WO2019050240A1 - New use of exosome kit comprising stem cell-derived exosome - Google Patents

New use of exosome kit comprising stem cell-derived exosome Download PDF

Info

Publication number
WO2019050240A1
WO2019050240A1 PCT/KR2018/010254 KR2018010254W WO2019050240A1 WO 2019050240 A1 WO2019050240 A1 WO 2019050240A1 KR 2018010254 W KR2018010254 W KR 2018010254W WO 2019050240 A1 WO2019050240 A1 WO 2019050240A1
Authority
WO
WIPO (PCT)
Prior art keywords
mask
exosome
patch
battery
skin
Prior art date
Application number
PCT/KR2018/010254
Other languages
French (fr)
Korean (ko)
Inventor
이용원
조병성
Original Assignee
주식회사 엑소코바이오
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR1020180101494A external-priority patent/KR102197562B1/en
Application filed by 주식회사 엑소코바이오 filed Critical 주식회사 엑소코바이오
Publication of WO2019050240A1 publication Critical patent/WO2019050240A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L33/00Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof
    • A23L33/10Modifying nutritive qualities of foods; Dietetic products; Preparation or treatment thereof using additives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • 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
    • 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
    • A61K35/48Reproductive organs
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/96Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution
    • A61K8/98Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution of animal origin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin

