WO2021086139A1 - Method for promoting stem cell-derived exosome production by means of magnetic nanoparticle cluster - Google Patents

Method for promoting stem cell-derived exosome production by means of magnetic nanoparticle cluster Download PDF

Info

Publication number
WO2021086139A1
WO2021086139A1 PCT/KR2020/015093 KR2020015093W WO2021086139A1 WO 2021086139 A1 WO2021086139 A1 WO 2021086139A1 KR 2020015093 W KR2020015093 W KR 2020015093W WO 2021086139 A1 WO2021086139 A1 WO 2021086139A1
Authority
WO
WIPO (PCT)
Prior art keywords
stem cells
polymer
magnetic nanoparticle
nanoparticle cluster
exosomes
Prior art date
Application number
PCT/KR2020/015093
Other languages
French (fr)
Korean (ko)
Inventor
서영준
박동준
기재홍
Original Assignee
연세대학교 원주산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 연세대학교 원주산학협력단 filed Critical 연세대학교 원주산학협력단
Publication of WO2021086139A1 publication Critical patent/WO2021086139A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • CCHEMISTRY; METALLURGY
    • C01INORGANIC CHEMISTRY
    • C01GCOMPOUNDS CONTAINING METALS NOT COVERED BY SUBCLASSES C01D OR C01F
    • C01G49/00Compounds of iron
    • C01G49/02Oxides; Hydroxides
    • CCHEMISTRY; METALLURGY
    • C01INORGANIC CHEMISTRY
    • C01PINDEXING SCHEME RELATING TO STRUCTURAL AND PHYSICAL ASPECTS OF SOLID INORGANIC COMPOUNDS
    • C01P2004/00Particle morphology
    • C01P2004/60Particles characterised by their size
    • C01P2004/64Nanometer sized, i.e. from 1-100 nanometer
    • CCHEMISTRY; METALLURGY
    • C01INORGANIC CHEMISTRY
    • C01PINDEXING SCHEME RELATING TO STRUCTURAL AND PHYSICAL ASPECTS OF SOLID INORGANIC COMPOUNDS
    • C01P2006/00Physical properties of inorganic compounds
    • C01P2006/42Magnetic properties
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2529/00Culture process characterised by the use of electromagnetic stimulation

Definitions

  • the present invention relates to a method for promoting the generation of stem cell-derived exosomes using magnetic nanoparticle clusters, and more particularly, to a method for promoting the generation of stem cell-derived exosomes using a polymer-magnetic nanoparticle cluster.
  • Stem cells are well known as cells that have the ability to proliferate indefinitely and differentiate into various cells under specific conditions. Stem cell therapeutics are in the spotlight as the only future therapeutics that can treat diseases that are currently incurable or incurable by using these differentiation capabilities. Stem cell therapy can regenerate damaged cells and tissues without immune rejection by differentiating and proliferating the stem cells extracted from the patient into necessary cells in the body, such as the cardiovascular system, nervous system, cartilage system, and skin, and injecting them back into the patient. .
  • mesenchymal stem cells have been widely studied for many years for their role in regulating the immune response, immune system activity, and the body's response to inflammation and disease. According to several studies, cultured mesenchymal stem cells have the ability to differentiate into bone and cartilage as well as other cell types and tissues in vitro and in vivo. In addition, mesenchymal stem cells secrete various cytokines and chemokines such as endothelial growth factor, IL-6 (interleukin-6), IL-8 (interleukin-8), etc. Promotes the formation of blood vessels. In addition, mesenchymal stem cells are known to secrete extracellular vesicles (EVs), and extracellular vesicles are known to affect a variety of aspects such as cell fate, function, and differentiation through signal transduction between cells.
  • IL-6 endothelial growth factor
  • IL-8 interleukin-8
  • extracellular vesicles are a generic term for vesicles having a membrane structure secreted by cells out of the cell, and are classified into microvesicles, exosomes, ectosomes, and apoptotic bodies.
  • Double exosomes are defined as extracellular vesicles having a diameter of about 40 to 150 nm and a density of 1.09 to 1.18 g/ml, and have various kinds of active functions. Exosomes contain cell-specific components that reflect the unique biological function of the cell of origin (donor cell). In addition to phospholipids, messenger RNA (mRNA), microRNA (microRNA, miRNA), various soluble proteins, exogenous proteins, and It includes transmembrane protein components and the like.
  • mRNA messenger RNA
  • microRNA microRNA
  • miRNA various soluble proteins
  • exogenous proteins and It includes transmembrane protein components and the like.
  • CD63 and CD81 are well known. These include receptors on the surface of cells such as EGFR, molecules involved in signal transduction, proteins involved in cell adhesion, and heat shock proteins. protein) and Alix related to endoplasmic reticulum formation. These exosomes can be isolated from various types of body fluids, such as saliva, urine, plasma, serum, and amniotic fluid, and can also be isolated from various types of cell culture supernatant.
  • exosomes have been found to have clinical therapeutic efficacy in vitro and in vivo , so exosomes against various diseases that have been attempted using mesenchymal stem cells. It is emerging as a new alternative. Exosomes also play a role in immune regulation and signal transduction between cells, inducing functional changes in cells to activate cell regeneration programs, and because they are cell-free system, there is no risk of tumor formation and at -20°C without cryopreservatives It is preserved in a state in which biological activity is maintained for months and is encapsulated, so there is an advantage that substances in exosomes are not decomposed.
  • the present inventors have endeavored to develop a method that can increase the occurrence of exosomes in mesenchymal stem cells, and as a result, the amount of inflow into the mesenchymal stem cells varies depending on the surface modification properties and magnetic stimulation of the magnetic nanoparticle cluster. , It was confirmed that the amount of exosomes was affected by this. Therefore, it was found that the generation and activity of exosomes in mesenchymal stem cells can be maximized using magnetic nanoparticle clusters and magnetic stimulation, and thus the present invention was completed.
  • An object of the present invention is to provide a method for promoting the generation of exosomes derived from stem cells.
  • the present invention provides a method for promoting the generation of stem cells-derived exosomes using a polymer-magnetic nanoparticle cluster.
  • the generation of stem cell-derived exosomes was promoted when a polymer-magnetic nanoparticle cluster was treated on stem cells and a magnetic force was applied to the stem cells.
  • a method of promoting production it has the advantage of providing a large amount of high-quality exosomes with clinical therapeutic efficacy in vitro and in vivo.
  • FIG. 1 is a schematic diagram showing a mechanism by which the generation of exosomes in stem cells is promoted by polymer-magnetic nanoparticle clusters and magnetic stimulation.
  • FIG. 2 is a diagram showing a polymer-superparamagnetic nanoparticle cluster surface-modified with negative charge (FIG. 2, upper) or positive charge (FIG. 2, lower).
  • 3A is a diagram illustrating the size of a polymer-magnetic nanoparticle cluster surface-modified with a positive charge.
  • 3B is a diagram showing a polymer-magnetic nanoparticle cluster observed by zeta potential ( ⁇ (zeta, z)-potential), SEM and TEM of a polymer-magnetic nanoparticle cluster.
  • FIG. 4 is a diagram showing the expression level of exosomes according to the surface charge and concentration of the polymer-magnetic nanoparticle cluster.
  • 5 is a diagram showing the size of the expressed exosomes.
  • CD9 is a diagram showing the expression levels of CD9, CD63, and CD81, which are exosome-specific markers according to the strength of magnetic force.
  • the present invention is a.
  • step 2 2) applying a magnetic force to the stem cells of step 1); provides a method for promoting the generation of stem cell-derived exosomes.
  • the polymer of step 1) may be a biocompatible polymer, and more specifically, as the biocompatible polymer, poly(beta-hydroxyethyl methacrylate) (Poly(beta-hydroxyethyl Methacrylate), PHEMA); Polyacrylamide (PA); Polyvinyl alcohol (PVA); Polyacrylic acid (PAA), its salt and its derivative; Polymethacrylic acid (Poly (metha acrylic acid), PMAA) or a derivative thereof; Poly(acrylic amide) or a derivative thereof; Poly(undecenoic acid) or a derivative thereof, or a copolymer of the polymer; Dextran or a derivative thereof, polyvinylpyrrolidone (PVP); Polyethylene oxide (PEO); Polyethyleneglycol (PEG) or a derivative thereof; Polypropylene Glycol (PPG) or a derivative thereof; A copolymer of the polyethylene glycol and polypropylene glycol, or a monoesterified derivative thereof; Poly(ethylene oxide-b-
  • the magnetism of step 1) may be superparamagnetic, but is not limited thereto.
  • the magnetic nanoparticles of step 1) are ferric oxide (iron(II) oxide: FeO), iron(III) oxide, magnemite; Fe 2 O 3 , ⁇ -Fe 2 O 3 , ⁇ - Fe 2 O 3 , ⁇ -Fe 2 O 3 , ⁇ -Fe 2 O 3 ), magnetite (Fe 3 O 4 ) and iron(II, III) oxides; Fe 4 O 5 , Fe 5 O 6 , Fe 5 O 7 , Fe 13 O 19 , Fe 25 O 32 ) may be one or more selected from the group consisting of, but is not limited thereto.
  • the polymer-magnetic nanoparticle cluster in step 1) is preferably a polymer-supermagnetic nanoparticle cluster surface-modified with a positive charge.
  • the positively charged polymer-magnetic nanoparticle cluster may have a cationic transformant attached to the surface of the polymer-magnetic nanoparticle cluster, and more specifically, polyethyleneimine (PEI) as the cationic transformant, Pegyl Pegylated polyethyleneimine, Histidylated polyethyleneimine, Lactosylated polyethyleneimine, Folate-conjugated polyethyleneimine, Melittin-conjugated Polyethyleneimine derivatives such as polyethylinimine; Polylysine, Spermine, Protamine sulfate, Polyamidoamine (PAMAM), Polypropyleneimine, Polybrene, and DEAE-dextran At least one cationic transformant selected from the group consisting of may be attached, but is not limited thereto.
  • PEI polyethyleneimine
  • Pegyl Pegylated polyethyleneimine Histidylated polyethyleneimine, Lactosylated polyethyleneimine, Folate-con
  • the polymer-magnetic nanoparticle cluster may be prepared by the following method, but is not limited thereto:
  • step c) mixing the polymer of step a) and the magnetic nanoparticles of step b);
  • step d) ultrasonicating the mixture of step c) and stirring at room temperature.
