어서, 단계 (d) 후 재조합 인간 (recombinant human) TGF-β로 자극하는 단계를 추가로 포함하는 것인 엑소좀의 제조방법.The method according to claim 3, further comprising the step of stimulation with recombinant human TGF-β after step (d).
  7. 청구항 1에 있어서, 상기 엑소좀을 분리하는 단계는 하기 단계를 포함하는 것인 엑소좀의 제조방법:The method according to claim 1, wherein the step of separating the exosomes comprises the following steps:
    (a) 유도된 중간엽 줄기세포를 배지에서 36 내지 60 시간 동안 배양하는 단계;(a) culturing the induced mesenchymal stem cells in a medium for 36 to 60 hours;
    (b) 상기 배양된 중간엽 줄기세포 배양액을 0 내지 5℃에서 200 내지 400 g로 5 내지 15 분 동안 원심분리하는 단계;(b) centrifuging the cultured mesenchymal stem cell culture medium at 200 to 400 g at 0 to 5° C. for 5 to 15 minutes;
    (c) 상기 (b) 단계에서 원심분리된 배양액의 상층액을 0 내지 5℃에서 1500 내지 2500 g로 15 내지 30 분 동안 원심분리하는 단계;(c) centrifuging the supernatant of the culture medium centrifuged in step (b) at 1500 to 2500 g at 0 to 5° C. for 15 to 30 minutes;
    (d) 상기 (c) 단계에서 원심분리된 배양액의 상층액을 0 내지 5℃에서 8,000 내지 12,000 g로 20 내지 40 분 동안 원심분리하는 단계;(d) centrifuging the supernatant of the culture solution centrifuged in step (c) at 8,000 to 12,000 g at 0 to 5° C. for 20 to 40 minutes;
    (e) 상기 (d) 단계에서 원심분리된 배양액의 상층액을 여과지로 필터링하는 단계; 및(e) filtering the supernatant of the culture medium centrifuged in step (d) with a filter paper; and
    (f) 상기 (e) 단계에서 필터링된 배양액을 0 내지 5℃에서 80,000 내지 120,000 g에서 100 내지 140 분 동안 원심분리하는 단계.(f) centrifuging the culture solution filtered in step (e) at 80,000 to 120,000 g at 0 to 5° C. for 100 to 140 minutes.
  8. 청구항 7에 있어서, 상기 (a) 단계의 배지는 SF-DMEM (serum free-Dulbecco Modified Eagle Medium)인 것인 엑소좀의 제조방법.The method of claim 7, wherein the medium in step (a) is SF-DMEM (serum free-Dulbecco Modified Eagle Medium).
  9. 청구항 1에 의해 제조된 엑소좀을 포함하는 자가면역 질환의 예방 또는 치료용 약학적 조성물.A pharmaceutical composition for preventing or treating an autoimmune disease comprising the exosome prepared according to claim 1.
  10. 청구항 9에 있어서, 상기 자가면역 질환은 류마티스 관절염인 것인 자가면역 질환의 예방 또는 치료용 약학적 조성물.The pharmaceutical composition for preventing or treating an autoimmune disease according to claim 9, wherein the autoimmune disease is rheumatoid arthritis.
PCT/KR2021/015286 2020-11-18 2021-10-28 Method for producing exosomes isolated from induced pluripotent stem cell-derived mesenchymal stem cells, and use thereof WO2022108165A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2020-0154551 2020-11-18
KR20200154551 2020-11-18

Publications (1)

Publication Number Publication Date
WO2022108165A1 true WO2022108165A1 (en) 2022-05-27

Family

ID=81709224

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2021/015286 WO2022108165A1 (en) 2020-11-18 2021-10-28 Method for producing exosomes isolated from induced pluripotent stem cell-derived mesenchymal stem cells, and use thereof

Country Status (2)

Country Link
KR (1) KR20220068148A (en)
WO (1) WO2022108165A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117004562A (en) * 2023-04-03 2023-11-07 广东医科大学附属医院 Autophagy activated mesenchymal stem cell exosome, and preparation method and application thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20190083932A (en) * 2018-01-05 2019-07-15 재단법인 아산사회복지재단 Composition for Improving Skin Conditions, or Preventing or Treating Skin Diseases comprising Induced Pluripotent Stem Cell-derived Mesenchymal Stem Cell and Exosomes derived therefrom
WO2019238693A1 (en) * 2018-06-11 2019-12-19 Health And Biotech France (H & B France) Extracellular vesicles derived from mesenchymal stem cells
KR20200088408A (en) * 2017-11-16 2020-07-22 보드 오브 리전츠, 더 유니버시티 오브 텍사스 시스템 Method for producing exosome derived from MSC
US20200297761A1 (en) * 2019-03-18 2020-09-24 Buddhist Tzu Chi Medical Foundation Mesenchymal stem cell derived exosomes and method for preventing or treating a joint disorder by administering a composition comprising the same