Definitions

  • the present invention relates to a new use of an exosome kit comprising exosomes derived from stem cells.
  • the skin of the human body is an organ that physically and chemically protects the body from the outside and performs the biochemical functions necessary for the metabolism of the whole body.
  • inflammatory skin diseases that appear on human skin are called dermatitis.
  • Relatively common dermatitis includes atopic dermatitis, contact dermatitis, and seborrhoic dermatitis.
  • Contact dermatitis is a dermatitis caused by contact with foreign substances. Contact dermatitis is characterized by irritant contact dermatitis and allergic contact dermatitis, depending on the mechanism by which one substance can cause both of these reactions simultaneously. Most of the substances that cause contact dermatitis are organic compounds. When the dermatitis-inducing substance comes into contact with the skin sensitized to these substances, it is known that the memory cell senses it and secretes various chemicals to cause inflammation.
  • Atopic dermatitis is a chronic inflammatory skin disease characterized by itchy and eczematous lesions. According to the results of previous studies, it is known that various factors are involved in the onset of atopic dermatitis. When atopic dermatitis is induced, inflammatory cells such as eosinophil, neutrophil and mast cell are observed in the lesion and immunoglobulin IgE produced from mast cells increases. In addition, IL-4, IL-5, and IL-13, which are inflammatory cytokines, are amplified and amplified by this process.
  • TSLP Thimic stromal lymphopoietin
  • seborrheic dermatitis is a chronic inflammatory skin disease that occurs due to increased activity of sebaceous gland, which occurs in the scalp and face, especially around eyebrows, nose, lips, ears, underarms, chest, groin and the like.
  • Drug remedies developed to date include steroids, antihistamines, and immunosuppressants such as cyclosporin A.
  • these drugs have serious problems such as skin atrophy, vasodilation, pigment desalination, hypersensitivity of injection site, tolerance and neutropenia.
  • exosome which has intercellular signal transduction
  • Extracellular vesicles Cells release various membrane-type vesicles in the extracellular environment, and these release vesicles are commonly referred to as extracellular vesicles (EVs).
  • the extracellular endoplasmic reticulum is sometimes referred to as cell membrane-derived endoplasmic reticulum, ectosomes, shedding vesicles, microparticles, exosomes and, in some cases, differentiated from exosomes.
  • Exosome is an endoplasmic reticulum of several tens to several hundreds of nanometers in size, composed of double lipid membranes identical to the cell membrane structure, and contains proteins, nucleic acids (mRNA, miRNA, etc.) called exosomal cargo inside.
  • Exosomal cargo contains a wide range of signaling factors, which are known to be specific for cell types and differentially regulated by the environment of the secretory cells.
  • Exosome is an intercellular signaling mediator that is secreted by the cell, and various cell signals transmitted through it regulate cell behavior including activation, growth, migration, differentiation, de-differentiation, apoptosis, and necrosis of target cells It is known.
  • Exosomes contain specific genetic material and bioactivity factors depending on the nature and condition of the derived cells.
  • the proliferating stem cell-derived exosomes regulate cell behavior such as cell migration, proliferation and differentiation, and reflect the characteristics of stem cells involved in tissue regeneration (Nature Review Immunology 2002 (2) 569-579).
  • the present inventors have conducted intensive studies on the development of new formulations capable of stably maintaining and storing exosomes and the combination of medical or cosmetic techniques, and have found that exosomes which can be applied to prevent, inhibit, alleviate, The kit was developed to complete the present invention.
  • the object of the present invention is to provide an exosome kit comprising exosome derived from stem cells and an application thereof.
  • the object of the present invention is to provide a mask pack, a mask sheet or a patch which is coated or immersed with a composition containing an exosome derived from a stem cell as an active ingredient for the prevention, inhibition, alleviation, improvement or treatment of dermatitis, And an iontophoresis device for use in the treatment of cancer.
  • Another object of the present invention is to provide a cosmetic method for controlling the condition of mammalian skin through prevention, suppression, alleviation or improvement of dermatitis except for therapeutic use using the exosomic kit.
  • the present invention provides a method for preventing, suppressing, alleviating, ameliorating, or treating dermatitis, which comprises applying a mask pack containing a composition containing an exosome derived from stem cells as an active ingredient, Or patch, and an iontophoresis device.
  • exosomes refers to an endoplasmic reticulum of several tens to several hundred nanometers (preferably about 30 to 200 nm) in size, consisting of double lipid membranes identical in structure to the cell membrane The particle size of the exosome can be varied according to the cell type, the separation method and the measurement method) (Vasiliy S. Chernyshev et al., "Size and shape characterization of hydrated and desiccated exosomes", Anal Bioanal Chem, DOI 10.1007 / s00216-015-8535-3). Exosomes contain proteins called exosomal cargo (cargo), nucleic acids (mRNA, miRNA, etc.).
  • Exosomal cargo contains a wide range of signaling factors, which are known to be specific for cell types and differentially regulated by the environment of the secretory cells.
  • Exosome is an intercellular signaling mediator that is secreted by the cell, and various cell signals transmitted through it regulate cell behavior including activation, growth, migration, differentiation, de-differentiation, apoptosis, and necrosis of target cells It is known.
  • exosome refers to a vesicle that has a nano-sized vesicle structure secreted from various stem cells and released into the extracellular space and has a composition similar to exosome (e.g., - pseudo-bezacl).
  • the type of the stem cell is not limited, but may be, for example, a mesenchymal stem cell, for example, a fat, a bone marrow, an umbilical cord or a cord blood derived stem cell, Derived stem cells.
  • the type of the adipose-derived stem cell is not limited as long as it does not cause a risk of infection by a pathogen and does not cause an immune rejection reaction, but it may be preferably a human adipose-derived stem cell.
  • the stem cell-derived exosome used in the present invention is effective for preventing, suppressing, alleviating, ameliorating or treating dermatitis and not causing adverse effects on the human body, so long as the stem cell-derived exosome can be used in a variety of stem cell- Of course, exosomes can be used. Therefore, it should be understood that the exosome isolated according to the separation method of the following embodiments should be understood as an example of exosome that can be used in the present invention, and the present invention is not limited thereto.
  • iontophoresis refers to a method in which an ionized active ingredient is allowed to permeate through the skin by an electrical repulsive force by applying a minute current to the skin to which the effective substance is applied, .
  • the iontophoresis used in one embodiment of the present invention is a method in which a current flows from an external power source to an electrode patch on the skin to introduce minute current into the skin, A method in which minute current is introduced, a method in which minute current is introduced into skin through a patch equipped with a reversed electrodialysis means for generating current through a difference in ion concentration between a high concentration electrolyte solution and a low concentration electrolyte solution, and the like .
  • the present invention is not limited thereto, and it is needless to say that various types of iontophoresis can be used.
  • the exosome kit of one embodiment of the present invention comprises a first mask pack, a mask sheet or a patch which is applied or immersed with a composition containing an exosome derived from a stem cell as an active ingredient, And an iontophoresis device for flowing an electric current.
  • an exosome kit is characterized in that the first mask pack, the mask sheet or the patch is brought into contact with or attached to the skin of the mammal, and the ion mask By positioning the phoresis device, the first mask pack, mask sheet or patch can be contacted or attached to be used in such a way that micro-currents flow to the skin of the mammal to which the composition is applied.
  • the composition may be used in combination with a conventionally used dermatitis remedy and / or moisturizer to the extent that its action (alleviation or improvement of dermatitis symptoms) is not impaired.
  • the composition may be supported on or mixed with at least one of hydrogel, hyaluronic acid, hyaluronic acid salt (for example, sodium hyaluronate), or hyaluronic acid gel.
  • the type of the hydrogel is not limited, but it may preferably be a hydrogel obtained by dispersing a gelated polymer in a polyhydric alcohol.
  • the gel polymer is at least one selected from the group consisting of pluronic, purified agar, agarose, gellan gum, alginic acid, carrageenan, cacao gum, xanthan gum, galactomannan, glucomannan, pectin, cellulose, guar gum and locust bean gum
  • the polyhydric alcohol may be at least one selected from the group consisting of ethylene glycol, propylene glycol, 1,3-butylene glycol, isobutylene glycol, dipropylene glycol, sorbitol, xylitol and glycerin.
  • the iontophoresis device may be applied to a flexible battery, a lithium ion secondary battery, an alkaline battery, a dry battery, a mercury battery, a lithium battery, a nickel-cadmium battery, Or a second mask pack, a mask sheet, or a patch having the at least one battery mounted thereon.
  • the second mask pack, the mask sheet or the patch may be laminated on the first mask pack, the mask sheet or the patch.
  • At least one surface of the first mask pack, the mask sheet or the patch, at least one surface of the first mask pack, the mask sheet or the patch may be coated with a hydrogel, hyaluronic acid, hyaluronic acid salt (for example, sodium hyaluronate) At least one of the gels may be applied.
  • the type of the hydrogel is not limited, but it may preferably be a hydrogel obtained by dispersing a gelated polymer in a polyhydric alcohol. The gelled polymer and the polyhydric alcohol may be exemplified in the above description.
  • the micro current flows from the iontophoresis device so that the exosomes can effectively penetrate the epidermis and penetrate deeply into the skin.
  • the exosome kit of one embodiment of the present invention can be effectively used for preventing, suppressing, ameliorating, alleviating or treating various dermatitis accompanied by itch, and is preferably used for the treatment of contact dermatitis, irritant contact dermatitis, allergic contact dermatitis, Allergic contact dermatitis, contact urticaria, atopic dermatitis, seborrheic dermatitis, autoimmune dermatitis, autoimmune progesterone dermatitis, dermatitis, acne or eczema.
  • Another embodiment of the present invention provides a cosmetic method for controlling the condition of a mammalian skin by preventing, suppressing, alleviating or improving dermatitis except for therapeutic use using the exosomic kit.
  • conditioning of the skin means improvement of the condition of the skin and / or prevention of the condition of the skin
  • improvement of the condition of the skin means visual and / Or a tactile perceptible positive change.
  • improvement of the condition of the skin may be mitigation or improvement of dermatitis related symptoms.
  • the cosmetic method of one embodiment of the present invention can be carried out using the exosome kit as described above.
  • the cosmetic method of one embodiment of the present invention comprises the steps of (a) contacting a first mask pack, a mask sheet or a patch with a composition containing an exosome derived from a stem cell as an active ingredient, (B) positioning the iontophoresis device on the first mask pack, mask sheet or patch, and (c) contacting the first mask pack, mask sheet or patch with the iontophoresis device, Performing iontophoresis to flow a microcurrent into the skin of the mammal to which the composition is applied; and (d) delivering the exosomes through the microcurrent to the inside of the mammalian skin.
  • At least one surface of the first mask pack, the mask sheet or the patch, at least one surface of the first mask pack, the mask sheet or the patch may be coated with a hydrogel, hyaluronic acid, hyaluronate (e.g., sodium hyaluronate) At least one of the gels may be applied.
  • the type of the hydrogel is not limited, but it may preferably be a hydrogel obtained by dispersing a gelated polymer in a polyhydric alcohol. The gelled polymer and the polyhydric alcohol may be exemplified in the above description.
  • the iontophoresis device comprises a flexible battery, a lithium ion secondary battery, an alkaline battery, a dry battery, a mercury battery, a lithium battery, a nickel-cadmium battery, Or a second mask pack, a mask sheet or a patch having the at least one battery mounted thereon.
  • the step (b) may be performed by laminating the second mask pack, the mask sheet or the patch on the first mask pack, the mask sheet or the patch.
  • the exosome kits of the present invention exhibit excellent effects on dermatitis symptoms and skin cosmetics such as improving or restoring the dermatological conditions deteriorated due to the dermatitis symptoms, and it is possible to alleviate or improve the condition of the skin condition due to dermatitis, .
  • the exosome kit of the present invention can prevent the exosomes applied or deposited on the mask pack, the mask sheet or the patch by the iontophoresis action to effectively penetrate the epidermis and penetrate deeply into the skin, Thereby exhibiting an effect of improving or alleviating the skin condition.
  • FIG. 1 shows the results of physical property analysis of exosomes obtained according to one embodiment of the present invention.
  • Figure 1A shows the particle size distribution and number of particles by TRPS (tunable resistive pulse sensing) analysis.
  • Figure 1B shows particle size distribution and particle number by NTA (nanoparticle tracking analysis) analysis.
  • Fig. 1C shows particle images by transmission electron microscopy (TEM) according to magnification.
  • TEM transmission electron microscopy
  • Figure 1D shows the Western blot results of exosomes obtained according to one embodiment of the present invention.
  • RTI ID 0.0 > 1E < / RTI > shows flow cytometric analysis results for CD63 and CD81 in marker assays for exosomes obtained according to one embodiment of the present invention.
  • FIG. 2 shows the result of confirming the absence of cytotoxicity after treatment of exosomes obtained according to one embodiment of the present invention in human skin fibroblast HS68 cells.
  • FIG. 3 shows that the amount of mRNA expression of TNF- ⁇ , IL-6, IL-1 ⁇ and iNOS induced by LPS was decreased when the exosome used in the exosome kit of the present invention was treated with LPS for RAW 264.7 cells And a real-time PCR result.
  • FIG. 4 shows experimental results showing that NO formation, which is a type of inflammatory reaction, is reduced by the exosome used in the exosome kit of the present invention.
  • PBS is phosphate-buffered saline
  • DEX is dexamethasone
  • EXO is exosome
  • CM is conditioned medium
  • CM-EXO is adipocyte- (Exosome-depeleted conditioned media).
  • FIG. 5 shows experimental results confirming that TNF-.alpha. Formation, which is an inflammatory cytokine, is reduced by exosomes used in the exosome kit of the present invention.
  • PBS represents phosphate-buffered saline
  • DEX represents dexamethasone
  • each numeral represents exosome throughput ( ⁇ g / mL).
  • FIG. 6 shows the results of co-treatment of exosomes used in the exosomal kit of the present invention together with LPS and IFN-y for THP-1-derived macrophages obtained after differentiating THP-1 monocytes into macrophages Time PCR results confirming that the mRNA expression level of the pro-inflammatory cytokine IL-6 was decreased.
  • FIGS. 7 to 9 show the results of pre-treatment of the exosomes used in the exosomal kit of the present invention and culturing for a predetermined period of time on THP-1-derived macrophages obtained after differentiating THP-1 monocytes into macrophages
  • FIGS. 7 to 9 show the results of pre-treatment of the exosomes used in the exosomal kit of the present invention and culturing for a predetermined period of time on THP-1-derived macrophages obtained after differentiating THP-1 monocytes into macrophages
  • FIGS. 7 to 9 show the results of pre-treatment of the exosomes used in the exosomal kit of the present invention and culturing for a predetermined period of time on THP-1-derived macrophages obtained after differentiating THP-1 monocytes into macrophages
  • FIGS. 7 to 9 show the results of pre-treatment of the exosomes used in the exosomal kit of the present invention and culturing for
  • FIG. 10 shows fluorescence intensity measurement results of exosomes stained with PKH67.
  • Fig. 11 is a fluorescence microscope image showing the degree of transfer of fluorescent stained exosomes into pig skin tissue.
  • Fig. 12 is a confocal fluorescence microscope image showing the extent of fluorescence-stained exosomes transferred into the mouse skin tissue.
  • FIG. 13 is a graph showing a comparison of the total fluorescence intensity obtained by measuring the fluorescence intensity on each image in Fig.
  • FIG. 14 is a photograph showing the skin condition before and after the treatment of human skin with the exosomal kit of the present invention.
  • FIG. 15 is a graph showing the results of application of iontophoresis for applying a microcurrent to a skin (affected part) coated with the composition after applying the composition containing exosome used in the exosome kit of the present invention to human skin As a result, it is a photograph showing that the erythema of the skin (the affected part) which has a severe dermatitis is remarkably improved.
  • 16 is a perspective view showing a configuration of the exosome kit 100 according to one embodiment of the present invention.
  • Mouse macrophage line RAW 264.7 was purchased from Korean Cell Line Bank and cultured.
  • DMEM purchased from ThermoFisher Scientific
  • medium containing 10% fetal bovine serum purchased from ThermoFisher Scientific
  • 1% antibiotic-antimycotics purchased from ThermoFisher Scientific 2 , and 37 ° C.
  • HS68 cells a human dermal fibroblast, were purchased from ATCC and cultured in RPMI 1640 supplemented with 10% fetal bovine serum (purchased from ThermoFisher Scientific) and 1% antibiotic-antimycotics (purchased from ThermoFisher Scientific) The cells were subcultured in DMEM (purchased from ThermoFisher Scientific) medium containing 5% CO 2 at 37 ° C.
  • adipose-derived stem cells were cultured at 5% CO 2 and 37 ° C. Then, the cells were washed with a phosphate-buffered saline (purchased from ThermoFisher Scientific), replaced with serum-free, non-phenol red medium, cultured for 1 to 10 days, and the supernatant .
  • a phosphate-buffered saline purchased from ThermoFisher Scientific
  • Trehalose was added to the culture medium in an amount of 2% by weight in order to obtain an exosome having a uniform particle size distribution and high purity in the process of separating exosome.
  • the culture was filtered with a 0.22 ⁇ m filter to remove impurities such as cellular debris, waste products and large particles.
  • the filtered cultures were immediately separated to isolate exosomes.
  • the filtered culture was stored in a refrigerator (image below 10 ° C) and used for exosome isolation.
  • the filtered culture was frozen in an ultra-low temperature freezer at -60 ° C or lower, and thawed, followed by exosome isolation. Then, the exosomes were separated from the culture medium by using Tangential Flow Filtration (TFF).
  • TMF Tangential Flow Filtration
  • Example 1 a TFF (Tangential Flow Filtration) method was used for separating, concentrating, desalting and diafiltration exosomes from a culture filtrated with a 0.22 ⁇ m filter.
  • a cartridge filter also called a hollow fiber filter (purchased from GE Healthcare) or a cassette filter (purchased from Pall or Sartorius or Merck Millipore) was used.
  • the TFF filter can be selected by a variety of molecular weight cutoffs (MWCO). Exosomes were selectively isolated and concentrated by selected MWCOs, and particles, proteins, lipids, nucleic acids, low molecular weight compounds, etc. smaller than MWCO were removed.
  • MWCO molecular weight cutoffs
  • TFF filters of MWCO 100,000 Da (Dalton), 300,000 Da, or 500,000 Da were used.
  • the culture medium was concentrated to a volume of about 1/100 to 1/25 using the TFF method, and substances smaller than MWCO were removed to separate the exosomes.
  • Separated and concentrated exosomal solutions were further desalted and buffered (diafiltration) using the TFF method.
  • the desalination and buffer exchange are performed by continuous diafiltration or discontinuous diafiltration, and at least 4 times, preferably 6 times to 10 times or more, more preferably, Was performed using a buffer solution having a volume of 12 times or more.
  • 2% by weight of trehalose dissolved in PBS was added to obtain an exosome having a uniform particle size distribution and high purity.
  • NTA nanoparticle tracking analysis
  • TRPS tunable resistive pulse sensing
  • FIG. 1D shows the presence of CD9, CD63, CD81 and TSG101 markers as a result of performing Western blotting on isolated exosomes according to the method of one embodiment of the present invention.
  • Anti-CD9 purchased from Abcam
  • anti-CD63 purchasedd from System Biosciences
  • anti-CD81 purchasedd from System Biosciences
  • anti-TSG101 purchasedd from Abcam
  • FIG. 1E shows the presence of CD63 and CD81 markers as a result of analysis using flow cytometry on exosomes isolated according to the method of one embodiment of the present invention.
  • Human CD63 isolation / detection kit purchased from ThermoFisher Scientific
  • PE-mouse anti-human CD63 PE-Mouse anti markers were stained using the PE-mouse CD63 (purchased from BD) and PE-mouse anti-human CD81 (purchased from BD), and analyzed using a flow cytometer (ACEA Biosciences) Respectively.
  • exosomes derived from stem cells used in the present invention are not limited to the exosomes of the above-mentioned embodiments, and it is possible to use various stem cell-derived exosomes which are used in the art or can be used in the future Of course. It is to be understood that the exosomes isolated according to the above embodiments are to be understood as an example of exosomes derived from stem cells which can be used in the present invention, and the present invention is not limited thereto.
  • exosomes were treated by concentration to the cells and the proliferation rate of the cells was confirmed.
  • HS68 cells were suspended in DMEM containing 10% FBS, and the cells were mixed with 80 ⁇ 90% confluency and cultured in a 5% CO 2 incubator at 37 ° C for 24 hours. After 24 hours, the culture solution was removed, and the cell survival rate was evaluated by culturing the exosome prepared in Example 2 for each concentration and culturing for 24 to 72 hours.
  • WST-1 reagent purchased from Takara
  • MTT reagent purchased from Sigma
  • CellTiter-Glo reagent purchased from Promega
  • Aramar Blue reagent alamarBlue reagent purchased from ThermoFisher Scientific
  • a microplate reader purchased from Molecular Devices
  • the comparison group was based on the number of cells cultured in the normal cell culture medium not treated with exosome, and it was confirmed that the cytosolic effect of exosome derived from stem cells did not appear within the tested concentration range (FIG. 2).
  • RAW 264.7 cells were suspended in DMEM medium containing 10% FBS and dispensed into each well of a multiwell plate to have a confluency of 80-90%.
  • the stem cell-derived exosomes (used as the exosome prepared in Example 2 as used in the exosome kit of the present invention) diluted in a new serum-free medium containing LPS were treated for 1 to 24 hours at an appropriate concentration and cultured .
  • the cultured supernatant was taken and NO and inflammatory cytokines present in the culture were measured to confirm the inflammatory response.
  • the inflammatory response in the culture medium was measured using a NO detection kit (purchased from Intron Bio or Promega).
  • ELISA kit purchased from the R & D system
  • Dexamethasone purchased from Sigma
  • cDNA was prepared from the total RNA obtained from the RAW 264.7 cells treated as described above, and the amount of mRNA change of iNOS, TNF- ⁇ , IL-6 and IL-1 ⁇ was measured using a real-time PCR method.
  • the GAPDH gene was used as a standard gene for quantifying the above genes.
  • the types and sequences of the primers used in the real-time PCR are shown in Table 1 below.
  • RAW 264.7 cells which are macrophages of mice
  • NO production which is an inflammatory reaction induced by LPS
  • FIG. 5 RAW 264.7 cells, macrophages of mice, were treated with exosomes used in the exosomal kit of the present invention in the presence of LPS.
  • LPS-induced inflammatory cytokine TNF-a production As shown in FIG. 4, RAW 264.7 cells, which are macrophages of mice, were treated with exosomes used in the exosomal kit of the present invention in the presence of LPS.
  • exosome used in the exosomal kit of the present invention has an activity of reducing the inflammatory reaction induced by LPS, which is useful for preventing, inhibiting, improving, alleviating or treating dermatitis, It strongly suggests that exosome kits may be useful for the prevention, inhibition, amelioration, palliation or treatment of dermatitis.
  • THP-1 monocyte THP-1 human monocyte
  • the anti-inflammatory effect of exosomes used in the exosome kit of the present invention was confirmed as follows.
  • THP-1 monocytes were cultured in RPMI supplemented with 10% FBS (Fetal Bovine Serum), 1% penicillin-streptomycin and 100 nM Phorbol 12-myristate 13-acetate (purchased from Sigma) 1640 (purchased from ThermoFisher Scientific) medium at 1640 (purchased from ThermoFisher Scientific), and then seeded in a 12-well plate at a density of 6 ⁇ 10 5 cells / mL and cultured in a 5% CO 2 incubator at 37 ° C. for 48 hours to obtain THP-1 Derived macrophage (THP-1 derived macrophage). The differentiated cells were then washed once with PBS and cultured in RPMI 1640 medium without formalin 12-myristate 13-acetate for 24 hours to stabilize the cells.
  • FBS Fetal Bovine Serum
  • penicillin-streptomycin 100 nM Phorbol 12-myristate 13-acetate
  • 1640 purchased from ThermoFisher
  • RPMI 1640 medium without FBS Fetal Bovine Serum
  • 100 ng / mL LPS purchased from Sigma
  • 20 ng / mL IFN - ⁇ purchased from Peprotech
  • RPMI 1640 medium containing no FBS (Fetal Bovine Serum) for the THP-1 derived macrophages and dexamethasone purchased from Sigma
  • the exosomes used in the kit were cultured and cultured for 24 hours and then treated with 100 ng / mL LPS (purchased from Sigma) and 20 ng / mL IFN-y (purchased from Peprotech) And incubated for 4 hours to activate THP-1-derived macrophages.
  • CDNA was prepared from the RNA isolated from cultured THP-1 derived macrophages, and cDNA was prepared from the pro-inflammatory cytokines IL-6, IL-1 ⁇ And TNF-alpha mRNA expression levels were measured. GAPDH was used as a standard gene for quantifying the genes.
  • the types and sequences of the primers used in the PCR are shown in Table 2 below.
  • the forward primer (5 '- > 3')
  • the reverse primer (5 '- > 3') IL-6 AAGCCAGAGCTGTGCAGATGAGTA (SEQ ID NO: 11) TGTCCTGCAGCCACTGGTTC (SEQ ID NO: 12) IL-1?
  • THP-1 derived macrophages obtained after differentiating THP-1 monocytes into macrophages were treated with LPS and IFN- co-treatment
  • mRNA expression of IL-6, a proinflammatory cytokine was decreased compared to negative control.
  • THP-1 derived macrophages obtained after differentiating THP-1 monocytes into macrophages were used in the exosome kit of the present invention before LPS and IFN- IL-6, IL-1 ⁇ and TNF- ⁇ mRNA expression levels were decreased in the pre-treatment of exosomes compared to negative control.
  • the proinflammatory cytokines IL-6, IL-1 [beta] and TNF- [alpha] increase expression in proinflammatory M1 macrophages and decrease expression in anti-inflammatory M2 macrophages.
  • the exosome used in the exosome kit of the present invention can inhibit, alleviate and reduce the inflammatory reaction and the dermatitis caused thereby, and the exosome kit of the present invention can prevent, , which may be useful for improving, alleviating, or treating a disease.
  • Example 7 Skin permeability test of exosome used in the exosome kit of the present invention
  • PKH67 dye (purchased from Sigma) was used to prepare fluorescently stained exosomes. 1 mM PKH67 was diluted in Diluent C (purchased from Sigma) to prepare a 10 ⁇ M PKH67 solution, mixed with an appropriate concentration of exosome solution, and allowed to react at room temperature for 10 minutes in the absence of light. After the reaction, an MW3000 spin column (purchased from ThermoFisher Scientific) was used to remove the remaining PKH67 dye from the exosomes stained with PKH67 (hereinafter abbreviated as "PKH-exosomes"). After confirming the use of a fluorescence meter (purchased from Molecular Devices), PKH67 not reacted with exosomes was removed, and fluorescence of sufficient intensity was detected in PKH-exosomes (FIG. 10).
  • PKH-exosomes were dispersed in phosphate buffer (PBS) at a proper concentration, for example, 1 ⁇ 10 5 to 1 ⁇ 10 9 particles / mL, and then applied to the outer surface of the pig skin.
  • PBS phosphate buffer
  • the nonwoven fabric was covered and allowed to react for an appropriate period of time, for example, 30 minutes to 1 hour to allow the PKH-exosomes to be delivered to the subcutaneous tissues.
  • the PKH-exosomes were applied to the outer surface of the pig skin and covered with the nonwoven fabric, followed by flowing a microcurrent for a predetermined time, for example, 30 minutes to 1 hour.
  • the pig skin tissue was immersed in a 3.7% formaldehyde solution, reacted overnight, and washed three times for 5 minutes each with phosphate buffered saline.
  • the washed pig skin tissue was immersed in a 30% sucrose solution and treated with OCT compound.
  • sections were prepared on a longitudinal section using a microtome, and tissue sections were placed on a slide glass.
  • the preparation of the tissue section may be carried out before the tissue is fixed with formaldehyde solution. Fluorescence detected from PKH-exosomes in tissue sections was observed using fluorescence microscopy.
  • the exosomes used in the exosome kit of the present invention can be effectively passed through the epidermis and deeply transferred into the skin tissue, and can be effectively absorbed to the skin. Therefore, the exosomic kit of the present invention is expected to be usefully used for preventing, suppressing, improving, alleviating or treating dermatitis.
  • PKH-exosomes were dispersed in phosphate buffer (PBS) at a proper concentration, for example, 1 ⁇ 10 5 to 1 ⁇ 10 9 particles / mL, and then applied to the outside of the skin tissue of the mice.
  • PBS phosphate buffer
  • the nonwoven fabric was pre-positioned outside the mouse skin tissue, and PKH-exosomes were injected between the nonwoven fabric and skin tissue. Then, the reaction was carried out for 30 minutes to 1 hour.
  • the PKH-exosomes were injected between the nonwoven fabric and the skin tissue, and the microcurrent was flowed for a predetermined time, for example, 30 minutes to 1 hour.
  • the PKH-exosomes transferred into the skin tissue were confirmed immediately using a confocal fluorescence microscope (Leica, SP8X) or the skin tissue and the PKH-exosomal solution were further reacted for 1 hour to 6 hours , And PKH-exosomes were identified by confocal fluorescence microscopy.
  • a confocal fluorescence microscope Leica, SP8X
  • PKH-exosomes were identified by confocal fluorescence microscopy.
  • exosome kits of the present invention can effectively pass through the epidermis to effectively prevent the dermatitis from being prevented, inhibited, alleviated, improved or treated when the exosome kit of the present invention is applied to the skin.
  • Example 8 Treatment of exosome kit against human skin
  • the exosome kit 100 includes a liquid immersion sheet mask 10 (a white sheet mask on which a composition containing a stem cell-derived exosome is applied or immersed) And a transdermal permeation promoting sheet mask (silver sheet mask) 20 which is an iontophoresis device.
  • a stem cell-derived exosome (12) is applied or deposited on the surface or inside of the liquid immersion sheet mask (10).
  • a battery 22 for generating a microcurrent is mounted on the percutaneous permeation enhancing sheet mask 20 and a conductive pattern 24 electrically connected to the cell 22 is attached to the percutaneous permeation enhancing sheet mask 20, So that the micro current generated from the battery 22 can be uniformly transmitted to the entirety of the percutaneous permeation enhancing sheet mask 20.
  • the conductive pattern 24 is formed by printing or plating a conductive metal such as gold, silver, or copper.
  • exosome kit (100) having the above-described structure is applied once to the face of a person having a flushing symptom and skin troubles (hereinafter referred to as "case 1") related to the dermatitis symptom, It was evaluated whether the symptom of trouble and redness was alleviated or improved.
  • the exosome kit 100 of the present invention is applied once to the face of a person having flushing symptoms (hereinafter referred to as " case 2 ") associated with dermatitis symptoms to evaluate whether the overall skin tone and flushing symptoms are improved Respectively.
  • a liquid immersion sheet mask 10 containing 5 ⁇ 10 5 exosomes 12 was uniformly adhered to the entire faces of the cases 1 and 2 by aligning them with the eyes and mouth parts and then a microcurrent of about 0.3 to 0.4 mA
  • An activated percutaneous permeation enhancing sheet mask 20 which was activated to flow was laminated on the liquid immersion sheet mask 10 and used for about 25 to 30 minutes.
  • the exosome kit 100 composed of the liquid-immersion sheet mask 10 containing the stem cell-derived exosome 12 at a concentration of 5 ⁇ 10 5 particles / mask and the percutaneous permeation- (See case 1 in FIG. 14), the overall skin tone was improved in "case 2" and the flushing was alleviated (see case 2 in FIG. 14) . Therefore, the exosomal kit (present invention) comprising the liquid immersion sheet mask 10 in which the composition containing the stem cell-derived exosome as an active ingredient is immersed and the percutaneous permeation promoting sheet mask (iontophoresis device) 100) can be said to have an effect of preventing, suppressing, alleviating or alleviating dermatitis symptoms and redness symptoms and skin troubles associated therewith.
  • compositions containing a stem cell-derived exosome at a concentration of 0.29 x 10 8 particles / mL was applied to the affected part (hand, neck, arm, etc.) of three severe atopic patients complaining of itching Iontophoresis was carried out by using a iontophoresis device (IONZYME DF MACHINE) (purchased from Environ) to flow a micro current of 0.4 mA for 20 minutes to the affected part of the composition.
  • IONZYME DF MACHINE purchased from Environ
  • the exosome kit 100 of the present invention can be effectively used for preventing, suppressing, ameliorating, alleviating or treating dermatitis and symptoms associated therewith, as confirmed by the clinical test as described above.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Birds (AREA)
  • Reproductive Health (AREA)
  • Dermatology (AREA)
  • Mycology (AREA)
  • Nutrition Science (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention provides an exosome kit comprising an iontophoresis device with a mask pack, a mask sheet or a patch to which a composition comprising a stem cell-derived exosome as an active ingredient is applied or immersed in for preventing, suppressing, alleviating, ameliorating, or treating dermatitis. The exosome kit of the present invention exhibits excellent effects in skin care such as ameliorating dermatitis symptoms and skin condition that has deteriorated thereby and recovering the same to a normal condition, and exhibits effects of alleviating and ameliorating a diseased skin condition due to dermatitis or recovering the diseased skin condition to a normal condition. Particularly, the exosome kit of the present invention allows the exosomes applied to or immersed in the mask pack, the mask sheet or the patch to effectively pass through the epidermis and to permeate deeply into the skin by an iontophoresis action, and thus exhibits an effect of alleviating or ameliorating the dermatitis symptoms and the deteriorated skin condition related thereto.