  • step a') may be added to the step a):
  • step a′ adding and reacting a cationic transformant and a linker connecting the cationic transformant and the polymer to step a) to obtain a polymer to which the cationic transformant is attached.
  • the linker may be specifically N,N'-dicyclohexylcarbodiimide (N,N'-Dicyclohexylcarbodiimide, DCC) and N-hydroxysuccinimide (N-Hydroxysuccinimide, NHS), but is not limited thereto.
  • step c) may be performed by vortexing for 3 to 7 minutes, but is not limited thereto.
  • a fluorescently attached polymer may be added in step c) to prepare a fluorescently-labeled polymer-magnetic nanoparticle cluster.
  • the ultrasonic treatment in step d) may be specifically performed for 1 to 5 minutes, more specifically for 2 to 4 minutes.
  • Stirring in step d) may be performed for 1 to 24 hours, specifically for 4 to 8 hours, and more specifically for 5 to 7 hours.
  • the polymer-magnetic nanoparticle cluster prepared by the above method has a cluster form in which a polymer and magnetic nanoparticles are aggregated.
  • polymer-magnetic nanoparticle cluster may be treated at 1 to 1000 ⁇ g/ml in step 1), but is not limited thereto.
  • the stem cells may be derived from one or more tissues selected from the group consisting of bone marrow, adipose tissue, peripheral blood, liver, muscle, lung, amniotic fluid, placental chorion, and umbilical cord blood, but is not limited thereto.
  • the stem cells may be embryonic stem cells or adult stem cells, but are not limited thereto.
  • the adult stem cells may be one or more adult stem cells selected from the group consisting of mesenchymal stem cells, human tissue-derived mesenchymal stromal cells, human tissue-derived mesenchymal stem cells, and multipotent stem cells, but are not limited thereto. .
  • the magnetic force may be applied in an amount of 0.1, 0.2, 0.3, 0.4, 0.6, or 1.0 T or more, specifically 0.1 to 2 T, and more specifically 0.3 to 1 T, but is not limited thereto. Does not.
  • the present invention comprises the steps of: 1) treating a polymer-magnetic nanoparticle cluster on stem cells;
  • step 3 separating the exosomes from the stem cells of step 2); and a method for preparing an exosome including, and an exosome prepared according to the preparation method.
  • the present invention provides a composition for promoting the generation of exosomes derived from stem cells comprising a polymer-magnetic nanoparticle cluster (cluster).
  • any general medium used for culturing stem cells may be used.
  • DMEM Dulbecco's Modified Eagle's Medium
  • MEM Minimal Essential Medium
  • BME Base Medium Eagle
  • RPMI 1640 DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10)
  • DMEM/F-12 Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12
  • ⁇ -MEM ⁇ -Minimal Essential Medium
  • G-MEM Gasgow's Minimal Essential Medium
  • IMDM Isocove's Modified Dulbecco's Medium
  • KnockOut DMEM more preferably a minimum essential nutrient medium (minimum essential medium, MEM) It may be, but is not limited thereto.
  • the medium may contain other components, such as antibiotics or antifungal agents (eg, penicillin, streptomycin), glutamine, and the like.
  • antibiotics or antifungal agents eg, penicillin, streptomycin
  • glutamine eg, glutamine, and the like.
  • the present inventors confirmed that the generation of exosomes derived from mesenchymal stem cells is promoted by polymer-magnetic nanoparticle clusters and magnetic stimulation.
  • the present inventors confirmed that when a macromolecular-magnetic nanoparticle cluster was treated on stem cells and a magnetic force was applied to the stem cells, the generation of stem cell-derived exosomes was promoted, so that the generation of the exosomes
  • the method of maximizing the value can be usefully used.
  • MSC Mesenchymal stem cells
  • MUBMX-01001 Mesenchymal stem cells
  • FBS fetal bovine serum
  • DPBS Dulbecco's Phosphate-Buffered Saline
  • PCS nanoparticles Polymerized clustered superparamagnetic iron oxide (PCS) nanoparticles were made of iron oxide-based nanoparticles, and the size of the nanoparticles was about 100 nm. In order to determine the PCS nanoparticle treatment conditions, the concentrations of the nanoparticles were prepared at 20, 40 and 60 ⁇ g/ml for 24 hours.
  • poly(D, L-lactide-co-glycolide) Poly(D, L-lactide-co-glycolide, PLGA)) (50:50, M W 38,000 to 54,000) (Sigma-Aldrich, US), superparamagnetic iron oxide (SPIO) nanoparticles, SPIONs) (Sigma-Aldrich, US) were used, and 1-ethyl-3-(3-dimethylaminopropyl) Carbodiimide hydrochloride (1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride, EDC) (Thermo-fisher Scientigic, US) and N-hydroxysuccinimide (NHS) were used as fluorescent molecules (fluorescence).
  • cyanine 5.5 (Cyanine5.5, Cy5.5) (Cyanine5.5 amine, Lumiprobe Co., US) was used as a fluorescent molecule to detect nanoparticles in cells.
  • Positively charged surface-modified PCS nanoparticles were prepared as follows. In order to determine the PCS nanoparticle treatment conditions, the concentrations of the nanoparticles were prepared at 20, 40 and 60 ⁇ g/ml for 24 hours.
  • poly(D, L-lactide-co-glycolide) Poly(D, L-lactide-co-glycolide, PLGA))(50:50, M W 38,000 ⁇ 54,000) (Sigma-Aldrich, US) and polyethyleneimine (PEI) (M W ⁇ 25,000 by LS, M n ⁇ 10,000 by GPC) (Sigma-Aldrich, US) was used, and N,N'-dicyclo Hexylcarbodiimide (N,N'-Dicyclohexylcarbodiimide, DCC) and N-Hydroxysuccinimide (NHS) were used to prepare a linker between PLGA and PEI.
  • the PLGA-Cy5.5 fluorescent polymer was synthesized in the same manner as described in ⁇ Example 2>.
  • PLGA-PEI copolymer PLGA-Cy5.5 and SPIONs were used.
  • 0.4 mg of PLGA-Cy5.5 and 0.1 mg of PLGA-PEI were dissolved in 0.1 ml of DMSO, and 0.1 ml of SPIONs (5 mg/ml) was added dropwise to 3 ml of deionized water. Thereafter, the vial was vortexed for 5 minutes, sonicated for 3 minutes, and stirred at room temperature for 6 hours to obtain positively charged surface-modified PCS nanoparticles. The obtained positively charged surface-modified PCS nanoparticles were freeze-dried to obtain a powder sample (Fig. 2, bottom).
  • the mixture was incubated at 6° C. overnight, and then centrifuged at 6° C. for 1 hour with a centrifugal force of 10,000 xg, and the supernatant was discarded and a pellet settled on the bottom of the tube was recovered. Subsequently, the recovered pellet was re-suspended in 1X PBS and stored at -20°C for a long period of time.
  • the cells were fixed in 2.5% glutaraldehyde for 2 hours at 4°C, and solidified with 2% agar. After washing with 0.1 M cacodylate buffer, it was fixed in 1% osmium tetroxide (OsO 4 ). The dehydration step was carried out using 50-100% ethanol and embedded in Epon resin. Ultrathin sections were cut using an ultramicrotome, and stained using uranyl acetate and lead citrate. Then, it was observed using TEM under the condition of 63 kV.
  • the size and surface charge of the PCS nanoparticles surface-modified with positive charges prepared according to the ⁇ Example 3> were measured by DLS (dynamic light scattering) Zetasizer Nano-ZS90 (Malvern Instruments Ltd., UK).
  • the PCS nanoparticles were placed on a carbonyl-coated 400 mesh copper grid (CF400-CU-UL, Electron Microscopy Sciences, US) for 15 minutes. Then, 10 ⁇ l of 2% uranyl acetate was placed on the grid for 10 minutes, dried and negatively stained. Then, using an electron microscope (JEM-2100F, JEOL Ltd, Japan), a TEM (transmission electron microscopic) image was obtained under the condition of 63 kV to confirm the structure of the nanoparticles. In addition, a scanning electron microscopic (SEM) image was obtained using an electron microscope to confirm the shape of the nanoparticles.
  • JEM-2100F transmission electron microscopic
  • the PCS nanoparticles surface-modified with a positive charge exhibited a cluster form in which PLGA and iron oxide nanoparticles were aggregated as shown in FIG. 2, and have a positive charge. I did. Accordingly, the PCS nanoparticles were named as polymer-magnetic nanoparticle clusters, and the PCS nanoparticles surface-modified with positive charges were polymer-magnetic nanoparticle clusters surface-modified with positive charges, and the PCS nanoparticles surface-modified with negative charges were negatively charged. It was named as a surface-modified polymer-magnetic nanoparticle cluster.
  • the concentration of the polymer-magnetic nanoparticle cluster surface-modified with positive/negative charges prepared in ⁇ Examples 2 and 3> above.
  • the mesenchymal stem cells were treated and micromagnetic was applied, and then the amount of exosomes produced in the mesenchymal stem cells was analyzed.
  • the polymer-magnetic nanoparticle cluster surface-modified with a positive charge prepared in ⁇ Example 3> was treated on the mesenchymal stem cells at a concentration of 40 ⁇ g/ml in the same manner as in the method described in ⁇ Example 4>. , 0.3 T, 0.4 T, 0.6 T and 1.0 T magnetic force was applied. Then, the amount of exosomes produced from mesenchymal stem cells was analyzed through TEM observation in the same manner as described in ⁇ Example 5>.
  • the polymer-magnetic nanoparticle clusters surface-modified with positive charges prepared in ⁇ Example 3> were treated at concentrations of 20, 40, and 60 ⁇ g/mL, and after applying magnetic force, mesenchymal stem The amount of exosomes produced from cells was analyzed.
  • mesenchymal stem cells without treatment of polymer-magnetic nanoparticle clusters were used.
  • the polymer-magnetic nanoparticle cluster surface-modified with the positive charge prepared in ⁇ Example 3> was treated to the mesenchymal stem cells, and micromagnets were applied by varying the strength. Afterwards, the amount of exosomes produced in mesenchymal stem cells was analyzed.
  • the polymer-magnetic nanoparticle clusters surface-modified with positive charges prepared in ⁇ Example 3> were treated at concentrations of 20, 40 and 60 ⁇ g/ml, and 0.3
  • Western blotting was performed in the same manner as described in ⁇ Example 5> to measure the expression level of exosome proteins.