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20200088408A (en) * 2017-11-16 2020-07-22 보드 오브 리전츠, 더 유니버시티 오브 텍사스 시스템 Method for producing exosome derived from MSC
KR20190083932A (en) * 2018-01-05 2019-07-15 재단법인 아산사회복지재단 Composition for Improving Skin Conditions, or Preventing or Treating Skin Diseases comprising Induced Pluripotent Stem Cell-derived Mesenchymal Stem Cell and Exosomes derived therefrom
WO2019238693A1 (en) * 2018-06-11 2019-12-19 Health And Biotech France (H & B France) Extracellular vesicles derived from mesenchymal stem cells
US20200297761A1 (en) * 2019-03-18 2020-09-24 Buddhist Tzu Chi Medical Foundation Mesenchymal stem cell derived exosomes and method for preventing or treating a joint disorder by administering a composition comprising the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
AHMED ABDAL DAYEM, LEE SOO BIN, KIM KYEONGSEOK, LIM KYUNG MIN, JEON TAK-IL, SEOK JAEKWON, CHO SSANG-GOO: "Production of Mesenchymal Stem Cells Through Stem Cell Reprogramming", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 20, no. 8, 18 April 2019 (2019-04-18), pages 1922, XP055763938, DOI: 10.3390/ijms20081922 *

Also Published As

Publication number Publication date
KR20220068148A (en) 2022-05-25

Similar Documents

Publication Publication Date Title
WO2019135644A1 (en) Composition for improving, preventing or treating skin diseases comprising induced pluripotent stem cell-derived mesenchymal stem cell and exosome derived therefrom
WO2018074758A2 (en) Method for sorting highly effective stem cells for treating immune disorder
WO2019135645A1 (en) Composition for improving, preventing or treating skin disease comprising induced pluripotency stem cell-derived mesenchymal stem cells pretreated with interferon gamma and exosomes derived therefrom
WO2019198995A1 (en) Exosome-based conversion method for immune cells
WO2019117633A1 (en) Cosmetic composition and pharmaceutical composition for alleviating atopic dermatitis, hair loss, and wounds or reducing skin wrinkles
WO2017164467A1 (en) Mesenchymal stem cell culture for prevention or treatment of immune disease or inflammatory disease and preparation method thereof
WO2022169049A1 (en) Method for obtaining mesenchymal stem cell-derived exosome
Seguier et al. Inhibition of the differentiation of monocyte-derived dendritic cells by human gingival fibroblasts
WO2022108165A1 (en) Method for producing exosomes isolated from induced pluripotent stem cell-derived mesenchymal stem cells, and use thereof
WO2021241798A1 (en) Method for preparation of mesenchymal stem cell-derived exosome and cell culture prepared therefrom
WO2012008733A2 (en) Stem cells derived from primary placenta tissue and cellular therapeutic agent containing same
WO2017146538A1 (en) Pharmaceutical composition for preventing or treating regulatory t cell-mediated diseases
EP4004191A1 (en) Precursor cells of induced pluripotent stem cell-derived mesenchymal stem cells and method for preparing the same
WO2019035668A9 (en) Composition for treatment of thyroid associated ophthalmopathy, comprising mesenchymal stem cell
WO2019031729A2 (en) Use of composition comprising stem cell-derived exosome as effective ingredient in alleviating non-alcoholic simple steatosis or non-alcoholic steatohepatitis
WO2019004738A2 (en) Use of composition comprising adipose stem cell-derived exosome as effective ingredient in alleviating dermatitis
WO2019103436A9 (en) Composition for culturing nk cells and method for culturing nk cells using same
WO2013165120A1 (en) Method for culturing neural crest stem cells, and use thereof
WO2021020666A1 (en) Precursor cells of induced pluripotent stem cell-derived mesenchymal stem cells and method for preparing the same
WO2015023147A1 (en) Mtor/stat3 signal inhibitor-treated mesenchymal stem cell having immunomodulatory activity, and cell therapy composition comprising same, for preventing or treating immune disorders
WO2019216450A1 (en) Method for culturing stem cells secreting anti-inflammatory components, and anti-inflammatory composition containing said stem cell culture medium
WO2015023165A1 (en) Inflammation-controlling composite and stabilized mesenchymal stem cells having optimized immunity control function by blocking stat3 signal molecule
WO2020122405A1 (en) Composition for preventing or treating inflammatory diseases, comprising mesenchymal stem cells derived from dedifferentiated stem cells
WO2015194710A1 (en) Composition comprising mesenchymal stem cells treated with stat3 inhibitor as active ingredient for preventing or treating immune diseases
WO2019212123A1 (en) Method for increasing dendritic cell migration ability, and use thereof

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: 21894936

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21894936

Country of ref document: EP

Kind code of ref document: A1

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

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 24.10.2023)