Description

줄기세포 유래의 엑소좀을 포함하는 엑소좀 키트의 새로운 용도New Uses of Exosome Kit Containing Exosomes from Stem Cells
본 발명은 줄기세포 유래의 엑소좀을 포함하는 엑소좀 키트의 새로운 용도에 관한 것이다.The present invention relates to a new use of an exosome kit comprising exosomes derived from stem cells.
구체적으로, 본 발명은 피부염의 예방, 억제, 완화, 개선, 또는 치료를 위해, 줄기세포 유래의 엑소좀을 유효성분으로 포함하는 조성물이 도포되거나 침적된 마스크팩, 마스크시트 또는 패취와, 이온토포레시스(iontophoresis) 디바이스를 포함하는 엑소좀 키트를 제공하는 것에 관한 것이다. Specifically, the present invention provides a method for preventing, suppressing, alleviating, ameliorating, or treating dermatitis, which comprises applying a mask pack, a mask sheet or a patch containing a stem cell-derived exosome as an active ingredient, Lt; RTI ID = 0.0 > iontophoresis < / RTI > device.
인체의 피부는 물리적, 화학적으로 외부로부터 신체를 보호하는 동시에 전신의 대사에 필요한 생화학적 기능을 수행하는 기관이다. 일반적으로 사람의 피부에 나타나는 염증성 피부질환을 피부염이라고 한다. 비교적 흔한 피부염(dermatitis)으로는 아토피성 피부염(atopic dermatitis), 접촉성 피부염(contact dermatitis), 지루성 피부염(seborrhoic dermatitis) 등이 있다.The skin of the human body is an organ that physically and chemically protects the body from the outside and performs the biochemical functions necessary for the metabolism of the whole body. In general, inflammatory skin diseases that appear on human skin are called dermatitis. Relatively common dermatitis includes atopic dermatitis, contact dermatitis, and seborrhoic dermatitis.
접촉성 피부염은 외부 물질과의 접촉에 의해 발생하는 피부염이다. 접촉성 피부염은 발생하는 기전에 따라 자극성 접촉 피부염과 알레르기성 접촉 피부염이 있으며, 한 가지 물질이 이러한 두 가지 반응을 동시에 일으킬 수 있다. 접촉성 피부염을 일으키는 물질의 대부분은 유기 화합물이다. 이러한 물질에 감작된 피부에 재차 피부염 유발 물질이 접촉하게 되면 기억세포가 이를 감지하여 여러 화학 물질이 분비되어 염증을 일으키는 것으로 알려져 있다.Contact dermatitis is a dermatitis caused by contact with foreign substances. Contact dermatitis is characterized by irritant contact dermatitis and allergic contact dermatitis, depending on the mechanism by which one substance can cause both of these reactions simultaneously. Most of the substances that cause contact dermatitis are organic compounds. When the dermatitis-inducing substance comes into contact with the skin sensitized to these substances, it is known that the memory cell senses it and secretes various chemicals to cause inflammation.
아토피성 피부염은 가려움과 습진성 병변이 특징인 만성적인 염증성 피부질환이다. 지금까지의 연구결과에 따르면 아토피성 피부염의 발병에는 여러 가지 인자들이 관여하는 것으로 알려져 있다. 아토피성 피부염이 유발될 경우 호산구(eosinophil), 호중구(neutrophil) 및 비만세포(mast cell) 같은 염증 관련 세포가 병변 부위에서 관찰되며, 비만 세포로부터 생성되는 면역글로불린 IgE가 증가한다. 또한 T 세포의 이상증식이 나타나는데, 이 과정에서 염증성 사이토카인인 IL-4, IL-5, IL-13 등이 증가하여 면역반응을 증폭시킨다. 최근에는 TSLP(Thymic stromal lymphopoietin)가 피부염의 중증도와 연관되어 있고, 아토피성 피부염에서 나타나는 가려움 증세의 원인이라고 알려졌다. 또한, 지루성 피부염은 피지샘의 활동이 증가되어 피지 분비가 왕성한 두피와 얼굴, 그 중에서도 눈썹, 코, 입술 주위, 귀, 겨드랑이, 가슴, 서혜부 등에 발생하는 만성 염증성 피부 질환이다.Atopic dermatitis is a chronic inflammatory skin disease characterized by itchy and eczematous lesions. According to the results of previous studies, it is known that various factors are involved in the onset of atopic dermatitis. When atopic dermatitis is induced, inflammatory cells such as eosinophil, neutrophil and mast cell are observed in the lesion and immunoglobulin IgE produced from mast cells increases. In addition, IL-4, IL-5, and IL-13, which are inflammatory cytokines, are amplified and amplified by this process. Recently, TSLP (Thymic stromal lymphopoietin) has been associated with the severity of dermatitis and it is known to cause itching symptoms in atopic dermatitis. In addition, seborrheic dermatitis is a chronic inflammatory skin disease that occurs due to increased activity of sebaceous gland, which occurs in the scalp and face, especially around eyebrows, nose, lips, ears, underarms, chest, groin and the like.
염증 및 면역 억제를 통해 피부염 치료제를 개발하고자 하는 노력들이 있다. 현재까지 개발된 피부염 치료제로는 스테로이드, 항히스타민제, 및 사이클로스포린 A와 같은 면역 억제제 등이 있다. 그러나, 이들 약제는 피부 위축, 혈관 확장, 색소 탈실, 주사 부위의 과민반응, 내성 및 호중구감소증(neutropenia) 등의 심각한 부작용을 일으키는 문제점이 있다.There are efforts to develop dermatitis treatments through inflammation and immunosuppression. Drug remedies developed to date include steroids, antihistamines, and immunosuppressants such as cyclosporin A. However, these drugs have serious problems such as skin atrophy, vasodilation, pigment desalination, hypersensitivity of injection site, tolerance and neutropenia.
한편, 최근 세포 분비물(secretome)에 세포의 행동(behavior)을 조절하는 다양한 생체활성인자가 포함되어 있다는 연구가 보고되고 있으며, 특히 세포 분비물 내에는 세포 간 신호전달 기능을 갖는 '엑소좀(exosome)'이 포함되어 있어 그 성분과 기능에 대한 연구가 활발히 진행 중에 있다. Recently, there have been reported studies on the secretion of various bioactive factors that regulate cell behavior, and in particular, exosome (exosome), which has intercellular signal transduction, , And the research on the composition and function thereof is actively under way.
세포는 세포외 환경에 다양한 막(membrane) 유형의 소포체를 방출하는데, 통상 이러한 방출 소포체들을 세포외 소포체(Extracellular vesicles, EV)라고 부르고 있다. 세포외 소포체는 세포막 유래 소포체, 엑토좀(ectosomes), 쉐딩 소포체(shedding vesicles), 마이크로파티클(microparticles), 엑소좀 등으로 불려지기도 하며, 경우에 따라서는 엑소좀과는 구별되어 사용되기도 한다.Cells release various membrane-type vesicles in the extracellular environment, and these release vesicles are commonly referred to as extracellular vesicles (EVs). The extracellular endoplasmic reticulum is sometimes referred to as cell membrane-derived endoplasmic reticulum, ectosomes, shedding vesicles, microparticles, exosomes and, in some cases, differentiated from exosomes.
엑소좀은 세포막의 구조와 동일한 이중인지질막으로 이루어진 수십 내지 수백 나노미터 크기의 소포체로서 내부에는 엑소좀 카고(cargo)라고 불리는 단백질, 핵산(mRNA, miRNA 등) 등이 포함되어 있다. 엑소좀 카고에는 광범위한 신호전달 요소들(signaling factors)이 포함되며, 이들 신호전달 요소들은 세포 타입에 특이적이고 분비세포의 환경에 따라 상이하게 조절되는 것으로 알려져 있다. 엑소좀은 세포가 분비하는 세포 간 신호전달 매개체로서 이를 통해 전달된 다양한 세포 신호는 표적 세포의 활성화, 성장, 이동, 분화, 탈분화, 사멸(apoptosis), 괴사(necrosis)를 포함한 세포 행동을 조절한다고 알려져 있다. 엑소좀은 유래된 세포의 성질 및 상태에 따라 특이적인 유전물질과 생체활성 인자들이 포함되어 있다. 증식하는 줄기세포 유래 엑소좀의 경우 세포의 이동, 증식 및 분화와 같은 세포 행동을 조절하고, 조직 재생과 관련된 줄기세포의 특성이 반영되어 있다(Nature Review Immunology 2002 (2) 569-579). Exosome is an endoplasmic reticulum of several tens to several hundreds of nanometers in size, composed of double lipid membranes identical to the cell membrane structure, and contains proteins, nucleic acids (mRNA, miRNA, etc.) called exosomal cargo inside. Exosomal cargo contains a wide range of signaling factors, which are known to be specific for cell types and differentially regulated by the environment of the secretory cells. Exosome is an intercellular signaling mediator that is secreted by the cell, and various cell signals transmitted through it regulate cell behavior including activation, growth, migration, differentiation, de-differentiation, apoptosis, and necrosis of target cells It is known. Exosomes contain specific genetic material and bioactivity factors depending on the nature and condition of the derived cells. The proliferating stem cell-derived exosomes regulate cell behavior such as cell migration, proliferation and differentiation, and reflect the characteristics of stem cells involved in tissue regeneration (Nature Review Immunology 2002 (2) 569-579).
그러나 엑소좀을 이용한 특정 질환의 치료에 대한 가능성 제시 등 다양한 연구가 이루어지고 있음에도 불구하고, 엑소좀을 안정적으로 유지·보관할 수 있는 새로운 제형 개발과 엑소좀의 사용 편의성 및 효능 증대 등을 위한 다양한 의료 내지는 미용기술과의 접목은 상대적으로 주목받고 있지 못하고 있다. However, in spite of various studies such as the possibility of treatment of specific diseases using exosome, there have been many studies to develop a new formulation capable of stably maintaining and storing exosome, various medical treatments Or beauty technology has not received much attention.
본 발명자들은 엑소좀을 안정적으로 유지·보관할 수 있는 새로운 제형 개발과 의료 내지는 미용기술과의 접목에 대해 예의 연구를 거듭하던 중, 피부염 증상의 예방, 억제, 완화, 개선 또는 치료에 응용가능한 엑소좀 키트를 개발하여 본 발명을 완성하였다. The present inventors have conducted intensive studies on the development of new formulations capable of stably maintaining and storing exosomes and the combination of medical or cosmetic techniques, and have found that exosomes which can be applied to prevent, inhibit, alleviate, The kit was developed to complete the present invention.
한편, 상기한 배경기술로서 설명된 사항들은 본 발명의 배경에 대한 이해 증진을 위한 것일 뿐, 본 발명의 "선행 기술"로서 이용될 수 있다는 승인으로서 인용한 것은 아님을 이해하여야 한다.It should be understood, however, that the foregoing description of the background art is merely for the purpose of promoting an understanding of the background of the present invention, and not as an admission that it can be used as the " prior art "
본 발명의 목적은 줄기세포 유래의 엑소좀을 포함하는 엑소좀 키트 및 이의 응용을 제공하는데 있다. 구체적으로, 본 발명의 목적은 피부염의 예방, 억제, 완화, 개선, 또는 치료를 위해, 줄기세포 유래의 엑소좀을 유효성분으로 포함하는 조성물이 도포되거나 침적된 마스크팩, 마스크시트 또는 패취와, 이온토포레시스(iontophoresis) 디바이스를 포함하는 엑소좀 키트를 제공하는데 있다.It is an object of the present invention to provide an exosome kit comprising exosome derived from stem cells and an application thereof. Specifically, the object of the present invention is to provide a mask pack, a mask sheet or a patch which is coated or immersed with a composition containing an exosome derived from a stem cell as an active ingredient for the prevention, inhibition, alleviation, improvement or treatment of dermatitis, And an iontophoresis device for use in the treatment of cancer.
본 발명의 다른 목적은 상기 엑소좀 키트를 이용하여, 치료용을 제외한 피부염의 예방, 억제, 완화 또는 개선을 통해 포유동물 피부의 상태를 조절하는 미용방법을 제공하는데 있다.Another object of the present invention is to provide a cosmetic method for controlling the condition of mammalian skin through prevention, suppression, alleviation or improvement of dermatitis except for therapeutic use using the exosomic kit.
그러나, 전술한 바와 같은 본 발명의 과제는 예시적인 것으로, 이에 의해 본 발명의 범위가 제한되는 것은 아니다. 또한, 본 발명의 다른 목적 및 이점은 하기의 발명의 상세한 설명, 청구범위 및 도면에 의해 보다 명확하게 된다.However, the above-described problems of the present invention are illustrative and not intended to limit the scope of the present invention. Other objects and advantages of the present invention will become more apparent from the following detailed description of the invention, claims and drawings.
상기와 같은 목적을 달성하기 위하여, 본 발명은 피부염의 예방, 억제, 완화, 개선, 또는 치료를 위해, 줄기세포 유래의 엑소좀을 유효성분으로 포함하는 조성물이 도포되거나 침적된 마스크팩, 마스크시트 또는 패취와, 이온토포레시스(iontophoresis) 디바이스를 포함하는 엑소좀 키트를 제공한다.In order to achieve the above object, the present invention provides a method for preventing, suppressing, alleviating, ameliorating, or treating dermatitis, which comprises applying a mask pack containing a composition containing an exosome derived from stem cells as an active ingredient, Or patch, and an iontophoresis device.
본 명세서에서 용어, "엑소좀(exosomes)"은 세포막의 구조와 동일한 이중인지질막으로 이루어진 수십 내지 수백 나노미터(바람직하게는 대략 30~200 nm) 크기의 소포체를 의미한다(단, 분리 대상이 되는 세포 종류, 분리방법 및 측정방법에 따라 엑소좀의 입자 크기는 가변될 수 있음)(Vasiliy S. Chernyshev et al., "Size and shape characterization of hydrated and desiccated exosomes", Anal Bioanal Chem, (2015) DOI 10.1007/s00216-015-8535-3). 엑소좀에는 엑소좀 카고(cargo)라고 불리는 단백질, 핵산(mRNA, miRNA 등) 등이 포함되어 있다. 엑소좀 카고에는 광범위한 신호전달 요소들(signaling factors)이 포함되며, 이들 신호전달 요소들은 세포 타입에 특이적이고 분비세포의 환경에 따라 상이하게 조절되는 것으로 알려져 있다. 엑소좀은 세포가 분비하는 세포 간 신호전달 매개체로서 이를 통해 전달된 다양한 세포 신호는 표적 세포의 활성화, 성장, 이동, 분화, 탈분화, 사멸(apoptosis), 괴사(necrosis)를 포함한 세포 행동을 조절한다고 알려져 있다.As used herein, the term " exosomes " refers to an endoplasmic reticulum of several tens to several hundred nanometers (preferably about 30 to 200 nm) in size, consisting of double lipid membranes identical in structure to the cell membrane The particle size of the exosome can be varied according to the cell type, the separation method and the measurement method) (Vasiliy S. Chernyshev et al., "Size and shape characterization of hydrated and desiccated exosomes", Anal Bioanal Chem, DOI 10.1007 / s00216-015-8535-3). Exosomes contain proteins called exosomal cargo (cargo), nucleic acids (mRNA, miRNA, etc.). Exosomal cargo contains a wide range of signaling factors, which are known to be specific for cell types and differentially regulated by the environment of the secretory cells. Exosome is an intercellular signaling mediator that is secreted by the cell, and various cell signals transmitted through it regulate cell behavior including activation, growth, migration, differentiation, de-differentiation, apoptosis, and necrosis of target cells It is known.
한편, 본 명세서에서 사용된 "엑소좀"이란 용어는 다양한 줄기세포에서 분비되어 세포외 공간으로 방출된 나노 크기의 베지클 구조를 갖고 있고 엑소좀과 유사한 조성을 갖는 베지클(예를 들어, 엑소좀-유사 베지클)을 모두 포함하는 것을 의미한다. 상기 줄기세포의 종류는 제한되지 않으나, 본 발명을 한정하지 않는 하나의 예시로서, 바람직하게는 중간엽 줄기세포, 예를 들어 지방, 골수, 제대 또는 제대혈 유래 줄기세포일 수 있으며, 보다 바람직하게는 지방 유래 줄기세포일 수 있다. 상기 지방 유래 줄기세포의 종류는 병원체에 의한 감염의 위험이 없고 면역 거부 반응을 일으키지 않는 것이라면 제한되지 않으나, 바람직하게는 인간지방 유래 줄기세포일 수 있다.As used herein, the term " exosome " refers to a vesicle that has a nano-sized vesicle structure secreted from various stem cells and released into the extracellular space and has a composition similar to exosome (e.g., - pseudo-bezacl). The type of the stem cell is not limited, but may be, for example, a mesenchymal stem cell, for example, a fat, a bone marrow, an umbilical cord or a cord blood derived stem cell, Derived stem cells. The type of the adipose-derived stem cell is not limited as long as it does not cause a risk of infection by a pathogen and does not cause an immune rejection reaction, but it may be preferably a human adipose-derived stem cell.
그러나 본 발명에서 사용되는 줄기세포 유래의 엑소좀은 피부염의 예방, 억제, 완화, 개선 또는 치료에 효과가 있고 인체에 불리한 작용을 일으키지 않는 것이라면 당업계에서 사용되고 있거나 향후 사용될 수 있는 다양한 줄기세포 유래의 엑소좀을 사용할 수 있음은 물론이다. 따라서, 후술하는 실시예들의 분리방법에 따라 분리된 엑소좀은 본 발명에서 사용될 수 있는 엑소좀의 일례로서 이해되어야 하며, 본 발명은 이에 제한되는 것이 아님을 명백히 밝혀 둔다.However, the stem cell-derived exosome used in the present invention is effective for preventing, suppressing, alleviating, ameliorating or treating dermatitis and not causing adverse effects on the human body, so long as the stem cell-derived exosome can be used in a variety of stem cell- Of course, exosomes can be used. Therefore, it should be understood that the exosome isolated according to the separation method of the following embodiments should be understood as an example of exosome that can be used in the present invention, and the present invention is not limited thereto.
본 명세서에서 용어, "이온토포레시스(iontophoresis)"는 유효물질이 적용된 피부에 미세전류를 흐르게 하여 전위차를 주어 피부의 전기적 환경을 변화시킴으로써 이온화된 유효성분을 전기적 반발력으로 피부를 투과하게 하는 방법을 의미한다. 본 발명의 일 구체예에 사용되는 이온토포레시스(iontophoresis)는 피부 위의 전극 패치에 외부전원으로부터의 전류가 흘러들어가 피부에 미세전류가 도입되는 방식, 전극 패치 자체에 배터리가 장착되어 피부에 미세전류가 도입되는 방식, 고농도 전해질 용액 및 저농도 전해질 용액 간의 이온 농도 차이를 통해 전류를 발생시키는 역전기투석(Reversed Electrodialysis) 수단이 장착된 패치를 통해 피부에 미세전류가 도입되는 방식 등을 포함할 수 있다. 그러나, 본 발명은 이에 제한되는 것이 아니며, 다양한 방식의 이온토포레시스가 사용될 수 있음은 물론이다.The term " iontophoresis " as used herein refers to a method in which an ionized active ingredient is allowed to permeate through the skin by an electrical repulsive force by applying a minute current to the skin to which the effective substance is applied, . The iontophoresis used in one embodiment of the present invention is a method in which a current flows from an external power source to an electrode patch on the skin to introduce minute current into the skin, A method in which minute current is introduced, a method in which minute current is introduced into skin through a patch equipped with a reversed electrodialysis means for generating current through a difference in ion concentration between a high concentration electrolyte solution and a low concentration electrolyte solution, and the like . However, the present invention is not limited thereto, and it is needless to say that various types of iontophoresis can be used.
본 발명의 일 구체예의 엑소좀 키트는, 줄기세포 유래의 엑소좀을 유효성분으로 포함하는 조성물이 도포되거나 침적된 제1 마스크팩, 마스크시트 또는 패취와, 상기 조성물이 적용된 포유동물의 피부에 미세전류를 흘려 주는 이온토포레시스 디바이스를 포함한다. The exosome kit of one embodiment of the present invention comprises a first mask pack, a mask sheet or a patch which is applied or immersed with a composition containing an exosome derived from a stem cell as an active ingredient, And an iontophoresis device for flowing an electric current.
예를 들어, 본 발명의 일 구체예의 엑소좀 키트는, 상기 제1 마스크팩, 마스크시트 또는 패취를 포유동물의 피부 위에 접촉 또는 부착시키고, 상기 제1 마스크팩, 마스크시트 또는 패취 위에 상기 이온토포레시스 디바이스를 위치시킴으로써, 상기 제1 마스크팩, 마스크시트 또는 패취가 접촉 또는 부착되어 상기 조성물이 적용된 포유동물의 피부에 미세전류를 흘려 주는 방식으로 사용될 수 있다.For example, an exosome kit according to one embodiment of the present invention is characterized in that the first mask pack, the mask sheet or the patch is brought into contact with or attached to the skin of the mammal, and the ion mask By positioning the phoresis device, the first mask pack, mask sheet or patch can be contacted or attached to be used in such a way that micro-currents flow to the skin of the mammal to which the composition is applied.