  • the surface modification of the nanoparticle cluster causes a difference due to the zeta-potential, and the more the nanoparticle cluster has a positive property rather than a negative property, the greater the amount of the nanoparticle cluster flows into the stem cells. As a result, it can be seen that the amount of exosomes produced increases when magnetic force is applied.
  • the present invention relates to a method for promoting generation of exosomes derived from mesenchymal stem cells using magnetic nanoparticle clusters, and specifically, when a polymer-magnetic nanoparticle cluster is treated on stem cells and magnetic force is applied to the stem cells, Since it promotes the generation of stem cell-derived exosomes, it can be usefully used to provide a large amount of high-quality exosomes with clinical therapeutic efficacy both in vitro and in vivo.

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Inorganic Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention relates to a method for promoting mesenchymal stem cell-derived exosome production by means of a magnetic nanoparticle cluster. Particularly, it is confirmed that stem cell-derived exosome production is promoted by treating a stem cell with a polymer-magnetic nanoparticle cluster and applying a magnetic force to the stem cell. Therefore, a large amount of high-quality exosomes exhibiting a clinical therapeutic efficacy can be provided in vitro and in vivo.

Description

자성 나노입자 클러스터를 이용한 줄기세포 유래 엑소좀 생성 촉진 방법Stem cell-derived exosome generation promotion method using magnetic nanoparticle cluster
본 발명은 자성 나노입자 클러스터를 이용한 줄기세포 유래 엑소좀 생성 촉진 방법에 관한 것으로, 보다 구체적으로 고분자-자성 나노입자 클러스터를 이용한 줄기세포 유래 엑소좀 생성 촉진 방법에 관한 것이다.The present invention relates to a method for promoting the generation of stem cell-derived exosomes using magnetic nanoparticle clusters, and more particularly, to a method for promoting the generation of stem cell-derived exosomes using a polymer-magnetic nanoparticle cluster.
줄기세포는 무한히 증식하는 자가 생산능을 가지며, 특정 조건에서 다양한 세포로 분화 가능한 세포로 잘 알려져 있다. 이러한 분화 능력을 이용하여 현재 의학으로 치료 불가능하거나 난치성인 질환을 치료할 수 있는 유일한 미래 치료제로서 줄기세포 치료제가 각광받고 있다. 줄기세포 치료제는 환자에서 추출한 줄기세포를 체외에서 심혈관계, 신경계, 연골계, 피부 등 필요한 계열의 세포로 분화 및 증식하여 다시 환자에 주입하는 방식으로 면역거부반응 없이 손상된 세포 및 조직을 재생할 수 있다.Stem cells are well known as cells that have the ability to proliferate indefinitely and differentiate into various cells under specific conditions. Stem cell therapeutics are in the spotlight as the only future therapeutics that can treat diseases that are currently incurable or incurable by using these differentiation capabilities. Stem cell therapy can regenerate damaged cells and tissues without immune rejection by differentiating and proliferating the stem cells extracted from the patient into necessary cells in the body, such as the cardiovascular system, nervous system, cartilage system, and skin, and injecting them back into the patient. .
줄기세포 중에서도 중간엽줄기세포(mesenchymal stem cells, MSCs)는 면역 반응, 면역계 활동 및 염증과 질병에 대한 신체 반응의 조절에 있어서 그 역할이 수년 동안 널리 연구되어 왔다. 여러 연구에 의하면 배양된 중간엽줄기세포는 체외와 생체 내에서 다른 세포 유형과 조직뿐만 아니라 뼈와 연골로 분화하는 능력을 가지고 있다. 또한, 중간엽줄기세포는 내피세포 성장인자(endothelial growth factor), IL-6(interleukin-6), IL-8(interleukin-8) 등과 같은 다양한 사이토카인과 케모카인들을 분비함으로써 조직 재생과 손상 조직 내 혈관 형성을 촉진한다. 또한, 중간엽줄기세포는 세포 외 소포체(extracellular vesicle, EV)를 분비하는 것으로 알려져 있으며, 세포 외 소포체가 세포 간의 신호전달을 통해 세포의 운명, 기능, 분화 등 다방면에 영향을 미치는 것으로 알려져 있다.Among stem cells, mesenchymal stem cells (MSCs) have been widely studied for many years for their role in regulating the immune response, immune system activity, and the body's response to inflammation and disease. According to several studies, cultured mesenchymal stem cells have the ability to differentiate into bone and cartilage as well as other cell types and tissues in vitro and in vivo. In addition, mesenchymal stem cells secrete various cytokines and chemokines such as endothelial growth factor, IL-6 (interleukin-6), IL-8 (interleukin-8), etc. Promotes the formation of blood vessels. In addition, mesenchymal stem cells are known to secrete extracellular vesicles (EVs), and extracellular vesicles are known to affect a variety of aspects such as cell fate, function, and differentiation through signal transduction between cells.
한편, 세포 외 소포체는 세포가 세포 외로 분비하는 막 구조의 소포체의 총칭으로 마이크로소포체(microvesicle), 엑소좀(exosome), 엑토좀(ectosome), 사멸체(apoptotic body) 등으로 구분된다. 이중 엑소좀은 약 40 내지 150 ㎚의 직경과 1.09 내지 1.18 g/㎖의 밀도를 가지는 세포 외 소포체로 정의되는데, 다양한 종류의 활성 기능이 있다. 엑소좀은 기원 세포(공여세포) 특유의 생물학적 기능을 반영하는 세포특이적 구성 성분을 함유하며, 인지질, 메신저RNA(messengerRNA, mRNA), 마이크로RNA(microRNA, miRNA) 외에도 다양한 수용성 단백질, 외재성 단백질 및 막관통 단백질 성분 등을 포함한다. 엑소좀의 마커 단백질로는 CD63, CD81 등이 잘 알려져 있으며, 여기에는 주로 EGFR과 같은 세포 표면의 수용체, 신호전달에 관여하는 분자, 세포의 부착(adhesion)에 관여하는 단백질, 열충격 단백질(heat shock protein), 소포체 형성과 관련하는 Alix 등의 단백질이 포함되는 것으로 알려져 있다. 이러한 엑소좀은 다양한 종류의 체액, 예를 들어 침, 소변, 혈장, 혈청, 양수로부터 분리할 수 있고 여러 종류의 세포배양 상층액에서도 분리가 가능하다.On the other hand, extracellular vesicles are a generic term for vesicles having a membrane structure secreted by cells out of the cell, and are classified into microvesicles, exosomes, ectosomes, and apoptotic bodies. Double exosomes are defined as extracellular vesicles having a diameter of about 40 to 150 nm and a density of 1.09 to 1.18 g/ml, and have various kinds of active functions. Exosomes contain cell-specific components that reflect the unique biological function of the cell of origin (donor cell). In addition to phospholipids, messenger RNA (mRNA), microRNA (microRNA, miRNA), various soluble proteins, exogenous proteins, and It includes transmembrane protein components and the like. As marker proteins for exosomes, CD63 and CD81 are well known. These include receptors on the surface of cells such as EGFR, molecules involved in signal transduction, proteins involved in cell adhesion, and heat shock proteins. protein) and Alix related to endoplasmic reticulum formation. These exosomes can be isolated from various types of body fluids, such as saliva, urine, plasma, serum, and amniotic fluid, and can also be isolated from various types of cell culture supernatant.
아울러, 순수하게 분리한 엑소좀은 생체외(in vitro) 및 생체내(in vivo)에서 임상적 치료효능이 있는 것으로 밝혀지고 있어, 그동안 중간엽줄기세포를 이용하여 시도되던 여러 질환에 대해 엑소좀이 새로운 대안으로 떠오르고 있다. 엑소좀은 또한 면역조절 및 세포 간 신호전달 역할을 하며 세포의 기능적 변화를 유도하여 세포재생 프로그램을 활성화하며, 무세포계(cell free system)이므로 종양 형성의 위험이 없고 동결보존제 없이 영하 20℃에서 6개월간 생물학적 활성이 유지되는 상태로 보존되며 캡슐화(encapsulation)되어 있어 엑소좀 내 물질이 분해되지 않는 장점이 있다.In addition, purely isolated exosomes have been found to have clinical therapeutic efficacy in vitro and in vivo , so exosomes against various diseases that have been attempted using mesenchymal stem cells. It is emerging as a new alternative. Exosomes also play a role in immune regulation and signal transduction between cells, inducing functional changes in cells to activate cell regeneration programs, and because they are cell-free system, there is no risk of tumor formation and at -20℃ without cryopreservatives It is preserved in a state in which biological activity is maintained for months and is encapsulated, so there is an advantage that substances in exosomes are not decomposed.
다만, 이러한 우수한 효과에도 불구하고 엑소좀을 상업적으로 이용하기 위해서는 다량의, 그리고 양질의 엑소좀이 필요하다. 그러나 현재 중간엽줄기세포로부터 얻을 수 있는 엑소좀의 양은 매우 소량에 불과하고, 중간엽줄기세포 유래 엑소좀의 생산량을 증가시킬 수 있는 방법 및 물질에 대한 개발도 아직은 미비한 실정이다. 이에 따라 중간엽줄기세포 유래 엑소좀의 생산을 촉진하는 신규한 방법 및 물질 개발에 대한 필요성이 제기되었다.However, despite these excellent effects, in order to use exosomes commercially, a large amount of and high quality exosomes are required. However, the amount of exosomes that can be obtained from mesenchymal stem cells is currently only a very small amount, and the development of methods and materials capable of increasing the production of exosomes derived from mesenchymal stem cells is still insufficient. Accordingly, the need for the development of novel methods and materials for promoting the production of mesenchymal stem cells-derived exosomes has been raised.
이에 본 발명자들은 중간엽줄기세포 내 엑소좀의 발생을 증가시킬 수 있는 방법을 개발하기 위해 노력한 결과, 자성 나노입자 클러스터의 표면 개질 성질 및 자성자극에 따라 중간엽줄기세포 내부로의 유입량이 달라지고, 이로 인해 엑소좀의 발생량이 영향을 받는 것을 확인하였다. 따라서 자성 나노입자 클러스터 및 자성자극을 이용하여 중간엽줄기세포에서 엑소좀의 발생 및 활성이 극대화할 수 있음을 밝혔으며, 이에 본 발명을 완성하였다.Accordingly, the present inventors have endeavored to develop a method that can increase the occurrence of exosomes in mesenchymal stem cells, and as a result, the amount of inflow into the mesenchymal stem cells varies depending on the surface modification properties and magnetic stimulation of the magnetic nanoparticle cluster. , It was confirmed that the amount of exosomes was affected by this. Therefore, it was found that the generation and activity of exosomes in mesenchymal stem cells can be maximized using magnetic nanoparticle clusters and magnetic stimulation, and thus the present invention was completed.
본 발명의 목적은 줄기세포 유래 엑소좀 생성 촉진 방법을 제공하기 위한 것이다.An object of the present invention is to provide a method for promoting the generation of exosomes derived from stem cells.
상기 목적을 달성하기 위하여, 본 발명은 고분자-자성 나노입자 클러스터를 이용한 줄기세포 유래 엑소좀 생성 촉진 방법을 제공한다.In order to achieve the above object, the present invention provides a method for promoting the generation of stem cells-derived exosomes using a polymer-magnetic nanoparticle cluster.
본 발명에서는 고분자-자성 나노입자 클러스터(cluster)를 줄기세포에 처리하고 상기 줄기세포에 자기력을 가했을 때, 줄기세포 유래 엑소좀의 생성이 촉진되는 것을 확인하였으므로, 본 발명에 따른 줄기세포 유래 엑소좀 생성 촉진 방법으로 생체외(in vitro) 및 생체내(in vivo)에서 임상적 치료효능이 있는 양질의 엑소좀을 다량으로 제공할 수 있는 이점을 가진다.In the present invention, it was confirmed that the generation of stem cell-derived exosomes was promoted when a polymer-magnetic nanoparticle cluster was treated on stem cells and a magnetic force was applied to the stem cells. As a method of promoting production, it has the advantage of providing a large amount of high-quality exosomes with clinical therapeutic efficacy in vitro and in vivo.
도 1은 고분자-자성 나노입자 클러스터(cluster) 및 자성자극에 의해 줄기세포에서 엑소좀의 생성이 촉진되는 기전을 모식도로 나타낸 도이다.1 is a schematic diagram showing a mechanism by which the generation of exosomes in stem cells is promoted by polymer-magnetic nanoparticle clusters and magnetic stimulation.
도 2는 음전하(도 2, 위) 또는 양전하(도 2, 아래)로 표면 개질된 고분자-초상자성 나노입자 클러스터를 나타낸 도이다.FIG. 2 is a diagram showing a polymer-superparamagnetic nanoparticle cluster surface-modified with negative charge (FIG. 2, upper) or positive charge (FIG. 2, lower).
도 3a는 양전하로 표면 개질된 고분자-자성 나노입자 클러스터의 크기를 확인한 도이다.3A is a diagram illustrating the size of a polymer-magnetic nanoparticle cluster surface-modified with a positive charge.
도 3b는 고분자-자성 나노입자 클러스터의 제타 전위(ζ(zeta, z)-potential), SEM 및 TEM으로 관찰한 고분자-자성 나노입자 클러스터를 나타낸 도이다.3B is a diagram showing a polymer-magnetic nanoparticle cluster observed by zeta potential (ζ(zeta, z)-potential), SEM and TEM of a polymer-magnetic nanoparticle cluster.
도 4는 고분자-자성 나노입자 클러스터의 표면 전하 및 농도에 따른 엑소좀의 발현 수준을 나타낸 도이다.4 is a diagram showing the expression level of exosomes according to the surface charge and concentration of the polymer-magnetic nanoparticle cluster.
도 5는 발현된 엑소좀의 크기를 나타낸 도이다.5 is a diagram showing the size of the expressed exosomes.
도 6은 자기력 세기에 따른 엑소좀 특이적 마커인 CD9, CD63 및 CD81의 발현 수준을 나타낸 도이다.6 is a diagram showing the expression levels of CD9, CD63, and CD81, which are exosome-specific markers according to the strength of magnetic force.
이하, 본 발명을 보다 상세히 설명한다.Hereinafter, the present invention will be described in more detail.
본 발명은The present invention
1) 고분자-자성 나노입자 클러스터(cluster)를 줄기세포에 처리하는 단계; 및1) treating a macromolecular-magnetic nanoparticle cluster on stem cells; And
2) 상기 단계 1)의 줄기세포에 자기력을 가하는 단계;를 포함하는 줄기세포 유래 엑소좀의 생성 촉진 방법을 제공한다.2) applying a magnetic force to the stem cells of step 1); provides a method for promoting the generation of stem cell-derived exosomes.
상기 단계 1)의 고분자는 생체적합성 고분자일 수 있고, 보다 구체적으로 상기 생체적합성 고분자로 폴리(베타-히드록시 에틸 메타아크릴레이트)(Poly(beta-hydroxyethyl Methacrylate), PHEMA); 폴리아크릴아미드(Polyacrylamide, PA); 폴리비닐 알코올(Polyvinyl Alcohol, PVA); 폴리아크릴산(Polyacrylic Acid, PAA), 그 염 및 그 유도체; 폴리메타아크릴산(Poly(metha acrylic Acid), PMAA) 또는 그 유도체; 폴리아크릴아미드(Poly(acrylic amide)) 또는 그 유도체; 폴리언데세노산(Poly(undecenoic acid)) 또는 그 유도체, 또는 상기 고분자의 공중합체; 덱스트란(Dextran) 또는 그 유도체, 폴리비닐피롤리돈(Polyvinylpyrrolidone, PVP); 폴리에틸렌 옥사이드(Polyethyleneoxide, PEO); 폴리에틸렌글리콜(Polyethyleneglycol, PEG) 또는 그 유도체; 폴리프로필렌글리콜(Polypropylene Glycol, PPG) 또는 그 유도체; 상기 폴리에틸렌글리콜 및 폴리프로필렌글리콜의 공중합체, 또는 이들의 모노에스테르화 유도체; 폴리(에틸렌옥시드-b-프로필렌 옥사이드(Poly(ethylene oxide-b-propylene oxide), PEO-PPO); 폴리-l-라이신(Polyl-l-lysine); 폴리에틸렌이민(Polyethylenimine); 폴리락트산(Poly lactic acid, PLA); 폴리글리콜산(Polyglycolic acid, PGA); 폴리(락트산-co-글리콜산)(Poly(lactic- co -glycolic acid), PLGA) 공중합체; 키토산(Chitosan); 히알루론산(Hyaluronic acid); 및 이들의 혼합물로 이루어진 군으로부터 선택된 1종 이상일 수 있으나, 이에 한정되지 않는다.The polymer of step 1) may be a biocompatible polymer, and more specifically, as the biocompatible polymer, poly(beta-hydroxyethyl methacrylate) (Poly(beta-hydroxyethyl Methacrylate), PHEMA); Polyacrylamide (PA); Polyvinyl alcohol (PVA); Polyacrylic acid (PAA), its salt and its derivative; Polymethacrylic acid (Poly (metha acrylic acid), PMAA) or a derivative thereof; Poly(acrylic amide) or a derivative thereof; Poly(undecenoic acid) or a derivative thereof, or a copolymer of the polymer; Dextran or a derivative thereof, polyvinylpyrrolidone (PVP); Polyethylene oxide (PEO); Polyethyleneglycol (PEG) or a derivative thereof; Polypropylene Glycol (PPG) or a derivative thereof; A copolymer of the polyethylene glycol and polypropylene glycol, or a monoesterified derivative thereof; Poly(ethylene oxide-b-propylene oxide, PEO-PPO); Polyl-l-lysine; Polyethylenimine; Polylactic acid lactic acid, PLA); Polyglycolic acid (PGA); Poly(lactic-co-glycolic acid) (PLGA) copolymer; Chitosan; Hyaluronic acid (Hyaluronic acid) acid); And may be one or more selected from the group consisting of a mixture thereof, but is not limited thereto.
또한, 상기 단계 1)의 자성은 초상자성일 수 있으나, 이에 한정되지 않는다.In addition, the magnetism of step 1) may be superparamagnetic, but is not limited thereto.
상기 단계 1)의 자성 나노입자는 산화 제2철(iron(Ⅱ) oxide: FeO), 산화 제3철(iron(Ⅲ) oxide, magnemite; Fe2O3, α-Fe2O3, β-Fe2O3, γ-Fe2O3, ε-Fe2O3), 마그네타이트(magnetite; Fe3O4) 및 산화 제2, 3철(iron(Ⅱ, Ⅲ) oxides; Fe4O5, Fe5O6, Fe5O7, Fe13O19, Fe25O32)로 이루어진 군으로부터 선택된 1종 이상일 수 있으나, 이에 한정되지 않는다.The magnetic nanoparticles of step 1) are ferric oxide (iron(II) oxide: FeO), iron(III) oxide, magnemite; Fe 2 O 3 , α-Fe 2 O 3 , β- Fe 2 O 3 , γ-Fe 2 O 3 , ε-Fe 2 O 3 ), magnetite (Fe 3 O 4 ) and iron(II, Ⅲ) oxides; Fe 4 O 5 , Fe 5 O 6 , Fe 5 O 7 , Fe 13 O 19 , Fe 25 O 32 ) may be one or more selected from the group consisting of, but is not limited thereto.
또한, 상기 단계 1)에서 고분자-자성 나노입자 클러스터는 양전하로 표면 개질된 고분자-초자성 나노입자 클러스터인 것이 바람직하다.In addition, the polymer-magnetic nanoparticle cluster in step 1) is preferably a polymer-supermagnetic nanoparticle cluster surface-modified with a positive charge.