또한, 상기 조성물은 엑소좀 이외에, 그 작용(피부염 증상 완화 내지는 개선 등)을 손상시키지 않는 한도에서 종래부터 사용된 피부염 치료제 및/또는 보습제를 함께 혼합하여 사용할 수 있다. 예를 들어, 상기 조성물은 하이드로겔, 히알루론산, 히알루론산 염(예를 들어 히알루론산 나트륨 등), 또는 히알루론산 겔 중 적어도 1종에 담지되거나 혼합될 수 있다. 상기 하이드로겔의 종류는 제한되지 않으나, 바람직하게는 겔화 고분자를 다가 알코올에 분산시켜 얻은 하이드로겔일 수 있다. 상기 겔화 고분자는 플루로닉, 정제한천, 아가로오스, 젤란검, 알긴산, 카라기난, 카시아검, 잔탄검, 갈락토만난, 글루코만난, 펙틴, 셀룰로오스, 구아검 및 로커스트빈검으로 이루어진 군으로부터 선택된 적어도 1종일 수 있고, 상기 다가 알코올은 에틸렌글리콜, 프로필렌글리콜, 1,3-부틸렌글리콜, 이소부틸렌글리콜, 디프로필렌글리콜, 소르비톨, 자일리톨 및 글리세린으로 이루어진 군으로부터 선택된 적어도 1종일 수 있다.In addition to the exosome, the composition may be used in combination with a conventionally used dermatitis remedy and / or moisturizer to the extent that its action (alleviation or improvement of dermatitis symptoms) is not impaired. For example, the composition may be supported on or mixed with at least one of hydrogel, hyaluronic acid, hyaluronic acid salt (for example, sodium hyaluronate), or hyaluronic acid gel. The type of the hydrogel is not limited, but it may preferably be a hydrogel obtained by dispersing a gelated polymer in a polyhydric alcohol. Wherein the gel polymer is at least one selected from the group consisting of pluronic, purified agar, agarose, gellan gum, alginic acid, carrageenan, cacao gum, xanthan gum, galactomannan, glucomannan, pectin, cellulose, guar gum and locust bean gum And the polyhydric alcohol may be at least one selected from the group consisting of ethylene glycol, propylene glycol, 1,3-butylene glycol, isobutylene glycol, dipropylene glycol, sorbitol, xylitol and glycerin.
본 발명의 일 구체예의 엑소좀 키트에 있어서, 상기 이온토포레시스 디바이스는 가요성 배터리, 리튬이온 이차 전지, 알칼리 전지, 건전지, 수은 전지, 리튬 전지, 니켈-카드뮴 전지, 및 역전기 투석 전지로 구성된 군으로부터 선택된 적어도 1종의 전지를 포함하거나, 상기 적어도 1종의 전지가 장착된 제2 마스크팩, 마스크시트 또는 패취일 수 있다.The iontophoresis device according to one embodiment of the present invention may be applied to a flexible battery, a lithium ion secondary battery, an alkaline battery, a dry battery, a mercury battery, a lithium battery, a nickel-cadmium battery, Or a second mask pack, a mask sheet, or a patch having the at least one battery mounted thereon.
본 발명의 일 구체예의 엑소좀 키트에 있어서, 상기 제1 마스크팩, 마스크시트 또는 패취 위에 상기 제2 마스크팩, 마스크시트 또는 패취가 적층되어 사용될 수 있다. In the exosome kit according to one embodiment of the present invention, the second mask pack, the mask sheet or the patch may be laminated on the first mask pack, the mask sheet or the patch.
본 발명의 일 구체예의 엑소좀 키트에 있어서, 상기 제1 마스크팩, 마스크시트 또는 패취의 적어도 일면(一面)에는 하이드로겔, 히알루론산, 히알루론산 염(예를 들어 히알루론산 나트륨), 또는 히알루론산 겔 중 적어도 1종이 도포될 수 있다. 상기 하이드로겔의 종류는 제한되지 않으나, 바람직하게는 겔화 고분자를 다가 알코올에 분산시켜 얻은 하이드로겔일 수 있다. 상기 겔화 고분자 및 다가 알코올은 앞선 설명들에서 예시된 것일 수 있다.In at least one surface of the first mask pack, the mask sheet or the patch, at least one surface of the first mask pack, the mask sheet or the patch may be coated with a hydrogel, hyaluronic acid, hyaluronic acid salt (for example, sodium hyaluronate) At least one of the gels may be applied. The type of the hydrogel is not limited, but it may preferably be a hydrogel obtained by dispersing a gelated polymer in a polyhydric alcohol. The gelled polymer and the polyhydric alcohol may be exemplified in the above description.
본 발명의 일 구체예의 엑소좀 키트에 따르면, 상기 이온토포레시스 디바이스로부터 미세전류가 흐름으로써 엑소좀이 표피를 효과적으로 통과하고 피부 내부 깊숙이 침투할 수 있게 된다.According to the exosome kit of one embodiment of the present invention, the micro current flows from the iontophoresis device so that the exosomes can effectively penetrate the epidermis and penetrate deeply into the skin.
본 발명의 일 구체예의 엑소좀 키트는 가려움을 수반하는 다양한 피부염의 예방, 억제, 개선, 완화 또는 치료에 효과적으로 이용될 수 있으며, 바람직하게는 접촉피부염, 자극 접촉피부염, 알레르기 접촉피부염, 광독성 및 광알레르기 접촉피부염, 접촉 두드러기 증후군, 아토피성 피부염, 지루성 피부염, 자가감작 피부염, 자가면역 프로게스테론 피부염, 울체 피부염, 여드름 또는 습진에 이용될 수 있다.The exosome kit of one embodiment of the present invention can be effectively used for preventing, suppressing, ameliorating, alleviating or treating various dermatitis accompanied by itch, and is preferably used for the treatment of contact dermatitis, irritant contact dermatitis, allergic contact dermatitis, Allergic contact dermatitis, contact urticaria, atopic dermatitis, seborrheic dermatitis, autoimmune dermatitis, autoimmune progesterone dermatitis, dermatitis, acne or eczema.
본 발명의 다른 구체예는 상기 엑소좀 키트를 이용하여, 치료용을 제외한 피부염의 예방, 억제, 완화 또는 개선을 통해 포유동물 피부의 상태를 조절하는 미용방법을 제공한다. 본 발명의 미용방법에 있어서, 피부의 상태 조절이란 피부의 상태를 개선시키고/시키거나 피부의 상태를 예방적으로 조절하는 것을 의미하고, 피부의 상태 개선이란 피부 조직의 외관 및 느낌의 시각적 및/또는 촉각적으로 지각할 수 있는 긍정적인 변화를 의미한다. 예를 들어, 피부의 상태 개선이란 피부염 관련 증상 완화 내지는 개선일 수 있다.Another embodiment of the present invention provides a cosmetic method for controlling the condition of a mammalian skin by preventing, suppressing, alleviating or improving dermatitis except for therapeutic use using the exosomic kit. In the cosmetic method of the present invention, conditioning of the skin means improvement of the condition of the skin and / or prevention of the condition of the skin, and improvement of the condition of the skin means visual and / Or a tactile perceptible positive change. For example, improvement of the condition of the skin may be mitigation or improvement of dermatitis related symptoms.
본 발명의 일 구체예의 미용방법은, 전술한 바와 같은 엑소좀 키트를 이용하여 수행될 수 있다. The cosmetic method of one embodiment of the present invention can be carried out using the exosome kit as described above.
예를 들어, 본 발명의 일 구체예의 미용방법은 (a) 줄기세포 유래의 엑소좀을 유효성분으로 포함하는 조성물이 도포되거나 침적된 제1 마스크팩, 마스크시트 또는 패취를 포유동물의 피부 위에 접촉 또는 부착하는 단계와, (b) 상기 제1 마스크팩, 마스크시트 또는 패취 위에 이온토포레시스 디바이스를 위치시키는 단계와, (c) 상기 제1 마스크팩, 마스크시트 또는 패취가 접촉 또는 부착되어 상기 조성물이 적용된 포유동물의 피부에 미세전류를 흘려 주는 이온토포레시스를 수행하는 단계와, (d) 상기 미세전류를 통하여 상기 엑소좀을 포유동물 피부 내부로 전달하는 단계를 포함한다.For example, the cosmetic method of one embodiment of the present invention comprises the steps of (a) contacting a first mask pack, a mask sheet or a patch with a composition containing an exosome derived from a stem cell as an active ingredient, (B) positioning the iontophoresis device on the first mask pack, mask sheet or patch, and (c) contacting the first mask pack, mask sheet or patch with the iontophoresis device, Performing iontophoresis to flow a microcurrent into the skin of the mammal to which the composition is applied; and (d) delivering the exosomes through the microcurrent to the inside of the mammalian skin.
본 발명의 일 구체예의 미용 방법에 있어서, 상기 제1 마스크팩, 마스크시트 또는 패취의 적어도 일면(一面)에는 하이드로겔, 히알루론산, 히알루론산 염(예를 들어 히알루론산 나트륨 등), 또는 히알루론산 겔 중 적어도 1종이 도포될 수 있다. 상기 하이드로겔의 종류는 제한되지 않으나, 바람직하게는 겔화 고분자를 다가 알코올에 분산시켜 얻은 하이드로겔일 수 있다. 상기 겔화 고분자 및 다가 알코올은 앞선 설명들에서 예시된 것일 수 있다.In at least one surface of the first mask pack, the mask sheet or the patch, at least one surface of the first mask pack, the mask sheet or the patch may be coated with a hydrogel, hyaluronic acid, hyaluronate (e.g., sodium hyaluronate) At least one of the gels may be applied. The type of the hydrogel is not limited, but it may preferably be a hydrogel obtained by dispersing a gelated polymer in a polyhydric alcohol. The gelled polymer and the polyhydric alcohol may be exemplified in the above description.
본 발명의 일 구체예의 미용 방법에 있어서, 상기 이온토포레시스 디바이스는 가요성 배터리, 리튬이온 이차 전지, 알칼리 전지, 건전지, 수은 전지, 리튬 전지, 니켈-카드뮴 전지, 및 역전기 투석 전지로 구성된 군으로부터 선택된 적어도 1종의 전지를 포함하거나, 상기 적어도 1종의 전지가 장착된 제2 마스크팩, 마스크시트 또는 패취일 수 있다. 상기 (b) 단계는 상기 제1 마스크팩, 마스크시트 또는 패취 위에 상기 제2 마스크팩, 마스크시트 또는 패취를 적층하여 수행될 수 있다.In the cosmetic method according to one embodiment of the present invention, the iontophoresis device comprises a flexible battery, a lithium ion secondary battery, an alkaline battery, a dry battery, a mercury battery, a lithium battery, a nickel-cadmium battery, Or a second mask pack, a mask sheet or a patch having the at least one battery mounted thereon. The step (b) may be performed by laminating the second mask pack, the mask sheet or the patch on the first mask pack, the mask sheet or the patch.
본 발명의 엑소좀 키트는 피부염 증상 및 이로 인해 나빠진 피부 상태를 개선 또는 정상상태로 회복시키는 등의 피부 미용에 있어서 우수한 효과를 나타내고, 피부염으로 인해 피부 상태가 병적 상태인 것을 완화, 개선 내지는 정상 상태로 회복시켜 주는 효과를 나타낸다. 특히, 본 발명의 엑소좀 키트는 이온토포레시스 작용에 의해 마스크팩, 마스크시트 또는 패취에 도포되거나 침적되어 있는 엑소좀이 표피를 효과적으로 통과하게 하고 피부 내부 깊숙이 침투시켜 피부염 증상 및 이와 관련하여 나빠진 피부상태를 개선 내지는 호전시키는 효과를 나타낸다.The exosome kits of the present invention exhibit excellent effects on dermatitis symptoms and skin cosmetics such as improving or restoring the dermatological conditions deteriorated due to the dermatitis symptoms, and it is possible to alleviate or improve the condition of the skin condition due to dermatitis, . Particularly, the exosome kit of the present invention can prevent the exosomes applied or deposited on the mask pack, the mask sheet or the patch by the iontophoresis action to effectively penetrate the epidermis and penetrate deeply into the skin, Thereby exhibiting an effect of improving or alleviating the skin condition.
한편, 전술한 바와 같은 효과들에 의해 본 발명의 범위가 제한되는 것은 아니다.On the other hand, the scope of the present invention is not limited by the above-mentioned effects.
도 1은 본 발명의 일 구체예에 따라 얻어진 엑소좀의 물리적 특성 분석 결과를 도시한 것이다. "도 1A"는 TRPS(tunable resistive pulse sensing) 분석에 의한 입자 크기 분포와 입자수를 나타낸다. "도 1B"는 NTA(nanoparticle tracking analysis) 분석에 의한 입자 크기 분포와 입자수를 나타낸다. "도 1C"는 TEM(transmitted electron microscopy) 분석에 의한 입자 이미지를 배율에 따라 도시하였다. "도 1D"는 본 발명의 일 구체예에 따라 얻어진 엑소좀의 웨스턴 블랏 결과를 나타낸다. "도 1E"는 본 발명의 일 구체예에 따라 얻어진 엑소좀에 대한 마커 분석에 있어서 CD63 및 CD81에 대한 유세포분석 결과를 나타낸다. FIG. 1 shows the results of physical property analysis of exosomes obtained according to one embodiment of the present invention. &Quot; Figure 1A " shows the particle size distribution and number of particles by TRPS (tunable resistive pulse sensing) analysis. &Quot; Figure 1B " shows particle size distribution and particle number by NTA (nanoparticle tracking analysis) analysis. &Quot; Fig. 1C " shows particle images by transmission electron microscopy (TEM) according to magnification. &Quot; Figure 1D " shows the Western blot results of exosomes obtained according to one embodiment of the present invention. ≪ RTI ID = 0.0 > 1E < / RTI > shows flow cytometric analysis results for CD63 and CD81 in marker assays for exosomes obtained according to one embodiment of the present invention.
도 2는 인체 피부섬유아세포인 HS68 세포에 본 발명의 일 구체예에 따라 수득된 엑소좀을 처리한 후 세포 독성이 없음을 확인한 결과를 도시한다.FIG. 2 shows the result of confirming the absence of cytotoxicity after treatment of exosomes obtained according to one embodiment of the present invention in human skin fibroblast HS68 cells.
도 3은 RAW 264.7 세포에 대해 LPS와 함께 본 발명의 엑소좀 키트에 사용되는 엑소좀을 처리한 경우 LPS에 의해 유도되는 TNF-α, IL-6, IL-1β 및 iNOS의 mRNA 발현량이 감소한 것을 확인한 리얼타임 PCR 결과를 도시한 그래프이다.FIG. 3 shows that the amount of mRNA expression of TNF-α, IL-6, IL-1β and iNOS induced by LPS was decreased when the exosome used in the exosome kit of the present invention was treated with LPS for RAW 264.7 cells And a real-time PCR result.
도 4는 본 발명의 엑소좀 키트에 사용되는 엑소좀에 의해 염증 반응의 일종인 NO 형성이 감소되는 것을 확인한 실험 결과를 도시한다. 도 4에서 PBS는 인산염 완충용액(phosphate-buffered saline), DEX는 덱사메타손(dexamethasone), EXO는 엑소좀, CM은 지방줄기세포 배양액(conditioned media), CM-EXO는 엑소좀이 제거된 지방줄기세포 배양액(exosome-depeleted conditioned media)을 나타낸다.FIG. 4 shows experimental results showing that NO formation, which is a type of inflammatory reaction, is reduced by the exosome used in the exosome kit of the present invention. In FIG. 4, PBS is phosphate-buffered saline, DEX is dexamethasone, EXO is exosome, CM is conditioned medium, and CM-EXO is adipocyte- (Exosome-depeleted conditioned media).
도 5는 본 발명의 엑소좀 키트에 사용되는 엑소좀에 의해 염증성 사이토카인인 TNF-α 형성이 감소되는 것을 확인한 실험 결과를 도시한다. 도 5에서, PBS는 인산염 완충용액(phosphate-buffered saline), DEX는 덱사메타손(dexamethasone), 각 숫자는 엑소좀 처리량(μg/mL)을 나타낸다.FIG. 5 shows experimental results confirming that TNF-.alpha. Formation, which is an inflammatory cytokine, is reduced by exosomes used in the exosome kit of the present invention. In FIG. 5, PBS represents phosphate-buffered saline, DEX represents dexamethasone, and each numeral represents exosome throughput (μg / mL).
도 6은 THP-1 단핵구를 대식세포로 분화시킨 후 얻은 THP-1 유래 대식세포에 대해, LPS 및 IFN-γ와 함께 본 발명의 엑소좀 키트에 사용되는 엑소좀을 처리(co-treatment)한 경우, 친염증성 사이토카인인 IL-6의 mRNA 발현량이 감소한 것을 확인한 리얼타임 PCR 결과를 도시한 그래프이다.FIG. 6 shows the results of co-treatment of exosomes used in the exosomal kit of the present invention together with LPS and IFN-y for THP-1-derived macrophages obtained after differentiating THP-1 monocytes into macrophages Time PCR results confirming that the mRNA expression level of the pro-inflammatory cytokine IL-6 was decreased.
도 7 내지 도 9는 THP-1 단핵구를 대식세포로 분화시킨 후 얻은 THP-1 유래 대식세포에 대해, 본 발명의 엑소좀 키트에 사용되는 엑소좀을 전처리(pre-treatment)하고 일정 시간 배양한 다음 LPS 및 IFN-γ를 처리한 경우, 친염증성 사이토카인인 IL-6, IL-1β 및 TNF-α의 mRNA 발현량이 감소한 것을 확인한 리얼타임 PCR 결과를 도시한 그래프이다.FIGS. 7 to 9 show the results of pre-treatment of the exosomes used in the exosomal kit of the present invention and culturing for a predetermined period of time on THP-1-derived macrophages obtained after differentiating THP-1 monocytes into macrophages Next, it is a graph showing real-time PCR results in which the amount of mRNA expression of IL-6, IL-1 [beta] and TNF- [alpha], which are proinflammatory cytokines, is decreased when LPS and IFN-y are treated.
도 10은 PKH67로 염색된 엑소좀을 확인한 형광강도 측정 결과를 도시한다.FIG. 10 shows fluorescence intensity measurement results of exosomes stained with PKH67.
도 11은 돼지 피부조직 내부로, 형광염색된 엑소좀이 전달된 정도를 확인한 형광현미경 이미지이다. Fig. 11 is a fluorescence microscope image showing the degree of transfer of fluorescent stained exosomes into pig skin tissue. Fig.
도 12는 생쥐 피부조직 내부로, 형광염색된 엑소좀이 전달된 정도를 확인한 공초점형광현미경 이미지이다. Fig. 12 is a confocal fluorescence microscope image showing the extent of fluorescence-stained exosomes transferred into the mouse skin tissue.
도 13은 도 12의 각 이미지 상의 형광강도를 측정하여 얻은 총 형광강도를 비교하여 도시한 그래프이다.13 is a graph showing a comparison of the total fluorescence intensity obtained by measuring the fluorescence intensity on each image in Fig.
도 14는 본 발명의 엑소좀 키트를 사람 피부에 처리하고 처리 전후의 피부 상태를 비교하는 사진이다. 14 is a photograph showing the skin condition before and after the treatment of human skin with the exosomal kit of the present invention.
도 15는 본 발명의 엑소좀 키트에 사용되는 엑소좀을 포함하는 조성물을 사람의 피부(환부)에 도포한 후 상기 조성물이 도포된 피부(환부)에 미세전류를 흘려주는 이온토포레시스를 수행한 결과, 피부염이 심한 피부(환부)의 홍반 등이 현저히 개선된 것을 보여주는 사진이다. FIG. 15 is a graph showing the results of application of iontophoresis for applying a microcurrent to a skin (affected part) coated with the composition after applying the composition containing exosome used in the exosome kit of the present invention to human skin As a result, it is a photograph showing that the erythema of the skin (the affected part) which has a severe dermatitis is remarkably improved.
도 16은 본 발명의 일 구체예에 따른 엑소좀 키트(100)의 구성을 나타내는 사시도이다. 16 is a perspective view showing a configuration of the exosome kit 100 according to one embodiment of the present invention.
이하 본 발명을 하기 실시예에서 보다 상세하게 기술한다. 다만, 하기 실시예는 본 발명의 내용을 예시하는 것일 뿐 본 발명의 권리범위를 제한하거나 한정하는 것이 아니다. 본 발명의 상세한 설명 및 실시예로부터 본 발명이 속하는 기술분야의 통상의 기술자가 용이하게 유추할 수 있는 것은 본 발명의 권리범위에 속하는 것으로 해석된다. 본 발명에 인용된 참고문헌들은 본 발명에 참고로서 통합된다.Hereinafter, the present invention will be described in more detail in the following Examples. It should be noted, however, that the following examples are illustrative only and do not limit or limit the scope of the present invention. It will be understood by those of ordinary skill in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention as defined by the appended claims. The references cited in the present invention are incorporated herein by reference.
명세서 전체에서, 어떤 부분이 어떤 구성 요소를 "포함"한다고 할 때, 이는 특별히 반대되는 기재가 없는 한 다른 구성 요소를 제외하는 것이 아니라 다른 구성 요소를 더 포함할 수 있는 것을 의미한다.Throughout the specification, when an element is referred to as " comprising ", it means that it can include other elements as well, without excluding other elements unless specifically stated otherwise.
실시예Example
실시예 1: 세포의 배양Example 1: Culture of cells
마우스 대식세포주인 RAW 264.7은 한국세포주은행에서 구입하여 배양하였다. 세포 배양을 위해 10% 우태아 혈청 (fetal bovine serum: ThermoFisher Scientific에서 구입) 및 1% 항생제-항진균제 (antibiotics-antimycotics: ThermoFisher Scientific에서 구입)가 함유된 DMEM (ThermoFisher Scientific에서 구입) 배지에 5% CO2, 37℃ 조건에서 계대 배양하였다.Mouse macrophage line RAW 264.7 was purchased from Korean Cell Line Bank and cultured. For cell culture, DMEM (purchased from ThermoFisher Scientific) medium containing 10% fetal bovine serum (purchased from ThermoFisher Scientific) and 1% antibiotic-antimycotics (purchased from ThermoFisher Scientific) 2 , and 37 ° C.
인체 피부 섬유아세포(human dermal fibroblast)인 HS68 세포는 ATCC에서 구입하여, 10% 우태아 혈청 (fetal bovine serum: ThermoFisher Scientific에서 구입) 및 1% 항생제-항진균제 (antibiotics-antimycotics: ThermoFisher Scientific에서 구입)가 함유된 DMEM (ThermoFisher Scientific에서 구입) 배지에 5% CO2, 37℃ 조건에서 계대 배양하였다.HS68 cells, a human dermal fibroblast, were purchased from ATCC and cultured in RPMI 1640 supplemented with 10% fetal bovine serum (purchased from ThermoFisher Scientific) and 1% antibiotic-antimycotics (purchased from ThermoFisher Scientific) The cells were subcultured in DMEM (purchased from ThermoFisher Scientific) medium containing 5% CO 2 at 37 ° C.
당해 발명이 속하는 기술분야에 알려진 세포배양 방법에 따라 5% CO2, 37℃ 조건에서 지방 유래 줄기세포를 배양하였다. 그 다음, 인산염 완충용액(phosphate-buffered saline)(ThermoFisher Scientific에서 구입)으로 세척 후, 무혈청, 무페놀레드 배지로 교체하여 1일 내지 10일간 배양하고 그 상층액(이하, 배양액)을 회수하였다.According to the cell culturing method known in the art, adipose-derived stem cells were cultured at 5% CO 2 and 37 ° C. Then, the cells were washed with a phosphate-buffered saline (purchased from ThermoFisher Scientific), replaced with serum-free, non-phenol red medium, cultured for 1 to 10 days, and the supernatant .