*상기 양전하로 표면 개질된 고분자-자성 나노입자 클러스터는 고분자-자성 나노입자 클러스터 표면에 양이온성 형질 전환제가 부착될 수 있고, 보다 구체적으로 상기 양이온성 형질 전환제로 폴리에틸렌이민(Polyethyleneimine, PEI), 페길레이티드 (Pegylated) 폴리에틸렌이민, 히스티딜레이티드 (Histidylated) 폴리에틸렌이민, 락토실레이티드 (Lactosylated) 폴리에틸렌이민, 엽산 공유결합형(Folate-conjugated) 폴리에틸렌이민, 멜리틴 공유결합형 (Melittin-conjugated) 폴리에틸린이민 등의 폴리에틸렌이민 유도체; 폴리라이신(Polylysine), 스퍼민(Spermine), 프로타민 설페이트(Protamine sulfate), 폴리아미도아민 (Polyamidoamine, PAMAM), 폴리 프로필렌이민 (Polypropyleneimine), 폴리브렌 (Polybrene) 및 디이에이이 덱스트란 (DEAE-dextran)으로 이루어진 군으로부터 선택된 1종 이상의 양이온성 형질 전환제가 부착될 수 있으나, 이에 한정되지 않는다.*The positively charged polymer-magnetic nanoparticle cluster may have a cationic transformant attached to the surface of the polymer-magnetic nanoparticle cluster, and more specifically, polyethyleneimine (PEI) as the cationic transformant, Pegyl Pegylated polyethyleneimine, Histidylated polyethyleneimine, Lactosylated polyethyleneimine, Folate-conjugated polyethyleneimine, Melittin-conjugated Polyethyleneimine derivatives such as polyethylinimine; Polylysine, Spermine, Protamine sulfate, Polyamidoamine (PAMAM), Polypropyleneimine, Polybrene, and DEAE-dextran At least one cationic transformant selected from the group consisting of may be attached, but is not limited thereto.
상기 단계 1)에서 고분자-자성 나노입자 클러스터(cluster)는 다음의 방법으로 제조될 수 있으나, 이에 한정되지 않는다:In step 1), the polymer-magnetic nanoparticle cluster may be prepared by the following method, but is not limited thereto:
a) 고분자를 용해하는 단계;a) dissolving the polymer;
b) 자성나노입자를 적가하는 단계;b) adding magnetic nanoparticles dropwise;
c) 단계 a)의 고분자 및 단계 b)의 자성나노입자를 혼합하는 단계;c) mixing the polymer of step a) and the magnetic nanoparticles of step b);
d) 단계 c)의 혼합물을 초음파 처리한 뒤 실온에서 교반하는 단계.d) ultrasonicating the mixture of step c) and stirring at room temperature.
또한, 양전하로 표면 개질된 고분자-자성 나노입자 클러스터를 제조하기 위하여, 상기 단계 a)에 하기 단계 a’)를 추가할 수 있다:In addition, in order to prepare a polymer-magnetic nanoparticle cluster surface-modified with a positive charge, the following step a') may be added to the step a):
a’) 단계 a)에 양이온성 형질 전환체, 및 상기 양이온성 형질 전환체 및 고분자를 연결하는 링커를 추가 및 반응하여 양이온성 형질 전환체가 부착된 고분자를 획득하는 단계.a′) adding and reacting a cationic transformant and a linker connecting the cationic transformant and the polymer to step a) to obtain a polymer to which the cationic transformant is attached.
상기 링커는 구체적으로 N,N’-디사이클로헥실카르보디이미드(N,N′-Dicyclohexylcarbodiimide, DCC) 및 N-하이드록시석신이미드(N-Hydroxysuccinimide, NHS)일 수 있으나, 이에 한정되지 않는다. The linker may be specifically N,N'-dicyclohexylcarbodiimide (N,N'-Dicyclohexylcarbodiimide, DCC) and N-hydroxysuccinimide (N-Hydroxysuccinimide, NHS), but is not limited thereto.
또한, 상기 단계 c)에서 혼합은 3 내지 7분 동안 볼텍싱하여 수행될 수 있으나, 이에 한정되지 않는다.In addition, the mixing in step c) may be performed by vortexing for 3 to 7 minutes, but is not limited thereto.
또한, 형광 표지된 고분자-자성 나노입자 클러스터를 제조하기 위하여 상기 단계 c)에서 형광 부착된 고분자를 추가할 수 있다. In addition, a fluorescently attached polymer may be added in step c) to prepare a fluorescently-labeled polymer-magnetic nanoparticle cluster.
또한, 상기 단계 d)에서 초음파 처리는 구체적으로 1 내지 5분 동안 수행될 수 있고, 보다 구체적으로 2 내지 4 분 동안 수행될 수 있다. 상기 단계 d)에서 교반은 1 내지 24시간 동안 수행될 수 있고, 구체적으로 4 내지 8시간 동안 수행될 수 있으며, 보다 구체적으로 5 내지 7시간 동안 수행될 수 있다. In addition, the ultrasonic treatment in step d) may be specifically performed for 1 to 5 minutes, more specifically for 2 to 4 minutes. Stirring in step d) may be performed for 1 to 24 hours, specifically for 4 to 8 hours, and more specifically for 5 to 7 hours.
상기 방법으로 제조된 고분자-자성 나노입자 클러스터는 고분자와 자성 나노입자가 응집된 클러스터(cluster) 형태를 나타낸다.The polymer-magnetic nanoparticle cluster prepared by the above method has a cluster form in which a polymer and magnetic nanoparticles are aggregated.
또한, 상기 단계 1)에서 고분자-자성 나노입자 클러스터를 1 내지 1000 ㎍/㎖로 처리할 수 있으나, 이에 한정되지 않는다.In addition, the polymer-magnetic nanoparticle cluster may be treated at 1 to 1000 µg/ml in step 1), but is not limited thereto.
상기 줄기세포는 골수, 지방조직, 말초혈액, 간, 근육, 폐, 양수, 태반의 융모막 및 제대혈로 이루어진 군으로부터 선택된 1종 이상의 조직으로부터 유래되는 것일 수 있으나, 이에 한정되지 않는다.The stem cells may be derived from one or more tissues selected from the group consisting of bone marrow, adipose tissue, peripheral blood, liver, muscle, lung, amniotic fluid, placental chorion, and umbilical cord blood, but is not limited thereto.
또한, 상기 줄기세포는 배아 줄기세포 또는 성체 줄기세포일 수 있으나, 이에 한정되지 않는다.In addition, the stem cells may be embryonic stem cells or adult stem cells, but are not limited thereto.
상기 성체 줄기세포는 중간엽줄기세포, 인간 조직 유래 중간엽 기질세포, 인간 조직 유래 중간엽줄기세포 및 다분화능 줄기세포로 구성된 군에서 선택된 1종 이상의 성체 줄기세포인 것일 수 있으나, 이에 한정되지 않는다.The adult stem cells may be one or more adult stem cells selected from the group consisting of mesenchymal stem cells, human tissue-derived mesenchymal stromal cells, human tissue-derived mesenchymal stem cells, and multipotent stem cells, but are not limited thereto. .
상기 단계 2)에서 자기력을 0.1, 0.2, 0.3, 0.4, 0.6 또는 1.0 T 이상으로 가할 수 있고, 구체적으로 0.1 내지 2 T로 가할 수 있으며, 보다 구체적으로 0.3 내지 1T로 가할 수 있으나, 이에 한정되지 않는다.In step 2), the magnetic force may be applied in an amount of 0.1, 0.2, 0.3, 0.4, 0.6, or 1.0 T or more, specifically 0.1 to 2 T, and more specifically 0.3 to 1 T, but is not limited thereto. Does not.
또한, 본 발명은 1) 고분자-자성 나노입자 클러스터(cluster)를 줄기세포에 처리하는 단계; In addition, the present invention comprises the steps of: 1) treating a polymer-magnetic nanoparticle cluster on stem cells;
2) 상기 단계 1)의 줄기세포에 자기력을 가하는 단계; 및2) applying a magnetic force to the stem cells of step 1); And
3) 상기 단계 2)의 줄기세포로부터 엑소좀을 분리하는 단계;를 포함하는 엑소좀의 제조방법 및 상기 제조방법에 따라 제조된 엑소좀을 제공한다.3) separating the exosomes from the stem cells of step 2); and a method for preparing an exosome including, and an exosome prepared according to the preparation method.
또한, 본 발명은 고분자-자성 나노입자 클러스터(cluster)를 포함하는 줄기세포 유래 엑소좀 생성 촉진용 조성물을 제공한다.In addition, the present invention provides a composition for promoting the generation of exosomes derived from stem cells comprising a polymer-magnetic nanoparticle cluster (cluster).
상기 조성물을 배양하는데 이용되는 배지로는, 줄기세포의 배양에 이용되는 일반적인 어떠한 배지도 이용할 수 있다. As a medium used for culturing the composition, any general medium used for culturing stem cells may be used.
바람직하게는, 혈청(예컨대, 소태아 혈청, 말 혈청 및 인간 혈청)이 함유된 배지이며, 본 발명에서 이용될 수 있는 배지는, 예를 들어, DMEM (Dulbecco's Modified Eagle's Medium), MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10), DMEM/F-12 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12), α-MEM(α-Minimal essential Medium), G-MEM(Glasgow's Minimal Essential Medium), IMDM(Isocove's Modified Dulbecco's Medium), 및 KnockOut DMEM일 수 있고, 더욱 바람직하게는 최소필수영양배지(minimum essential medium, MEM)일 수 있으나, 이에 한정되지 않는다. Preferably, it is a medium containing serum (e.g., fetal bovine serum, horse serum, and human serum), and the medium that can be used in the present invention is, for example, DMEM (Dulbecco's Modified Eagle's Medium), MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10), DMEM/F-12 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12), α -MEM (α-Minimal Essential Medium), G-MEM (Glasgow's Minimal Essential Medium), IMDM (Isocove's Modified Dulbecco's Medium), and may be KnockOut DMEM, more preferably a minimum essential nutrient medium (minimum essential medium, MEM) It may be, but is not limited thereto.
또한, 상기 배지에는 다른 성분, 예를 들어 항생제 또는 항진균제(예컨대, 페니실린, 스트렙토마이신) 및 글루타민 등이 포함될 수 있다.In addition, the medium may contain other components, such as antibiotics or antifungal agents (eg, penicillin, streptomycin), glutamine, and the like.
본 발명의 구체적인 실시예에서, 본 발명자들은 고분자-자성 나노입자 클러스터 및 자성자극에 의해 중간엽줄기세포 유래 엑소좀 생성이 촉진되는 것을 확인하였다.In a specific embodiment of the present invention, the present inventors confirmed that the generation of exosomes derived from mesenchymal stem cells is promoted by polymer-magnetic nanoparticle clusters and magnetic stimulation.