엑소좀의 분리 과정에서 입자크기 분포가 균일하고 순도가 높은 엑소좀을 수득하기 위하여 배양액에 트레할로오스를 2 중량% 첨가하였다. 트레할로오스를 첨가한 후 배양액을 0.22 μm 필터로 여과하여 세포 잔해물, 노폐물 및 거대 입자 등의 불순물을 제거해 주었다. 여과된 배양액은 즉시 분리 과정을 통해 엑소좀을 분리하였다. 또한, 여과된 배양액은 냉장고(영상 10℃ 이하)에서 보관한 후 엑소좀 분리에 사용하였다. 또한, 여과된 배양액은 -60℃ 이하의 초저온 냉동고에서 동결 보관하였다가 해동시킨 후 엑소좀 분리를 수행하였다. 이후, 배양액으로부터 접선흐름여과장치(Tangential Flow Filtration; TFF)를 이용하여 엑소좀을 분리하였다.Trehalose was added to the culture medium in an amount of 2% by weight in order to obtain an exosome having a uniform particle size distribution and high purity in the process of separating exosome. After the addition of trehalose, the culture was filtered with a 0.22 μm filter to remove impurities such as cellular debris, waste products and large particles. The filtered cultures were immediately separated to isolate exosomes. In addition, the filtered culture was stored in a refrigerator (image below 10 ° C) and used for exosome isolation. In addition, the filtered culture was frozen in an ultra-low temperature freezer at -60 ° C or lower, and thawed, followed by exosome isolation. Then, the exosomes were separated from the culture medium by using Tangential Flow Filtration (TFF).
실시예 2: TFF 방법에 의한 엑소좀의 분리 및 정제Example 2: Isolation and purification of exosome by TFF method
실시예 1에서 0.22 μm 필터로 여과된 배양액으로부터 엑소좀을 분리, 농축, 탈염과 버퍼교환(diafiltration)을 위해 TFF(Tangential Flow Filtration) 방법을 사용하였다. TFF 방법을 위한 필터로는 카트리지 필터(cartridge filter, 일명 hollow fiber filter; GE Healthcare에서 구입) 또는 카세트 필터(cassette filter; Pall 또는 Sartorius 또는 Merck Millipore에서 구입)를 사용하였다. TFF 필터는 다양한 분자량 차단(molecular weight cutoff; MWCO)에 의해 선택될 수 있다. 선택된 MWCO에 의해 선별적으로 엑소좀을 분리, 농축하였고, MWCO보다 작은 입자나 단백질, 지질, 핵산, 저분자 화합물 등은 제거하였다.In Example 1, a TFF (Tangential Flow Filtration) method was used for separating, concentrating, desalting and diafiltration exosomes from a culture filtrated with a 0.22 μm filter. For the TFF method, a cartridge filter (also called a hollow fiber filter (purchased from GE Healthcare) or a cassette filter (purchased from Pall or Sartorius or Merck Millipore) was used. The TFF filter can be selected by a variety of molecular weight cutoffs (MWCO). Exosomes were selectively isolated and concentrated by selected MWCOs, and particles, proteins, lipids, nucleic acids, low molecular weight compounds, etc. smaller than MWCO were removed.
엑소좀을 분리, 농축하기 위하여 MWCO 100,000 Da(Dalton), 300,000 Da, 또는 500,000 Da의 TFF 필터를 사용하였다. 배양액을 TFF 방법을 이용하여 1/100 내지 1/25 정도의 부피가 될 때까지 농축하면서, MWCO보다 작은 물질들은 제거하여 엑소좀을 분리하였다.To separate and concentrate the exosomes, TFF filters of MWCO 100,000 Da (Dalton), 300,000 Da, or 500,000 Da were used. The culture medium was concentrated to a volume of about 1/100 to 1/25 using the TFF method, and substances smaller than MWCO were removed to separate the exosomes.
분리, 농축된 엑소좀 용액은 TFF 방법을 이용하여 추가로 탈염과 버퍼교환(diafiltration)을 수행하였다. 이때, 탈염과 버퍼교환은 연속적으로 수행(continuous diafiltration)하거나 단속적으로 수행(discontinuous diafiltration)하였으며, 시작 부피(starting volume)에 대하여 적어도 4배, 바람직하게는 6배 내지는 10배 이상, 보다 바람직하게는 12배 이상의 부피를 갖는 완충용액을 이용하여 수행하였다. 완충용액에는 입자크기 분포가 균일하고 순도가 높은 엑소좀을 수득하기 위하여 PBS에 녹인 2 중량%의 트레할로오스를 첨가하였다. Separated and concentrated exosomal solutions were further desalted and buffered (diafiltration) using the TFF method. At this time, the desalination and buffer exchange are performed by continuous diafiltration or discontinuous diafiltration, and at least 4 times, preferably 6 times to 10 times or more, more preferably, Was performed using a buffer solution having a volume of 12 times or more. To the buffer solution, 2% by weight of trehalose dissolved in PBS was added to obtain an exosome having a uniform particle size distribution and high purity.
실시예 3: 분리된 엑소좀의 특성 분석Example 3: Characterization of isolated exosomes
분리된 엑소좀은 나노입자 트랙킹 분석(nanoparticle tracking analysis: NTA; Malvern에서 구입) 또는 가변 저항펄스 감지(tunable resistive pulse sensing: TRPS; Izon Science에서 구입)에 의해 입자의 크기와 농도를 측정하였다. 분리된 엑소좀의 균일도와 크기는 투과전자현미경(transmitted electron microscopy: TEM)을 이용하여 분석하였다. 본 발명의 일 구체예에 따라 분리된 엑소좀의 TRPS, NTA, TEM 분석 결과는 도 1A 내지 도 1C에 도시하였다.Separated exosomes were measured for particle size and concentration by nanoparticle tracking analysis (NTA; purchased from Malvern) or tunable resistive pulse sensing (TRPS; purchased from Izon Science). The uniformity and size of isolated exosomes were analyzed by transmission electron microscopy (TEM). The results of TRPS, NTA and TEM analysis of the exosomes isolated according to one embodiment of the present invention are shown in Figs. 1A to 1C.
도 1D는 본 발명의 일 구체예의 방법에 따라 분리된 엑소좀에 대해 웨스턴 블랏을 수행한 결과로서, CD9, CD63, CD81 및 TSG101 마커의 존재를 확인하였다. 각 마커에 대한 항체로는 각각 항-CD9 (Abcam에서 구입), 항-CD63 (System Biosciences에서 구입), 항-CD81 (System Biosciences에서 구입), 및 항-TSG101 (Abcam에서 구입)을 사용하였다.FIG. 1D shows the presence of CD9, CD63, CD81 and TSG101 markers as a result of performing Western blotting on isolated exosomes according to the method of one embodiment of the present invention. Anti-CD9 (purchased from Abcam), anti-CD63 (purchased from System Biosciences), anti-CD81 (purchased from System Biosciences), and anti-TSG101 (purchased from Abcam) were used as the antibodies for each marker.
도 1E는 본 발명의 일 구체예의 방법에 따라 분리된 엑소좀에 대해 유세포분석기를 이용하여 분석한 결과로서 CD63 및 CD81 마커의 존재를 확인하였다. CD63에 대해 양성(positive)인 엑소좀을 분리하기 위하여 엑소좀-휴먼 CD63 분리/검출 키트(ThermoFisher Scientific에서 구입)를 제조사의 방법에 따라 사용하였고, PE-마우스 항-인간 CD63 (PE-Mouse anti-human CD63)(BD에서 구입) 및 PE-마우스 항-인간 CD81 (PE-mouse anti-human CD81)(BD에서 구입)을 사용하여 마커를 염색한 후, 유세포분석기 (ACEA Biosciences)를 이용하여 분석하였다.FIG. 1E shows the presence of CD63 and CD81 markers as a result of analysis using flow cytometry on exosomes isolated according to the method of one embodiment of the present invention. Human CD63 isolation / detection kit (purchased from ThermoFisher Scientific) was used according to the manufacturer's method to isolate the positive exosomes for CD63 and PE-mouse anti-human CD63 (PE-Mouse anti markers were stained using the PE-mouse CD63 (purchased from BD) and PE-mouse anti-human CD81 (purchased from BD), and analyzed using a flow cytometer (ACEA Biosciences) Respectively.
한편, 본 발명에서 사용되는 줄기세포 유래의 엑소좀은 전술한 바와 같은 실시예들의 엑소좀에 제한되는 것이 아니며, 당업계에서 사용되고 있거나 향후 사용될 수 있는 다양한 줄기세포 유래의 엑소좀을 사용할 수 있음은 물론이다. 상기 실시예들에 따라 분리된 엑소좀은 본 발명에서 사용될 수 있는 줄기세포 유래의 엑소좀의 일례로서 이해되어야 하며, 본 발명은 이에 제한되는 것이 아님을 명백히 밝혀 둔다.Meanwhile, the exosomes derived from stem cells used in the present invention are not limited to the exosomes of the above-mentioned embodiments, and it is possible to use various stem cell-derived exosomes which are used in the art or can be used in the future Of course. It is to be understood that the exosomes isolated according to the above embodiments are to be understood as an example of exosomes derived from stem cells which can be used in the present invention, and the present invention is not limited thereto.
실시예 4: 엑소좀 처리에 따른 세포 독성 측정Example 4: Measurement of cytotoxicity by treatment with exosome
인체 피부 섬유아세포인 HS68 세포에서 본 발명의 일 구체예의 분리 방법에 따라 수득된 엑소좀의 독성을 평가하기 위해 세포에 농도별로 엑소좀을 처리하고 세포의 증식률을 확인하였다. HS68 세포를 10% FBS를 포함한 DMEM에 현탁시킨 후 80 내지 90%의 밀집도(confluency)를 갖도록 분주하고 37℃, 5% CO2 인큐베이터에서 24시간 배양하였다. 24시간 후, 배양액을 제거하고 실시예 2에서 준비된 엑소좀을 농도 별로 처리하여 24 내지 72시간 동안 배양하면서 세포 생존율을 평가하였다. 세포 생존율을 WST-1 시약(WST-1 reagent)(Takara에서 구입), MTT 시약(Sigma에서 구입), 셀타이터-글로 시약(CellTiter-Glo reagent)(Promega에서 구입), 또는 아라마르 블루 시약(alamarBlue reagent)(ThermoFisher Scientific에서 구입)과 마이크로플레이트 리더(microplate reader)(Molecular Devices에서 구입)를 이용하여 측정하였다. In order to evaluate the toxicity of exosomes obtained according to the isolation method of one embodiment of the present invention in human skin fibroblast HS68 cells, exosomes were treated by concentration to the cells and the proliferation rate of the cells was confirmed. HS68 cells were suspended in DMEM containing 10% FBS, and the cells were mixed with 80 ~ 90% confluency and cultured in a 5% CO 2 incubator at 37 ° C for 24 hours. After 24 hours, the culture solution was removed, and the cell survival rate was evaluated by culturing the exosome prepared in Example 2 for each concentration and culturing for 24 to 72 hours. Cell viability was measured using WST-1 reagent (purchased from Takara), MTT reagent (purchased from Sigma), CellTiter-Glo reagent (purchased from Promega), or Aramar Blue reagent alamarBlue reagent (purchased from ThermoFisher Scientific) and a microplate reader (purchased from Molecular Devices).
비교군은 엑소좀이 처리되지 않은 일반 세포배양배지에서 배양된 세포수를 기준으로 하였고, 시험된 농도 범위 내에서 줄기세포 유래의 엑소좀에 의한 세포 독성이 나타나지 않음을 확인하였다(도 2).The comparison group was based on the number of cells cultured in the normal cell culture medium not treated with exosome, and it was confirmed that the cytosolic effect of exosome derived from stem cells did not appear within the tested concentration range (FIG. 2).
실시예 5: 마크로파지 세포주를 이용한 염증 반응 측정Example 5 Measurement of Inflammatory Response Using Macrophage Cell Lines
RAW 264.7 세포를 10% FBS를 포함한 DMEM 배지에 현탁시키고 이를 멀티웰 플레이트(multiwell plate)의 각 웰에 80 내지 90%의 밀집도(confluency)를 갖도록 분주하였다. 다음 날 LPS가 포함된 새로운 무혈청 배지에 희석한 줄기세포 유래의 엑소좀 (실시예 2에서 준비된 엑소좀으로서 본 발명의 엑소좀 키트에 사용됨)을 적정 농도로 1~24시간 동안 처리하여 배양하였다. 배양이 끝난 배양상층액을 취하고 배양액 내에 존재하는 NO 및 염증성 사이토카인을 측정하여 염증 반응을 확인하였다. 배양액 내의 염증 반응은 NO 검출키트(detection kit)(인트론바이오 혹은 프로메가에서 구입)를 이용하여 측정하였다. ELISA 키트 (R&D system에서 구입) 제조사의 매뉴얼대로 수행하여 LPS만을 처리한 군과 줄기세포 유래의 엑소좀이 함께 처리된 군의 염증성 사이토카인 TNF-α 양을 확인하였다. 양성대조군으로 덱사메타손(dexamethasone) (Sigma에서 구입)을 처리하였다. 또한, 위와 같이 처리된 RAW 264.7 세포로부터 얻은 총 RNA로부터 cDNA를 제조하였고 리얼타임 PCR 방법을 이용하여 iNOS, TNF-α, IL-6 및 IL-1β의 mRNA 변화량을 측정하였다. 상기 유전자들을 정량하기 위한 표준 유전자로서 GAPDH 유전자를 사용하였다. 리얼타임 PCR에 사용한 프라이머의 종류와 서열은 하기의 표 1과 같다.RAW 264.7 cells were suspended in DMEM medium containing 10% FBS and dispensed into each well of a multiwell plate to have a confluency of 80-90%. On the next day, the stem cell-derived exosomes (used as the exosome prepared in Example 2 as used in the exosome kit of the present invention) diluted in a new serum-free medium containing LPS were treated for 1 to 24 hours at an appropriate concentration and cultured . The cultured supernatant was taken and NO and inflammatory cytokines present in the culture were measured to confirm the inflammatory response. The inflammatory response in the culture medium was measured using a NO detection kit (purchased from Intron Bio or Promega). An ELISA kit (purchased from the R & D system) was performed according to the manufacturer's manual to confirm the amount of TNF-α, an inflammatory cytokine, in the group treated with LPS alone and the group treated with stem cell-derived exosomes. Dexamethasone (purchased from Sigma) was treated as a positive control. In addition, cDNA was prepared from the total RNA obtained from the RAW 264.7 cells treated as described above, and the amount of mRNA change of iNOS, TNF-α, IL-6 and IL-1β was measured using a real-time PCR method. The GAPDH gene was used as a standard gene for quantifying the above genes. The types and sequences of the primers used in the real-time PCR are shown in Table 1 below.
리얼타임 PCR에 사용된 프라이머 종류 및 염기서열Primer types and base sequences used in real-time PCR
유전자gene 서열order
정방향 프라이머 (5' → 3')The forward primer (5 '- > 3') 역방향 프라이머 (5' → 3')The reverse primer (5 '- > 3')
TNF-αTNF-a TCT CAT CAG TTC TAT GGC CCA GAC (서열번호 1)TCT CAT CAG TTC TAT GGC CCA GAC (SEQ ID NO: 1) GGC ACC ACT AGT TGG TTG TCT TTG (서열번호 2)GGC ACC ACT AGT TGG TTG TCT TTG (SEQ ID NO: 2)
iNOSiNOS GCT ACC ACA TTG AAG AAG CTG GTG (서열번호 3)GCT ACC ACA TTG AAG AAG CTG GTG (SEQ ID NO: 3) CCA TAG GAA AAG ACT GCA CCG AAG (서열번호 4)CCA TAG GAA AAG ACT GCA CCG AAG (SEQ ID NO: 4)
GAPDHGAPDH GAC ATC AAG AAG GTG GTG AAG CAG (서열번호 5)GAC ATC AAG AAG GTG GTG AAG CAG (SEQ ID NO: 5) CCC TGT TGC TGT AGC CGT ATT CAT (서열번호 6)CCC TGT TGC TGT AGC CGT ATT CAT (SEQ ID NO: 6)
IL-6IL-6 GCC AGA GTC CTT CAG AGA GAT ACA (서열번호 7)GCC AGA GTC CTT CAG AGA GAT ACA (SEQ ID NO: 7) ATT GGA TGG TCT TGG TCC TTA GCC (서열번호 8)ATT GGA TGG TCT TGG TCC TTA GCC (SEQ ID NO: 8)
IL-1βIL-1? GCA ACG ACA AAA TAC CTG TGG CCT (서열번호 9)GCA ACG ACA AAA TAC CTG TGG CCT (SEQ ID NO: 9) AGT TGG GGA ACT CTG CAG ACT CAA (서열번호 10)AGT TGG GGA ACT CTG CAG ACT CAA (SEQ ID NO: 10)
우선, 도 3에 도시된 바와 같이, 생쥐의 대식세포인 RAW 264.7 세포에 대해 LPS와 함께 본 발명의 엑소좀 키트에 사용되는 엑소좀을 처리한 경우 LPS에 의해 유도되는 염증성 사이토카인인 TNF-α, IL-6 및 IL-1β의 mRNA 발현량이 감소하였고, NO 생성효소인 iNOS의 mRNA 발현량이 감소하였다.First, as shown in FIG. 3, when RAW 264.7 cells, which are macrophages of mice, were treated with exosomes used in the exosomic kit of the present invention together with LPS, TNF-a, an inflammatory cytokine induced by LPS , IL-6 and IL-1β decreased and mRNA expression of iNOS, a NO-producing enzyme, decreased.
다음으로, 도 4에 도시된 바와 같이, 생쥐의 대식세포인 RAW 264.7 세포에 LPS 존재 하에서 본 발명의 엑소좀 키트에 사용되는 엑소좀을 처리한 결과 LPS에 의해 유도되는 염증 반응인 NO 생성을 농도의존적으로 감소시킴을 확인하였다. 또한, 도 5에 도시된 바와 같이, 생쥐의 대식세포인 RAW 264.7 세포에 LPS 존재 하에서 본 발명의 엑소좀 키트에 사용되는 엑소좀을 처리한 결과 LPS에 의해 유도되는 염증성 사이토카인인 TNF-α 생성을 감소시킴을 확인하였다. 이러한 결과들은 본 발명의 엑소좀 키트에 사용되는 엑소좀이 피부염의 예방, 억제, 개선, 완화 또는 치료를 위해 유용한 기능적 활성, 즉 LPS에 의해 유도된 염증 반응을 감소시키는 활성을 가지며, 본 발명의 엑소좀 키트는 피부염의 예방, 억제, 개선, 완화 또는 치료를 위해 유용하게 사용될 수 있는 것임을 강력히 시사한다. Next, as shown in FIG. 4, RAW 264.7 cells, which are macrophages of mice, were treated with exosomes used in the exosomal kit of the present invention in the presence of LPS, and as a result, NO production, which is an inflammatory reaction induced by LPS, Dependent manner. As shown in FIG. 5, RAW 264.7 cells, macrophages of mice, were treated with exosomes used in the exosomal kit of the present invention in the presence of LPS. As a result, LPS-induced inflammatory cytokine TNF-a production . These results indicate that the exosome used in the exosomal kit of the present invention has an activity of reducing the inflammatory reaction induced by LPS, which is useful for preventing, inhibiting, improving, alleviating or treating dermatitis, It strongly suggests that exosome kits may be useful for the prevention, inhibition, amelioration, palliation or treatment of dermatitis.
실시예 6: THP-1 단핵구를 이용한 항염증 효과 확인Example 6: Determination of anti-inflammatory effect using THP-1 monocyte
THP-1 단핵구(THP-1 human monocyte)를 대식세포로 분화시킨 후 본 발명의 엑소좀 키트에 사용되는 엑소좀의 항염증 효과를 다음과 같이 확인하였다. After the THP-1 monocyte (THP-1 human monocyte) was differentiated into macrophages, the anti-inflammatory effect of exosomes used in the exosome kit of the present invention was confirmed as follows.
THP-1 단핵구를, 10% FBS(Fetal Bovine Serum), 1% 페니실린-스트렙토마이신 및 100nM 포르볼 12-미리스테이트 13-아세테이트(Phorbol 12-myristate 13-acetate)(Sigma에서 구입)가 첨가된 RPMI 1640(Roswell Park Memorial Institute 1640; ThermoFisher Scientific에서 구입) 배지에 현탁시킨 후 12웰 플레이트에 6×105 세포/mL의 밀도로 접종하고 37℃, 5% CO2 배양기에서 48시간 배양하여 THP-1 유래 대식세포(THP-1 derived macrophage)로 분화를 유도하였다. 이후 분화된 세포를 PBS로 1회 세척하고, 포르볼 12-미리스테이트 13-아세테이트가 포함되어 있지 않은 RPMI 1640 배지에서 24시간 동안 배양하여 세포를 안정화하였다. THP-1 monocytes were cultured in RPMI supplemented with 10% FBS (Fetal Bovine Serum), 1% penicillin-streptomycin and 100 nM Phorbol 12-myristate 13-acetate (purchased from Sigma) 1640 (purchased from ThermoFisher Scientific) medium at 1640 (purchased from ThermoFisher Scientific), and then seeded in a 12-well plate at a density of 6 × 10 5 cells / mL and cultured in a 5% CO 2 incubator at 37 ° C. for 48 hours to obtain THP-1 Derived macrophage (THP-1 derived macrophage). The differentiated cells were then washed once with PBS and cultured in RPMI 1640 medium without formalin 12-myristate 13-acetate for 24 hours to stabilize the cells.
동시처리(co-treatment) 실험군의 경우, THP-1 유래 대식세포에 대해 FBS(Fetal Bovine Serum)가 포함되어 있지 않은 RPMI 1640 배지와 100 ng/mL LPS(sigma 에서 구입) 및 20 ng/mL IFN-γ(Peprotech에서 구입)를 처리하여 THP-1 유래 대식세포를 활성화시키고, 또한 양성대조군인 덱사메타손(Sigma에서 구입) 또는 본 발명의 엑소좀 키트에 사용되는 엑소좀(실시예 2에서 준비된 엑소좀)을 처리하여 24시간 배양하였다. 한편, 전처리(pre-treatment) 실험군의 경우, THP-1 유래 대식세포에 대해 FBS(Fetal Bovine Serum)가 포함되어 있지 않은 RPMI 1640 배지와 양성대조군인 덱사메타손(Sigma에서 구입) 또는 본 발명의 엑소좀 키트에 사용되는 엑소좀(실시예 2에서 준비된 엑소좀)을 처리하여 24시간 배양한 다음, 100 ng/mL LPS(sigma 에서 구입) 및 20 ng/mL IFN-γ(Peprotech에서 구입)를 처리하고 4시간 동안 배양하여 THP-1 유래 대식세포를 활성화하였다. For the co-treatment group, RPMI 1640 medium without FBS (Fetal Bovine Serum), 100 ng / mL LPS (purchased from Sigma) and 20 ng / mL IFN -γ (purchased from Peprotech) was activated to activate THP-1 derived macrophages and the exosome used in the positive control group dexamethasone (purchased from Sigma) or the exosome kit of the present invention (exosome prepared in Example 2) ) And cultured for 24 hours. On the other hand, in the case of the pre-treatment experimental group, RPMI 1640 medium containing no FBS (Fetal Bovine Serum) for the THP-1 derived macrophages and dexamethasone (purchased from Sigma) The exosomes used in the kit (the exosomes prepared in Example 2) were cultured and cultured for 24 hours and then treated with 100 ng / mL LPS (purchased from Sigma) and 20 ng / mL IFN-y (purchased from Peprotech) And incubated for 4 hours to activate THP-1-derived macrophages.
배양이 끝난 THP-1 유래 대식세포(THP-1 derived macrophage)로부터 분리한 RNA로부터 cDNA를 제조하고 리얼타임 PCR 방법을 이용하여 친염증성 사이토카인(Pro-inflammatory Cytokine)인 IL-6, IL-1β 및 TNF-α의 mRNA 발현량을 측정하였다. 상기 유전자들을 정량하기 위한 표준 유전자로서 GAPDH를 사용하였다. PCR에 사용한 프라이머의 종류와 서열은 하기의 표 2와 같다.CDNA was prepared from the RNA isolated from cultured THP-1 derived macrophages, and cDNA was prepared from the pro-inflammatory cytokines IL-6, IL-1β And TNF-alpha mRNA expression levels were measured. GAPDH was used as a standard gene for quantifying the genes. The types and sequences of the primers used in the PCR are shown in Table 2 below.
리얼타임 PCR에 사용된 프라이머 종류 및 염기서열Primer types and base sequences used in real-time PCR
유전자gene 서열order
정방향 프라이머 (5'→3')The forward primer (5 '- > 3') 역방향 프라이머 (5'→3')The reverse primer (5 '- > 3')
IL-6IL-6 AAGCCAGAGCTGTGCAGATGAGTA (서열번호 11)AAGCCAGAGCTGTGCAGATGAGTA (SEQ ID NO: 11) TGTCCTGCAGCCACTGGTTC (서열번호 12)TGTCCTGCAGCCACTGGTTC (SEQ ID NO: 12)
IL-1βIL-1? ACCTGTCCTGCGTGTTGAAAGATG (서열번호 13)ACCTGTCCTGCGTGTTGAAAGATG (SEQ ID NO: 13) TGGGCAGACTCAAATTCCAGCTTG (서열번호 14)TGGGCAGACTCAAATTCCAGCTTG (SEQ ID NO: 14)
TNF-αTNF-a CTGCCTGCTGCACTTTGGAG (서열번호 15)CTGCCTGCTGCACTTTGGAG (SEQ ID NO: 15) ACATGGGCTACAGGCTTGTCACT (서열번호 16)ACATGGGCTACAGGCTTGTCACT (SEQ ID NO: 16)
GAPDHGAPDH CTTTGGTATCGTGGAAGGACTC (서열번호 17)CTTTGGTATCGTGGAAGGACTC (SEQ ID NO: 17) GTAGAGGCAGGGATGATGTTCT (서열번호 18)GTAGAGGCAGGGATGATGTTCT (SEQ ID NO: 18)
도 6에 도시된 바와 같이, THP-1 단핵구를 대식세포로 분화시킨 후 얻은 THP-1 유래 대식세포에 대해, LPS 및 IFN-γ와 함께 본 발명의 엑소좀 키트에 사용되는 엑소좀을 처리(co-treatment)한 경우, 음성대조군에 비해 친염증성 사이토카인인 IL-6의 mRNA 발현량이 감소하였다. 또한, 도 7 내지 도 9에 도시된 바와 같이, THP-1 단핵구를 대식세포로 분화시킨 후 얻은 THP-1 유래 대식세포에 대해, LPS 및 IFN-γ 처리 전에 본 발명의 엑소좀 키트에 사용되는 엑소좀을 전처리(pre-treatment)한 경우, 음성대조군에 비해 친염증성 사이토카인인 IL-6, IL-1β 및 TNF-α의 mRNA 발현량이 감소하였다. 친염증성 사이토카인인 IL-6, IL-1β 및 TNF-α는 친염증성 M1 대식세포에서는 발현이 증가하고 항염증성 M2 대식세포에서는 발현이 감소한다. As shown in FIG. 6, for the THP-1 derived macrophages obtained after differentiating THP-1 monocytes into macrophages, the exosomes used in the exosomal kit of the present invention were treated with LPS and IFN- co-treatment), mRNA expression of IL-6, a proinflammatory cytokine, was decreased compared to negative control. In addition, as shown in FIGS. 7 to 9, THP-1 derived macrophages obtained after differentiating THP-1 monocytes into macrophages were used in the exosome kit of the present invention before LPS and IFN- IL-6, IL-1β and TNF-α mRNA expression levels were decreased in the pre-treatment of exosomes compared to negative control. The proinflammatory cytokines IL-6, IL-1 [beta] and TNF- [alpha] increase expression in proinflammatory M1 macrophages and decrease expression in anti-inflammatory M2 macrophages.
따라서, 상기 결과들은 본 발명의 엑소좀 키트에 사용되는 엑소좀이 염증반응 및 이로 인한 피부염 증상을 억제, 완화 및 감소시킬 수 있는 것을 시사하며, 또한 본 발명의 엑소좀 키트가 피부염의 예방, 억제, 개선, 완화 또는 치료를 위해 유용하게 사용될 수 있는 것임을 강력히 뒷받침하는 결과라고 할 수 있다.Therefore, the above results suggest that the exosome used in the exosome kit of the present invention can inhibit, alleviate and reduce the inflammatory reaction and the dermatitis caused thereby, and the exosome kit of the present invention can prevent, , Which may be useful for improving, alleviating, or treating a disease.
실시예 7: 본 발명의 엑소좀 키트에 사용되는 엑소좀의 피부 투과 능력 시험Example 7: Skin permeability test of exosome used in the exosome kit of the present invention
형광염색된 엑소좀을 제조하기 위하여 PKH67 염료(Sigma에서 구입)를 사용하였다. 1 mM의 PKH67을 Diluent C (Sigma에서 구입)에 희석하여 10 μM의 PKH67 용액을 제조한 후, 적정 농도의 엑소좀 용액과 혼합하여 상온에서 빛을 차단한 상태로 10분간 반응시켰다. 반응이 끝난 후, PKH67로 염색된 엑소좀(이하, "PKH-엑소좀"으로 약칭)으로부터 잔여 PKH67 염료를 제거하기 위하여 MW3000 스핀 컬럼(ThermoFisher Scientific에서 구입)을 사용하였다. 형광측정기(Molecular Devices에서 구입)를 이용하여 확인한 결과 엑소좀과 반응하지 않은 PKH67을 제거한 후, PKH-엑소좀에서 충분한 강도의 형광이 검출됨을 확인하였다(도 10). PKH67 dye (purchased from Sigma) was used to prepare fluorescently stained exosomes. 1 mM PKH67 was diluted in Diluent C (purchased from Sigma) to prepare a 10 μM PKH67 solution, mixed with an appropriate concentration of exosome solution, and allowed to react at room temperature for 10 minutes in the absence of light. After the reaction, an MW3000 spin column (purchased from ThermoFisher Scientific) was used to remove the remaining PKH67 dye from the exosomes stained with PKH67 (hereinafter abbreviated as "PKH-exosomes"). After confirming the use of a fluorescence meter (purchased from Molecular Devices), PKH67 not reacted with exosomes was removed, and fluorescence of sufficient intensity was detected in PKH-exosomes (FIG. 10).
PKH-엑소좀을 적정 농도, 예를 들어 1×105 입자/mL 내지 1×109 입자/mL 농도로 인산완충용액(PBS)에 분산시킨 후, 돼지 피부 바깥면에 도포하였다. 도포된 PKH-엑소좀의 건조를 막기 위하여 부직포를 덮어 준 후 적정 시간, 예를 들어 30분 내지 1시간 동안 반응시켜 PKH-엑소좀이 피하 조직에 전달되도록 하였다. 또한, PKH-엑소좀을 돼지 피부 바깥면에 도포하고 부직포를 덮어 준 후, 일정 시간, 예를 들어 30분 내지 1시간 동안 미세 전류를 흘려주었다. 반응이 종료된 후, 돼지 피부조직을 3.7% 포름알데히드 용액에 담궈 하룻밤 동안 반응시키고, 인산완충용액으로 5분씩 3회 세척하였다. 세척된 돼지 피부조직을 30% 수크로오스 용액에 담가 처리한 후, OCT 화합물을 처리하였다. 다시 인산완충용액으로 5분씩 3회 세척한 후, 마이크로톰을 이용하여 종단면으로 절편을 제작하여 슬라이드글라스 위에 조직 절편을 위치시켰다. 한편 조직 절편의 제작은 포름알데히드 용액으로 조직을 고정하기 전에 진행할 수도 있다. 조직 절편에서 PKH-엑소좀으로부터 검출되는 형광은 형광현미경을 이용하여 관찰하였다. 이상의 방법으로 돼지 피부조직의 표피를 투과하여 피하 조직으로 전달된 PKH-엑소좀을 확인하였다. 그 결과, 미세 전류를 흘려주는 경우(active transdermal delivery), 엑소좀이 표피를 보다 효과적으로 통과하여 피부조직 내부로 깊숙이 전달되었고, 피부에 대해 보다 효과적으로 흡수되는 것을 확인할 수 있었다(도 11 참조). 도 11에서 확인되는 바와 같이, 본 발명의 엑소좀 키트에 사용되는 엑소좀은 표피를 효과적으로 통과하여 피부조직 내부로 깊숙이 전달될 수 있고, 피부에 대해 효과적으로 흡수될 수 있다. 따라서, 본 발명의 엑소좀 키트는 피부염의 예방, 억제, 개선, 완화 또는 치료 등에 있어서 유용하게 사용될 수 있을 것으로 기대된다. PKH-exosomes were dispersed in phosphate buffer (PBS) at a proper concentration, for example, 1 × 10 5 to 1 × 10 9 particles / mL, and then applied to the outer surface of the pig skin. To prevent drying of the applied PKH-exosomes, the nonwoven fabric was covered and allowed to react for an appropriate period of time, for example, 30 minutes to 1 hour to allow the PKH-exosomes to be delivered to the subcutaneous tissues. In addition, the PKH-exosomes were applied to the outer surface of the pig skin and covered with the nonwoven fabric, followed by flowing a microcurrent for a predetermined time, for example, 30 minutes to 1 hour. After the reaction was completed, the pig skin tissue was immersed in a 3.7% formaldehyde solution, reacted overnight, and washed three times for 5 minutes each with phosphate buffered saline. The washed pig skin tissue was immersed in a 30% sucrose solution and treated with OCT compound. After washing three times for 5 minutes with phosphate buffer solution, sections were prepared on a longitudinal section using a microtome, and tissue sections were placed on a slide glass. On the other hand, the preparation of the tissue section may be carried out before the tissue is fixed with formaldehyde solution. Fluorescence detected from PKH-exosomes in tissue sections was observed using fluorescence microscopy. In this way, PKH-exosomes transmitted to the subcutaneous tissue through the epidermis of the pig skin tissue were identified. As a result, it was confirmed that the exosomes were more effectively passed through the epidermis and deeply transferred into the skin tissue and more effectively absorbed to the skin when active current was delivered (active transdermal delivery) (see FIG. 11). As shown in Fig. 11, the exosomes used in the exosome kit of the present invention can be effectively passed through the epidermis and deeply transferred into the skin tissue, and can be effectively absorbed to the skin. Therefore, the exosomic kit of the present invention is expected to be usefully used for preventing, suppressing, improving, alleviating or treating dermatitis.
다음으로, 헤어리스 마우스의 피부조직을 적출하여 프란츠 확산셀(Franz Diffusion Cell)의 챔버 위쪽에 위치시켰다. 이때, 확산셀의 내부는 인산완충용액으로 채워주었다. PKH-엑소좀을 적정 농도, 예를 들어 1×105 입자/mL 내지 1×109 입자/mL 농도로 인산완충용액(PBS)에 분산시킨 후, 생쥐의 피부조직 바깥쪽에 도포하였다. 이때, PKH-엑소좀의 건조를 막기 위하여 부직포를 생쥐 피부조직 바깥쪽에 미리 위치시키고, 부직포와 피부조직 사이에 PKH-엑소좀을 주입하였다. 그 후 30분 내지 1시간 동안 반응시켰다. 또한, PKH-엑소좀을 부직포와 피부조직 사이에 주입한 후, 일정 시간, 예를 들어 30분 내지 1시간 동안 미세 전류를 흘려주었다. 반응이 끝난 후, 즉시 공초점형광현미경(Leica, SP8X)을 이용하여 피부조직 내부로 전달된 PKH-엑소좀을 확인하거나, 1시간 내지 6시간 동안 피부조직과 PKH-엑소좀 용액을 추가로 반응시킨 후, 공초점형광현미경으로 PKH-엑소좀을 확인하였다. 그 결과, 미세 전류를 흘려주는 경우(active transdermal delivery), 엑소좀이 표피를 보다 효과적으로 통과하여 피부조직 내부로 깊숙이 전달되었고, 피부에 대해 보다 효과적으로 흡수되는 것을 확인할 수 있었다(도 12 및 도 13 참조). Next, the skin tissue of the hairless mouse was removed and placed on top of the chamber of the Franz Diffusion Cell. At this time, the inside of the diffusion cell was filled with a phosphate buffer solution. PKH-exosomes were dispersed in phosphate buffer (PBS) at a proper concentration, for example, 1 × 10 5 to 1 × 10 9 particles / mL, and then applied to the outside of the skin tissue of the mice. At this time, in order to prevent drying of PKH-exosomes, the nonwoven fabric was pre-positioned outside the mouse skin tissue, and PKH-exosomes were injected between the nonwoven fabric and skin tissue. Then, the reaction was carried out for 30 minutes to 1 hour. In addition, the PKH-exosomes were injected between the nonwoven fabric and the skin tissue, and the microcurrent was flowed for a predetermined time, for example, 30 minutes to 1 hour. After completion of the reaction, the PKH-exosomes transferred into the skin tissue were confirmed immediately using a confocal fluorescence microscope (Leica, SP8X) or the skin tissue and the PKH-exosomal solution were further reacted for 1 hour to 6 hours , And PKH-exosomes were identified by confocal fluorescence microscopy. As a result, it was confirmed that the exosomes were more efficiently passed through the epidermis, deeply transferred into the skin tissue, and more effectively absorbed to the skin when active current was delivered (active transdermal delivery) (see Figs. 12 and 13 ).
이상의 실험결과들을 종합하면, 본 발명의 엑소좀 키트를 피부에 적용할 경우, 엑소좀이 표피를 효과적으로 통과하여 피부염의 예방, 억제, 완화, 개선 또는 치료 등에 있어서 효과적으로 작용할 것으로 기대된다. It is expected that the exosome kits of the present invention can effectively pass through the epidermis to effectively prevent the dermatitis from being prevented, inhibited, alleviated, improved or treated when the exosome kit of the present invention is applied to the skin.
실시예 8: 사람 피부에 대한 엑소좀 키트의 처치Example 8: Treatment of exosome kit against human skin
도 16에 도시된 바와 같이, 본 발명의 일 구체예에 따른 엑소좀 키트(100)는 액상 침적 시트 마스크(10)(줄기세포 유래의 엑소좀을 함유하는 조성물이 도포되거나 침적된 흰색 시트 마스크)와 이온토포레시스 디바이스인 경피투과 촉진 시트 마스크(은색 시트 마스크)(20)로 구성되어 있다. 액상 침적 시트 마스크(10)의 표면 내지는 내부에는 줄기세포 유래의 엑소좀(12)이 도포되거나 침적되어 있다. 또한, 경피투과 촉진 시트 마스크(20)에는 미세전류를 발생시키는 전지(22)가 장착되어 있고, 이러한 전지(22)와 전기적으로 연결되어 있는 도전성 패턴(24)이 경피투과 촉진 시트 마스크(20) 상에 형성되어 전지(22)로부터 발생된 미세전류를 경피투과 촉진 시트 마스크(20) 전반에 골고루 전달할 수 있다. 도전성 패턴(24)은 금, 은, 또는 구리 등의 도전성 금속이 프린팅되거나 도금(plating)되어 형성된다. 16, the exosome kit 100 according to one embodiment of the present invention includes a liquid immersion sheet mask 10 (a white sheet mask on which a composition containing a stem cell-derived exosome is applied or immersed) And a transdermal permeation promoting sheet mask (silver sheet mask) 20 which is an iontophoresis device. A stem cell-derived exosome (12) is applied or deposited on the surface or inside of the liquid immersion sheet mask (10). A battery 22 for generating a microcurrent is mounted on the percutaneous permeation enhancing sheet mask 20 and a conductive pattern 24 electrically connected to the cell 22 is attached to the percutaneous permeation enhancing sheet mask 20, So that the micro current generated from the battery 22 can be uniformly transmitted to the entirety of the percutaneous permeation enhancing sheet mask 20. [ The conductive pattern 24 is formed by printing or plating a conductive metal such as gold, silver, or copper.
상기와 같은 구성을 갖는 "엑소좀 키트(100)"를 피부염 증상과 관련이 있는 홍조 증상과 피부 트러블이 심한 사람(이하, "case 1"이라 함)의 얼굴에 1회 적용하여 피부염으로 인한 피부 트러블과 홍조 증상이 완화 내지는 개선되는지를 평가하였다. 또한, 본 발명의 엑소좀 키트(100)를 피부염 증상과 관련이 있는 홍조 증상이 있는 사람(이하, "case 2"라 함)의 얼굴에 1회 적용하여 전체적인 피부톤과 홍조 증상이 개선되는지를 평가하였다. 5×105 입자의 엑소좀(12)이 포함된 액상 침적 시트 마스크(10)를 눈과 입 부분에 맞추어 case 1 및 2의 얼굴 전체에 고르게 밀착시킨 다음, 약 0.3~0.4 mA의 미세 전류가 흐르도록 액티베이션된 경피투과 촉진 시트 마스크(20)를 액상 침적 시트 마스크(10) 위에 적층시켜 25~30분 정도 사용하였다. The "exosome kit (100)" having the above-described structure is applied once to the face of a person having a flushing symptom and skin troubles (hereinafter referred to as "case 1") related to the dermatitis symptom, It was evaluated whether the symptom of trouble and redness was alleviated or improved. In addition, the exosome kit 100 of the present invention is applied once to the face of a person having flushing symptoms (hereinafter referred to as " case 2 ") associated with dermatitis symptoms to evaluate whether the overall skin tone and flushing symptoms are improved Respectively. A liquid immersion sheet mask 10 containing 5 × 10 5 exosomes 12 was uniformly adhered to the entire faces of the cases 1 and 2 by aligning them with the eyes and mouth parts and then a microcurrent of about 0.3 to 0.4 mA An activated percutaneous permeation enhancing sheet mask 20 which was activated to flow was laminated on the liquid immersion sheet mask 10 and used for about 25 to 30 minutes.
그 결과, 줄기세포 유래의 엑소좀(12)을 5×105 입자/마스크의 농도로 함유하는 액상 침적 시트 마스크(10)와 경피투과 촉진 시트 마스크(20)로 구성된 "엑소좀 키트(100)"의 한번의 사용만으로 "case 1"에서 홍조 및 피부 트러블이 현저히 개선되었고(도 14의 case 1 참조), "case 2"에서도 전체적인 피부톤이 개선되었으며 홍조가 완화되었다(도 14의 case 2 참조). 따라서, 줄기세포 유래의 엑소좀을 유효성분으로 함유하는 조성물이 침적된 액상 침적 시트 마스크(10)와 경피투과 촉진 시트 마스크(이온토포레시스 디바이스)(20)로 구성된 본 발명의 엑소좀 키트(100)는 피부염 증상과 이와 관련이 있는 홍조 증상 및 피부트러블을 예방, 억제, 완화 내지는 개선시키는 효과가 있다고 할 수 있다. As a result, the "exosome kit 100" composed of the liquid-immersion sheet mask 10 containing the stem cell-derived exosome 12 at a concentration of 5 × 10 5 particles / mask and the percutaneous permeation- (See case 1 in FIG. 14), the overall skin tone was improved in "case 2" and the flushing was alleviated (see case 2 in FIG. 14) . Therefore, the exosomal kit (present invention) comprising the liquid immersion sheet mask 10 in which the composition containing the stem cell-derived exosome as an active ingredient is immersed and the percutaneous permeation promoting sheet mask (iontophoresis device) 100) can be said to have an effect of preventing, suppressing, alleviating or alleviating dermatitis symptoms and redness symptoms and skin troubles associated therewith.
또한, 줄기세포 유래의 엑소좀을 0.29×108 입자/mL의 농도로 함유하는 조성물(엑소좀 현탁액)을, 가려움증을 호소하는 중증 아토피 환자 3인의 환부(손, 목, 팔 등)에 도포한 후 이온토포레시스 장비(IONZYME DF MACHINE)(Environ에서 구입)를 사용하여 상기 조성물이 도포된 환부에 20분 동안 0.4 mA의 미세전류를 흘려주는 이온토포레시스를 수행하였다. 상기와 같은 처치를 2주 동안 주3회에 걸쳐 시행한 결과, 환자들의 극심한 가려움증이 현저히 완화되었고, 홍반 증상 역시 현저히 개선되었다(도 15). 줄기세포 유래의 엑소좀을 함유하는 조성물을 적용한 환자들 모두 극심한 가려움증과 홍반이 완화되어 스테로이드나 항히스타민 제제의 처방을 중단해도 될 정도로 증상이 개선되었다. In addition, a composition (exosome suspension) containing a stem cell-derived exosome at a concentration of 0.29 x 10 8 particles / mL was applied to the affected part (hand, neck, arm, etc.) of three severe atopic patients complaining of itching Iontophoresis was carried out by using a iontophoresis device (IONZYME DF MACHINE) (purchased from Environ) to flow a micro current of 0.4 mA for 20 minutes to the affected part of the composition. The above procedure was repeated three times a week for two weeks, and the severe itching of the patients was remarkably alleviated, and the erythema symptoms were also markedly improved (FIG. 15). Patients who applied compositions containing stem cell-derived exosomes improved their symptoms to such an extent that severe itching and erythema could be relieved to stop the prescription of steroid or antihistamine preparations.
따라서, 본 발명의 엑소좀 키트(100)는 상기와 같은 임상시험을 통해 확인되는 바와 같이 피부염 및 이와 관련된 증상의 예방, 억제, 개선, 완화 또는 치료에 유용하게 사용될 수 있음을 알 수 있다.Accordingly, it can be seen that the exosome kit 100 of the present invention can be effectively used for preventing, suppressing, ameliorating, alleviating or treating dermatitis and symptoms associated therewith, as confirmed by the clinical test as described above.
이상, 본 발명을 상기 실시예를 들어 설명하였으나, 본 발명은 이에 제한되는 것이 아니다. 당업자라면 본 발명의 취지 및 범위를 벗어나지 않고 수정, 변경을 할 수 있으며 이러한 수정과 변경 또한 본 발명에 속하는 것임을 알 수 있을 것이다.Although the present invention has been described with reference to the above embodiments, the present invention is not limited thereto. It will be understood by those skilled in the art that modifications and variations may be made without departing from the spirit and scope of the invention, and that such modifications and variations are also contemplated by the present invention.