보다 구체적으로, 본 발명자들은 고분자-자성 나노입자 클러스터(cluster)를 줄기세포에 처리하고 상기 줄기세포에 자기력을 가했을 때, 줄기세포 유래 엑소좀의 생성이 촉진되는 것을 확인하였으므로, 상기 엑소좀의 발생을 극대화시키는 방법을 유용하게 이용할 수 있다.More specifically, the present inventors confirmed that when a macromolecular-magnetic nanoparticle cluster was treated on stem cells and a magnetic force was applied to the stem cells, the generation of stem cell-derived exosomes was promoted, so that the generation of the exosomes The method of maximizing the value can be usefully used.
이하 본 발명을 실시예, 실험예에 의해 상세히 설명한다.Hereinafter, the present invention will be described in detail by examples and experimental examples.
단, 하기 실시예 및 실험예는 본 발명을 예시하는 것일 뿐, 본 발명의 내용이 하기 실시예 및 실험예에 한정되는 것은 아니다.However, the following Examples and Experimental Examples are merely illustrative of the present invention, and the contents of the present invention are not limited to the following Examples and Experimental Examples.
<실시예 1> 세포배양<Example 1> Cell culture
중간엽줄기세포(mesenchymal stem cell, MSC)는 Cyagen사(MUBMX-01001, US)로부터 구입하였다. 중간엽줄기세포를 10% 소태아혈청(fetal bovine serum, FBS)(Gibco, US) 및 항생제(antibiotics)(Antibiotic-Antimycotic, US)가 포함된 MEM-α 배지(Hyclone, US)에서 배양하였다. 상기 배양한 중간엽줄기세포를 DPBS(Dulbecco’s Phosphate-Buffered Saline)로 세척한 후 3일마다 배지를 교체하였고, 37℃, 5 % CO2 로 유지하였다.Mesenchymal stem cells (MSC) were purchased from Cyagen (MUBMX-01001, US). Mesenchymal stem cells were cultured in MEM-α medium (Hyclone, US) containing 10% fetal bovine serum (FBS) (Gibco, US) and antibiotics (Antibiotic-Antimycotic, US). After washing the cultured mesenchymal stem cells with DPBS (Dulbecco's Phosphate-Buffered Saline), the medium was changed every 3 days, and maintained at 37°C and 5% CO 2.
<실시예 2> 음전하 표면개질 중합 클러스터드 초상자성 산화철(Polymeric clustered superparamagnetic iron oxide, PCS) 나노입자 합성<Example 2> Synthesis of negatively charged surface-modified polymerized clustered superparamagnetic iron oxide (PCS) nanoparticles
중합 클러스터드 초상자성 산화철(Polymeric clustered superparamagnetic iron oxide, PCS) 나노입자는 산화철 기반 나노입자로 이루어졌으며, 나노입자의 크기는 약 100 ㎚이다. PCS 나노입자 처리 조건을 결정하기 위하여, 나노입자의 농도를 24시간 동안 20, 40 및 60 ㎍/㎖로 제조하였다.Polymerized clustered superparamagnetic iron oxide (PCS) nanoparticles were made of iron oxide-based nanoparticles, and the size of the nanoparticles was about 100 nm. In order to determine the PCS nanoparticle treatment conditions, the concentrations of the nanoparticles were prepared at 20, 40 and 60 μg/ml for 24 hours.
음전하 표면개질 PCS 나노입자를 합성하기 위하여, 폴리(D, L-락타이드-코-글리코라이드)(Poly(D, L-lactide-co-glycolide, PLGA))(50:50, MW 38,000 내지 54,000)(Sigma-Aldrich, US), 초상자성 산화철 나노입자(Superparamagnetic iron oxide(SPIO) nanoparticles, SPIONs)(Sigma-Aldrich, US)를 사용하였고, 1-에틸-3-(3-디메틸아미노프로필) 카르보디미드 히드로클로라이드(1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride, EDC)(Thermo-fisher Scientigic, US) 및 N-하이드록시석신이미드(N-hydroxysuccinimide, NHS)를 형광 분자(fluorescence molecule) 사이의 링커(EDC-NHS)로 제조하기 위해 사용하였다. 또한, 세포 내의 나노입자를 검출하기 위해 형광 분자로 시아닌5.5(Cyanine5.5, Cy5.5)(Cyanine5.5 amine, Lumiprobe Co., US)을 사용하였다.To synthesize negatively charged surface-modified PCS nanoparticles, poly(D, L-lactide-co-glycolide) (Poly(D, L-lactide-co-glycolide, PLGA)) (50:50, M W 38,000 to 54,000) (Sigma-Aldrich, US), superparamagnetic iron oxide (SPIO) nanoparticles, SPIONs) (Sigma-Aldrich, US) were used, and 1-ethyl-3-(3-dimethylaminopropyl) Carbodiimide hydrochloride (1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride, EDC) (Thermo-fisher Scientigic, US) and N-hydroxysuccinimide (NHS) were used as fluorescent molecules (fluorescence). molecule) between the linker (EDC-NHS). In addition, cyanine 5.5 (Cyanine5.5, Cy5.5) (Cyanine5.5 amine, Lumiprobe Co., US) was used as a fluorescent molecule to detect nanoparticles in cells.
구체적으로, 330 ㎎의 PLGA를 60 ㎎의 EDC, 132 ㎎의 NHS 및 9.9 ㎎의 Cy5.5와 함께 DMSO 용액에 용해시키고 24시간 동안 배양하여 PLGA와 Cy5.5가 EDC-NHS에 의해 결합되도록 한 후, 반응 혼합물을 탈 이온수(deionized water)로 투석(Mw cutoff=10 k)하여 과량의 EDC, NHS 및 Cy5.5를 제거하여 PLGA-Cy5.5 형광 중합체(PLGA-Cy5.5)를 획득하였다.Specifically, 330 mg of PLGA was dissolved in DMSO solution with 60 mg of EDC, 132 mg of NHS and 9.9 mg of Cy5.5 and incubated for 24 hours so that PLGA and Cy5.5 were bound by EDC-NHS. Then, the reaction mixture was dialyzed with deionized water (M w cutoff = 10 k) to remove excess EDC, NHS and Cy5.5 to obtain a PLGA-Cy5.5 fluorescent polymer (PLGA-Cy5.5). I did.
그 다음, 0.4 ㎎의 PLGA-Cy5.5 및 0.1 ㎎의 PLGA를 0.1 ㎖의 DMSO에 용해시키고, 3 ㎖의 탈 이온수에 0.1 ㎖의 SPIONs(5 ㎎/㎖)를 적가(drop wise)하였다. 이후 바이알(Vial)을 5분 동안 볼텍싱하고, 3분 동안 초음파 처리한 뒤 6시간 동안 실온에서 교반하여 음전하 표면개질 PCS 나노입자를 획득하였다. 상기 획득한 음전하 표면 개질 PCS 나노입자를 동결 건조하여 분말 시료를 얻었다(도 2, 위).Then, 0.4 mg of PLGA-Cy5.5 and 0.1 mg of PLGA were dissolved in 0.1 ml of DMSO, and 0.1 ml of SPIONs (5 mg/ml) was added dropwise to 3 ml of deionized water. Thereafter, the vial was vortexed for 5 minutes, sonicated for 3 minutes, and stirred at room temperature for 6 hours to obtain negatively charged surface-modified PCS nanoparticles. The obtained negatively charged surface-modified PCS nanoparticles were freeze-dried to obtain a powder sample (Fig. 2, above).
<실시예 3> 양전하 표면개질 중합 클러스터드 초상자성 산화철(Polymeric clustered superparamagnetic iron oxide, PCS) 나노입자 합성<Example 3> Synthesis of positively charged surface-modified polymerized clustered superparamagnetic iron oxide (PCS) nanoparticles
양전하 표면개질 PCS 나노입자를 하기와 같이 제조하였다. PCS 나노입자 처리 조건을 결정하기 위하여, 나노입자의 농도를 24시간 동안 20, 40 및 60 ㎍/㎖로 제조하였다.Positively charged surface-modified PCS nanoparticles were prepared as follows. In order to determine the PCS nanoparticle treatment conditions, the concentrations of the nanoparticles were prepared at 20, 40 and 60 μg/ml for 24 hours.
양전하 표면개질 PCS 나노입자를 합성하기 위하여, 폴리(D, L-락타이드-코-글리코라이드)(Poly(D, L-lactide-co-glycolide, PLGA))(50:50, MW 38,000~54,000)(Sigma-Aldrich, US) 및 폴리에틸렌이민(Polyethyleneimine, PEI) (MW ~25,000 by LS, Mn ~ 10,000 by GPC)(Sigma-Aldrich, US)을 사용하였고, N,N’-디사이클로헥실카르보디이미드(N,N’-Dicyclohexylcarbodiimide, DCC) 및 N-하이드록시석신이미드(N-Hydroxysuccinimide, NHS)를 PLGA와 PEI 사이의 링커로 제조하기 위해 사용하였다.To synthesize positively charged surface-modified PCS nanoparticles, poly(D, L-lactide-co-glycolide) (Poly(D, L-lactide-co-glycolide, PLGA))(50:50, M W 38,000~ 54,000) (Sigma-Aldrich, US) and polyethyleneimine (PEI) (M W ~25,000 by LS, M n ~ 10,000 by GPC) (Sigma-Aldrich, US) was used, and N,N'-dicyclo Hexylcarbodiimide (N,N'-Dicyclohexylcarbodiimide, DCC) and N-Hydroxysuccinimide (NHS) were used to prepare a linker between PLGA and PEI.
PLGA-PEI 공중합체를 합성하기 위하여, 330 ㎎의 PLGA, 113.67 ㎎의 PEI, 18.33 ㎎의 DCC 및 11 ㎎의 NHS를 100 ㎖의 DMSO 용액에 용해시키고 24시간 동안 배양한 후, 반응 혼합물을 탈 이온수로 투석(Mw cutoff=10 k)하여 과량의 DCC, NHS 및 PEI를 제거하였다. 얻어진 PLGA-PEI 공중합체 욕액을 동결 건조하여 분말 시료를 얻었다.To synthesize the PLGA-PEI copolymer, 330 mg of PLGA, 113.67 mg of PEI, 18.33 mg of DCC and 11 mg of NHS were dissolved in 100 ml of DMSO solution and incubated for 24 hours, and the reaction mixture was then used in deionized water. Dialysis (M w cutoff = 10 k) was performed to remove excess DCC, NHS and PEI. The obtained PLGA-PEI copolymer bath liquid was freeze-dried to obtain a powder sample.