Claims (7)

  1. 줄기세포 유래의 엑소좀을 유효성분으로 포함하는 조성물이 도포되거나 침적된 제1 마스크팩, 마스크시트 또는 패취와, A first mask pack, a mask sheet or a patch which is applied or deposited with a composition containing an exosome derived from a stem cell as an active ingredient,
    상기 제1 마스크팩, 마스크시트 또는 패취가 적용된 포유동물의 피부에 미세전류를 흘려 주는 이온토포레시스 디바이스를 포함하는, 피부염의 예방, 억제, 완화, 개선 또는 치료용 엑소좀 키트.And an iontophoresis device for applying a microcurrent to the skin of the mammal to which the first mask pack, the mask sheet or the patch is applied, for preventing, suppressing, alleviating, improving or treating dermatitis.
  2. 제1항에 있어서,The method according to claim 1,
    상기 제1 마스크팩, 마스크시트 또는 패취를 포유동물의 피부 위에 접촉 또는 부착시키고, 상기 제1 마스크팩, 마스크시트 또는 패취 위에 상기 이온토포레시스 디바이스를 위치시킴으로써, 상기 제1 마스크팩, 마스크시트 또는 패취가 접촉 또는 부착되어 상기 조성물이 적용된 포유동물의 피부에 미세전류를 흘려 주는 것을 특징으로 하는, 엑소좀 키트.Wherein the first mask pack, the mask sheet or the patch is contacted or adhered onto the skin of the mammal, and the iontophoresis device is placed on the first mask pack, the mask sheet or the patch, Or patches are contacted or adhered to flow micro-currents into the skin of the mammal to which said composition is applied.
  3. 제1항 또는 제2항에 있어서,3. The method according to claim 1 or 2,
    상기 이온토포레시스 디바이스는 가요성 배터리, 리튬이온 이차 전지, 알칼리 전지, 건전지, 수은 전지, 리튬 전지, 니켈-카드뮴 전지, 및 역전기 투석 전지로 구성된 군으로부터 선택된 적어도 1종의 전지를 포함하거나, 상기 적어도 1종의 전지가 장착된 제2 마스크팩, 마스크시트 또는 패취인, 엑소좀 키트.The iontophoresis device includes at least one battery selected from the group consisting of a flexible battery, a lithium ion secondary battery, an alkaline battery, a dry battery, a mercury battery, a lithium battery, a nickel-cadmium battery, A second mask pack, a mask sheet or a patch, on which the at least one battery is mounted.
  4. 제3항에 있어서,The method of claim 3,
    상기 제1 마스크팩, 마스크시트 또는 패취 위에 상기 제2 마스크팩, 마스크시트 또는 패취가 적층되어 사용되는 것을 특징으로 하는, 엑소좀 키트.Wherein the second mask pack, the mask sheet or the patch is laminated on the first mask pack, the mask sheet or the patch.
  5. (a) 줄기세포 유래의 엑소좀을 유효성분으로 포함하는 조성물이 도포되거나 침적된 제1 마스크팩, 마스크시트 또는 패취를 포유동물의 피부 위에 접촉 또는 부착하는 단계와, (a) contacting or adhering a first mask pack, a mask sheet or a patch applied or immersed with a composition comprising an exosome derived from a stem cell as an active ingredient onto the skin of a mammal,
    (b) 상기 제1 마스크팩, 마스크시트 또는 패취 위에 이온토포레시스 디바이스를 위치시키는 단계와, (b) positioning the iontophoresis device on the first mask pack, mask sheet or patch,
    (c) 상기 제1 마스크팩, 마스크시트 또는 패취가 접촉 또는 부착되어 상기 조성물이 적용된 포유동물의 피부에 미세전류를 흘려 주는 이온토포레시스를 수행하는 단계와, (c) performing iontophoresis, wherein the first mask pack, mask sheet or patch is contacted or adhered to flow a microcurrent into the skin of the mammal to which the composition is applied;
    (d) 상기 미세전류를 통하여 상기 엑소좀을 포유동물 피부 내부로 전달하는 단계를 포함하는, 치료용을 제외한 피부염의 예방, 억제, 완화 또는 개선을 통해 포유동물 피부의 상태를 조절하는 미용방법.(d) delivering the exosomes to the interior of the mammalian skin through the microcurrent, wherein the condition is controlled by preventing, inhibiting, alleviating or ameliorating the dermatitis except for the treatment.
  6. 제5항에 있어서,6. The method of claim 5,
    상기 이온토포레시스 디바이스는 가요성 배터리, 리튬이온 이차 전지, 알칼리 전지, 건전지, 수은 전지, 리튬 전지, 니켈-카드뮴 전지, 및 역전기 투석 전지로 구성된 군으로부터 선택된 적어도 1종의 전지를 포함하거나, 상기 적어도 1종의 전지가 장착된 제2 마스크팩, 마스크시트 또는 패취인, 미용방법.The iontophoresis device includes at least one battery selected from the group consisting of a flexible battery, a lithium ion secondary battery, an alkaline battery, a dry battery, a mercury battery, a lithium battery, a nickel-cadmium battery, A second mask pack, a mask sheet or a patch, on which said at least one battery is mounted.
  7. 제6항에 있어서,The method according to claim 6,
    상기 (b) 단계는 상기 제1 마스크팩, 마스크시트 또는 패취 위에 상기 제2 마스크팩, 마스크시트 또는 패취를 적층하여 수행되는, 미용방법.Wherein the step (b) is performed by laminating the second mask pack, the mask sheet or the patch on the first mask pack, the mask sheet or the patch.
PCT/KR2018/010254 2017-09-07 2018-09-04 New use of exosome kit comprising stem cell-derived exosome WO2019050240A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
KR20170114487 2017-09-07
KR10-2017-0114487 2017-09-07
KR10-2018-0017355 2018-02-13
KR20180017355 2018-02-13
KR10-2018-0101494 2018-08-28
KR1020180101494A KR102197562B1 (en) 2017-09-07 2018-08-28 New use of exosome kit comprising exosomes derived from stem cells