PLGA-Cy5.5 형광 중합체는 상기 <실시예 2>에 기재된 방법과 동일한 방법으로 합성하였다.The PLGA-Cy5.5 fluorescent polymer was synthesized in the same manner as described in <Example 2>.
그 다음, PLGA-PEI 공중합체, PLGA-Cy5.5 및 SPIONs가 사용되었다. 0.4 ㎎의 PLGA-Cy5.5 및 0.1 ㎎의 PLGA-PEI를 0.1 ㎖의 DMSO에 용해시키고, 3 ㎖의 탈 이온수에 0.1 ㎖의 SPIONs(5 ㎎/㎖)를 적가(drop wise)하였다. 이후 바이알(Vial)을 5분 동안 볼텍싱하고, 3분 동안 초음파 처리한뒤 6시간 동안 실온에서 교반하여 양전하 표면개질 PCS 나노입자를 획득하였다. 상기 획득한 양전하 표면 개질 PCS 나노입자를 동결 건조하여 분말 시료를 얻었다(도 2, 아래).Then, PLGA-PEI copolymer, PLGA-Cy5.5 and SPIONs were used. 0.4 mg of PLGA-Cy5.5 and 0.1 mg of PLGA-PEI were dissolved in 0.1 ml of DMSO, and 0.1 ml of SPIONs (5 mg/ml) was added dropwise to 3 ml of deionized water. Thereafter, the vial was vortexed for 5 minutes, sonicated for 3 minutes, and stirred at room temperature for 6 hours to obtain positively charged surface-modified PCS nanoparticles. The obtained positively charged surface-modified PCS nanoparticles were freeze-dried to obtain a powder sample (Fig. 2, bottom).
<실시예 4> 미세자성(micro-magnetofection)을 가하여 PCS 나노입자 처리한 중간엽줄기세포로부터 엑소좀 분리<Example 4> Separation of exosomes from mesenchymal stem cells treated with PCS nanoparticles by applying micro-magnetofection
상기 <실시예 2 및 3>에서 제조한 PCS 나노입자를 상기 <실시예 1>의 중간엽줄기세포에 처리하고 자기력을 가한 후, 엑소좀을 분리하였다. After the PCS nanoparticles prepared in <Examples 2 and 3> were treated to the mesenchymal stem cells of <Example 1> and magnetic force was applied, exosomes were separated.
구체적으로, 상기 <실시예 1>의 중간엽줄기세포 2×10^6 개를 세포 배양 플레이트에서 배양하였다. 그다음, 상기 <실시예 2 및 3>에서 제조한 PCS 나노입자를 상기 배양한 중간엽줄기세포에 24시간 동안 처리한 후, 자기력을 24시간 동안 가하였다. 이후, 세포배양 배지(cell culture media)를 회수하여 2,000 xg의 원심력으로 30분 동안 원심분리한 후, 상층액을 새로운 튜브로 옮겼다. 새로운 튜브로 옮긴 상층액에 0.5 볼륨(volume)의 엑소좀 분리 시약(cat.4478359, Thermo-fisher Scientigic, US)을 첨가하고, 볼텍싱(vortexing) 또는 피펫팅(pipetting)으로 혼합하였다. 상기 혼합물을 6℃에서 밤새 배양한 후, 10,000 xg의 원심력으로 1시간 동안 6℃에서 원심분리하고, 상층액을 버리고 튜브의 바닥에 가라앉은 펠렛(pellet)을 회수하였다. 이후 회수한 펠렛은 1X PBS에 재현탁(re-suspended)하고, -20℃에서 장기간 보관하였다.Specifically, 2×10^6 mesenchymal stem cells of <Example 1> were cultured in a cell culture plate. Then, the PCS nanoparticles prepared in <Examples 2 and 3> were treated with the cultured mesenchymal stem cells for 24 hours, and then magnetic force was applied for 24 hours. Thereafter, the cell culture media was recovered and centrifuged for 30 minutes with a centrifugal force of 2,000 xg, and the supernatant was transferred to a new tube. 0.5 volume of exosome separation reagent (cat.4478359, Thermo-fisher Scientigic, US) was added to the supernatant transferred to a new tube, and mixed by vortexing or pipetting. The mixture was incubated at 6° C. overnight, and then centrifuged at 6° C. for 1 hour with a centrifugal force of 10,000 xg, and the supernatant was discarded and a pellet settled on the bottom of the tube was recovered. Subsequently, the recovered pellet was re-suspended in 1X PBS and stored at -20°C for a long period of time.
<실시예 5> 투과 전자 현미경(Transmission Electron Microscopy, TEM)<Example 5> Transmission Electron Microscopy (TEM)
세포를 4℃에서 2시간 동안 2.5 %의 글루타르알데하이드(glutaraldehyde)에 고정시키고, 2 % 한천(agar)으로 굳혔다. 이후 0.1 M의 카코딜레이트 완충액(cacodylate buffer)으로 세척한 뒤, 1 %의 오스뮴 테트록사이드(osmium tetroxide, OsO4)에 고정시켰다. 탈수 단계는 50 내지 100 %의 에탄올을 사용하여 실시되고, 에폰 레진(Epon resin)에 내장되었다. 초박절편(ultrathin sections)은 초마이크로톰(ultramicrotome)을 사용하여 절단하고, 우라닐 아세테이트(uranyle acetate) 및 납 시트레이트(lead citrate)를 사용하여 염색하였다. 그다음, 63 kV 조건에서 TEM을 사용하여 관찰하였다.The cells were fixed in 2.5% glutaraldehyde for 2 hours at 4°C, and solidified with 2% agar. After washing with 0.1 M cacodylate buffer, it was fixed in 1% osmium tetroxide (OsO 4 ). The dehydration step was carried out using 50-100% ethanol and embedded in Epon resin. Ultrathin sections were cut using an ultramicrotome, and stained using uranyl acetate and lead citrate. Then, it was observed using TEM under the condition of 63 kV.
<실시예 6> 웨스턴 블롯(Western blot)<Example 6> Western blot
상기 <실시예 4>에서 엑소좀을 제조한 후, 엑소좀의 특이적 마커(specific markers)를 엑소좀 단백질에 의해 측정하였다. CD9(ab92726, Abcam, US), CD63(ab216130, Abcam, US) 및 CD81(ab109201, Abcam, US)에 대한 1차 항체(primary antibodies)가 1:1000으로 사용되었고, 염소 항-토끼 면역글로불린 G(Goat anti-rabbit immunoglobulin G, Goat anti-rabbit IgG)는 2차 항체(secondary antibody)로 사용되었다. SPSS(Statistical Package for the Social Science)(SPSS Inc., US)를 사용하여 엑소좀 단백질의 발현 수준을 측정하고 표준화하였다.After preparing exosomes in <Example 4>, specific markers of exosomes were measured by exosome proteins. Primary antibodies against CD9 (ab92726, Abcam, US), CD63 (ab216130, Abcam, US) and CD81 (ab109201, Abcam, US) were used at 1:1000, goat anti-rabbit immunoglobulin G (Goat anti-rabbit immunoglobulin G, Goat anti-rabbit IgG) was used as a secondary antibody. The expression level of exosome proteins was measured and normalized using SPSS (Statistical Package for the Social Science) (SPSS Inc., US).
<실시예 7> 통계 분석<Example 7> Statistical analysis
SPSS 통계 패키지 버전 21.0(SPSS statistical package version 21.0, SPSS Inc., US)을 사용하여 통계 분석을 수행하였다. 연속 변수(continuous variables)의 기술적 결과(descriptive results)는 정규 분포 변수(normally distributed variables)의 평균(mean) ± 표준 편차(standard deviation, SD)로 표현하였다. 평균은 이원분산분석(two-way analysis of variance)을 이용해 비교하였고, 통계적 유의 수준은 0.05이다.Statistical analysis was performed using SPSS statistical package version 21.0 (SPSS Inc., US). Descriptive results of continuous variables were expressed as mean ± standard deviation (SD) of normally distributed variables. Means were compared using a two-way analysis of variance, and the statistical significance level was 0.05.
<실험예 1> PCS 나노입자의 특성 확인<Experimental Example 1> Confirmation of the properties of PCS nanoparticles
상기 <실시예 3>에 따라 제조한 양전하로 표면 개질된 PCS 나노입자의 크기 및 표면 전하를 분석하였다.The size and surface charge of the surface-modified PCS nanoparticles prepared according to <Example 3> were analyzed.
구체적으로, 상기 <실시예 3>에 따라 제조한 양전하로 표면 개질된 PCS 나노입자의 크기 및 표면 전하를 DLS(dynamic light scattering) Zetasizer Nano-ZS90(Malvern Instruments Ltd., UK)로 측정하였다.Specifically, the size and surface charge of the PCS nanoparticles surface-modified with positive charges prepared according to the <Example 3> were measured by DLS (dynamic light scattering) Zetasizer Nano-ZS90 (Malvern Instruments Ltd., UK).
또한, 상기 PCS 나노입자를 카보닐-코팅된 400 메시 구리 그리드(CF400-CU-UL, Electron Microscopy Sciences, US)에 15분 동안 놓았다. 그다음, 2 % 우라닐 아세테이트 10 ㎕를 상기 그리드 상에 10분간 둔 후, 건조하여 음성 염색하였다. 그다음, 전자현미경(JEM-2100F, JEOL Ltd, Japan)을 이용하여 63 kV 조건에서 TEM(transmission electron microscopic) 이미지를 획득하여 나노입자의 구조를 확인하였다. 또한, 전자현미경을 이용하여 SEM(scanning electron microscopic) 이미지를 획득하여 나노입자의 형태를 확인하였다.In addition, the PCS nanoparticles were placed on a carbonyl-coated 400 mesh copper grid (CF400-CU-UL, Electron Microscopy Sciences, US) for 15 minutes. Then, 10 µl of 2% uranyl acetate was placed on the grid for 10 minutes, dried and negatively stained. Then, using an electron microscope (JEM-2100F, JEOL Ltd, Japan), a TEM (transmission electron microscopic) image was obtained under the condition of 63 kV to confirm the structure of the nanoparticles. In addition, a scanning electron microscopic (SEM) image was obtained using an electron microscope to confirm the shape of the nanoparticles.