Publications (1)

Publication Number Publication Date
WO2019050240A1 true WO2019050240A1 (en) 2019-03-14

Family

ID=65635067

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2018/010254 WO2019050240A1 (en) 2017-09-07 2018-09-04 New use of exosome kit comprising stem cell-derived exosome

Country Status (1)

Country Link
WO (1) WO2019050240A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3639832A4 (en) * 2017-06-30 2021-04-21 Exocobio Inc. Use of composition comprising adipose stem cell-derived exosome as effective ingredient in alleviating dermatitis

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160033555A (en) * 2014-09-18 2016-03-28 (주)인성정보 The Sheet Mask for Iontophoresis and a mask assembly therewith
WO2016072821A1 (en) * 2014-11-07 2016-05-12 한양대학교 에리카산학협력단 Composition for differentiation induction of adipocyte containing stem cell-derived exosome, regeneration of adipose tissue, and skin whitening or wrinkle improvement
KR20170020245A (en) * 2015-08-12 2017-02-22 (주)프로스테믹스 Mixed culture of adult stem cell and differentiated cell and uses thereof
KR101753003B1 (en) * 2016-05-30 2017-07-03 에이비지노쓰아시아(주) Iontophoresis patch and iontophoresis apparatus having the same
KR20170085010A (en) * 2016-01-12 2017-07-21 주식회사 강스템바이오텍 Stem cell-derived exosomes containing a high amount of growth factors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160033555A (en) * 2014-09-18 2016-03-28 (주)인성정보 The Sheet Mask for Iontophoresis and a mask assembly therewith
WO2016072821A1 (en) * 2014-11-07 2016-05-12 한양대학교 에리카산학협력단 Composition for differentiation induction of adipocyte containing stem cell-derived exosome, regeneration of adipose tissue, and skin whitening or wrinkle improvement
KR20170020245A (en) * 2015-08-12 2017-02-22 (주)프로스테믹스 Mixed culture of adult stem cell and differentiated cell and uses thereof
KR20170085010A (en) * 2016-01-12 2017-07-21 주식회사 강스템바이오텍 Stem cell-derived exosomes containing a high amount of growth factors
KR101753003B1 (en) * 2016-05-30 2017-07-03 에이비지노쓰아시아(주) Iontophoresis patch and iontophoresis apparatus having the same

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3639832A4 (en) * 2017-06-30 2021-04-21 Exocobio Inc. Use of composition comprising adipose stem cell-derived exosome as effective ingredient in alleviating dermatitis
US11612621B2 (en) 2017-06-30 2023-03-28 Exocobio Inc. Use of composition comprising exosome derived from adipose-derived stem cell as effective ingredient in ameliorating dermatitis

Similar Documents

Publication Publication Date Title
Kou et al. The Fas/Fap-1/Cav-1 complex regulates IL-1RA secretion in mesenchymal stem cells to accelerate wound healing
WO2019088656A1 (en) Filler composition containing stabilized exosomes
JP6970459B2 (en) Uses for improving dermatitis of compositions containing adipose-derived stem cell-derived exosomes as an active ingredient
WO2019004757A9 (en) Use of composition comprising stem cell-derived exosome as effective ingredient for prevention or alleviation of pruritus
KR20190069301A (en) Cosmetic composition for preventing, suppressing, alleviating or improving sensitive skin comprising an exosome derived from stem cell as an active ingredient
WO2015088288A1 (en) Method for promoting generation of stem cell-derived exosome by using thrombin
WO2020027466A1 (en) Method for lyophilizing exosome
KR102045188B1 (en) A composition comprising an exosome derived from adipose-derived stem cell as an active ingredient and its application for improving dermatitis
KR20190003399A (en) A composition comprising an exosome derived from stem cell as an active ingredient and its application for improving skin fibrosis
KR20190069277A (en) A composition comprising an exosome and/or extracellular vesicle derived from stem cell as an active ingredient and its application for preventing, suppressing, alleviating or improving sensitive skin
WO2019182299A1 (en) Skin care method using ipl radiation on skin and stem cell-derived exosome treatment in combination
KR20200013599A (en) Freeze-dried formulation of exosomes derived from stem cell and composition for preventing, suppressing, alleviating, improving or treating acne comprising the same
WO2022071643A1 (en) Novel composition comprising edelweiss-derived exosome as active ingredient
WO2021246684A1 (en) Composition for anti-inflammation, wound healing or wound healing promotion, comprising rose stem cell-derived exosomes as active ingredient
KR20190136907A (en) Composition for reducing skin pore size comprising an exosome derived from stem cell as an active ingredient
KR102209937B1 (en) Manufacturing method of exosomes for increasing their productivity and bioactivity and its application
WO2019050240A1 (en) New use of exosome kit comprising stem cell-derived exosome
WO2019004738A2 (en) Use of composition comprising adipose stem cell-derived exosome as effective ingredient in alleviating dermatitis
KR20190069291A (en) Cosmetic composition for moisturizing skin comprising an exosome derived from stem cell as an active ingredient
WO2020027467A1 (en) Lyophilized preparation of stem cell-derived exosomes, and anti-inflammatory composition comprising same as active ingredient
WO2019088545A1 (en) Exosome kit and method for enhancing transdermal permeation of exosomes by using same
WO2019112238A2 (en) Cosmetic composition for moisturizing skin comprising stem cell-derived exosome as active ingredient
KR20190027720A (en) New use of exosome kit comprising exosomes derived from stem cells
WO2019112229A2 (en) New use of exosome kit comprising exosome derived from stem cell
WO2022139439A1 (en) Pharmaceutical composition for preventing or treating dermatitis, containing multifunctional exosomes derived from progenitor cells

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: 18853308

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18853308

Country of ref document: EP

Kind code of ref document: A1