그 결과, 도 3a 내지 도 3d에 나타낸 바와 같이, 양전하로 표면 개질된 PCS 나노입자가 도 2에 나타낸 바와 같이 PLGA와 산화철 나노입자가 응집된 클러스터(cluster) 형태를 나타내고, 양전하를 띠고 있음을 확인하였다. 이에 상기 PCS 나노입자를 고분자-자성 나노입자 클러스터로 명명하였고, 상기 양전하로 표면 개질된 PCS 나노입자는 양전하로 표면 개질된 고분자-자성 나노입자 클러스터로, 음전하로 표면 개질된 PCS 나노입자는 음전하로 표면 개질된 고분자-자성 나노입자 클러스터로 명명하였다.As a result, as shown in FIGS. 3A to 3D, it was confirmed that the PCS nanoparticles surface-modified with a positive charge exhibited a cluster form in which PLGA and iron oxide nanoparticles were aggregated as shown in FIG. 2, and have a positive charge. I did. Accordingly, the PCS nanoparticles were named as polymer-magnetic nanoparticle clusters, and the PCS nanoparticles surface-modified with positive charges were polymer-magnetic nanoparticle clusters surface-modified with positive charges, and the PCS nanoparticles surface-modified with negative charges were negatively charged. It was named as a surface-modified polymer-magnetic nanoparticle cluster.
<실험예 2> 고분자-자성 나노입자 클러스터의 표면 전하 및 농도에 따른 엑소좀의 생성 정도 확인<Experimental Example 2> Confirmation of the degree of generation of exosomes according to the surface charge and concentration of the polymer-magnetic nanoparticle cluster
고분자-자성 나노입자 클러스터의 표면 전하 및 농도에 따른 엑소좀의 생성 정도를 확인하기 위해, 상기 <실시예 2 및 3>에서 제조한 양전하/음전하로 표면 개질된 고분자-자성 나노입자 클러스터를 농도를 달리하여 중간엽줄기세포에 처리하고 미세자성을 가한 후 중간엽줄기세포에서 생성되는 엑소좀의 생성량을 분석하였다.In order to confirm the degree of generation of exosomes according to the surface charge and concentration of the polymer-magnetic nanoparticle cluster, the concentration of the polymer-magnetic nanoparticle cluster surface-modified with positive/negative charges prepared in <Examples 2 and 3> above. In different ways, the mesenchymal stem cells were treated and micromagnetic was applied, and then the amount of exosomes produced in the mesenchymal stem cells was analyzed.
구체적으로, 상기 <실시예 4>에 기재된 방법과 동일한 방법으로 상기 <실시예 3>에서 제조한 양전하로 표면 개질된 고분자-자성 나노입자 클러스터를 40 ㎍/㎖ 농도로 중간엽줄기세포에 처리하고, 0.3 T, 0.4 T, 0.6 T 및 1.0 T 세기의 자기력을 가하였다. 그다음, 상기 <실시예 5>에 기재된 방법과 동일한 방법으로 TEM 관찰을 통해 중간엽줄기세포로부터 엑소좀의 생성량을 분석하였다.Specifically, the polymer-magnetic nanoparticle cluster surface-modified with a positive charge prepared in <Example 3> was treated on the mesenchymal stem cells at a concentration of 40 μg/ml in the same manner as in the method described in <Example 4>. , 0.3 T, 0.4 T, 0.6 T and 1.0 T magnetic force was applied. Then, the amount of exosomes produced from mesenchymal stem cells was analyzed through TEM observation in the same manner as described in <Example 5>.
또한, 상기에 기재된 방법과 동일한 방법으로 상기 <실시예 3>에서 제조한 양전하로 표면 개질된 고분자-자성 나노입자 클러스터를 20, 40 및 60 ㎍/㎖ 농도로 처리하고 자기력을 가한 후 중간엽줄기세포로부터 엑소좀의 생성량을 분석하였다.In addition, in the same method as described above, the polymer-magnetic nanoparticle clusters surface-modified with positive charges prepared in <Example 3> were treated at concentrations of 20, 40, and 60 ㎍/mL, and after applying magnetic force, mesenchymal stem The amount of exosomes produced from cells was analyzed.
대조군으로 고분자-자성 나노입자 클러스터를 무처리한 중간엽줄기세포를 이용하였다.As a control, mesenchymal stem cells without treatment of polymer-magnetic nanoparticle clusters were used.
그 결과, 도 4 및 도 5에서 나타낸 바와 같이, 음전하로 표면 개질된 고분자-자성 나노입자 클러스터를 처리하였을 때는 엑소좀이 발생되지 않았지만, 양전하로 표면 개질된 고분자-자성 나노입자 클러스터를 처리하였을 때는 엑소좀의 발생이 유도되었고, 양전하로 표면 개질된 고분자-자성 나노입자 클러스터의 농도가 높아질수록 그 양 또한 증가되는 것을 관찰할 수 있었다(도 4). 특히, 양전하로 표면 개질된 고분자-자성 나노입자 클러스터를 처리한 경우 자기력에 의해 단시간에 많은 양의 나노입자 클러스터가 유입되고, 엔도시토시스(endocytosis)에 의해 유입된 나노입자 클러스터는 엑소좀을 거쳐 리소좀으로 이동하는 것을 확인하였다. 또한, 리소좀 내에서 나노입자 클러스터는 그대로 있고, 엑소좀의 생성량만 증가하며, 생성되는 엑소좀은 91 내지 169 ㎚의 크기를 가지고 있는 것으로 나타났다(도 5).As a result, as shown in FIGS. 4 and 5, when the polymer-magnetic nanoparticle clusters surface-modified with negative charges were treated, exosomes were not generated, but when the polymer-magnetic nanoparticle clusters surface-modified with positive charges were treated. The generation of exosomes was induced, and as the concentration of the polymer-magnetic nanoparticle clusters surface-modified with positive charges increased, the amount was also observed to increase (FIG. 4). In particular, when the polymer-magnetic nanoparticle clusters surface-modified with positive charges are treated, a large amount of nanoparticle clusters are introduced in a short time by magnetic force, and the nanoparticle clusters introduced by endocytosis pass through exosomes. It was confirmed that it migrates to the lysosome. In addition, the nanoparticle cluster in the lysosome remains as it is, only the amount of exosomes produced increases, and the resulting exosomes were found to have a size of 91 to 169 nm (FIG. 5).
<실험예 3> 자기력 세기에 따른 엑소좀의 생성 정도 확인<Experimental Example 3> Confirmation of the degree of generation of exosomes according to magnetic force strength
자기력 세기에 따른 엑소좀의 생성 정도를 확인하기 위해, 상기 <실시예 3>에서 제조한 양전하로 표면 개질된 고분자-자성 나노입자 클러스터를 중간엽줄기세포에 처리하고 미세자성을 세기를 달리하여 가한 후 중간엽줄기세포에서 생성되는 엑소좀의 생성량을 분석하였다.In order to confirm the degree of generation of exosomes according to the strength of magnetic force, the polymer-magnetic nanoparticle cluster surface-modified with the positive charge prepared in <Example 3> was treated to the mesenchymal stem cells, and micromagnets were applied by varying the strength. Afterwards, the amount of exosomes produced in mesenchymal stem cells was analyzed.
구체적으로, 상기 <실험예 3>에 기재된 방법과 동일한 방법으로 상기 <실시예 3>에서 제조한 양전하로 표면 개질된 고분자-자성 나노입자 클러스터를 20, 40 및 60 ㎍/㎖ 농도로 처리하고 0.3 T, 0.4 T, 0.6 T 및 1.0 T 세기의 자기력을 가한 후 상기 <실시예 5>에 기재된 방법과 동일한 방법으로 웨스턴 블럿을 수행하여 엑소좀 단백질 발현량을 측정하였다.Specifically, in the same method as described in <Experimental Example 3>, the polymer-magnetic nanoparticle clusters surface-modified with positive charges prepared in <Example 3> were treated at concentrations of 20, 40 and 60 µg/ml, and 0.3 After applying magnetic force of T, 0.4 T, 0.6 T, and 1.0 T intensity, Western blotting was performed in the same manner as described in <Example 5> to measure the expression level of exosome proteins.
그 결과, 도 6에 나타낸 바와 같이, 자기력에 노출되는 동안 엑소좀 특이적 마커인 CD9, CD63 및 CD81의 발현이 모두 증가된 수준으로 나타나고, 가하는 자기력 세기가 강할수록 발현 또한 증가되는 것을 관찰할 수 있었다(도 6).As a result, as shown in FIG. 6, it can be observed that the expression of all of the exosome-specific markers CD9, CD63, and CD81 during exposure to magnetic force increased at increased levels, and the expression also increased as the strength of the applied magnetic force increased. There was (Fig. 6).
상기 결과들을 통해 나노입자 클러스터의 표면 개질이 제타-전위에 의해 차이를 일으키며, 음성(negative)의 성질보다는 양성(positive)의 성질을 가질수록 더 많은 양의 나노입자 클러스터가 줄기세포 내부로 유입되며, 이에 따라 자기력을 가했을 때 엑소좀의 생성량이 증가함을 알 수 있다.Through the above results, the surface modification of the nanoparticle cluster causes a difference due to the zeta-potential, and the more the nanoparticle cluster has a positive property rather than a negative property, the greater the amount of the nanoparticle cluster flows into the stem cells. As a result, it can be seen that the amount of exosomes produced increases when magnetic force is applied.
본 발명은 자성 나노입자 클러스터를 이용한 중간엽줄기세포 유래 엑소좀 생성 촉진 방법에 관한 것으로, 구체적으로, 고분자-자성 나노입자 클러스터(cluster)를 줄기세포에 처리하고 상기 줄기세포에 자기력을 가했을 때, 줄기세포 유래 엑소좀의 생성을 촉진시키므로, 생체외(in vitro) 및 생체내(in vivo)에서 임상적 치료효능이 있는 양질의 엑소좀을 다량으로 제공하는데 유용하게 이용할 수 있다.The present invention relates to a method for promoting generation of exosomes derived from mesenchymal stem cells using magnetic nanoparticle clusters, and specifically, when a polymer-magnetic nanoparticle cluster is treated on stem cells and magnetic force is applied to the stem cells, Since it promotes the generation of stem cell-derived exosomes, it can be usefully used to provide a large amount of high-quality exosomes with clinical therapeutic efficacy both in vitro and in vivo.

Claims (17)

1) 고분자-자성 나노입자 클러스터(cluster)를 줄기세포에 처리하는 단계; 및1) treating a macromolecular-magnetic nanoparticle cluster on stem cells; And
2) 상기 단계 1)의 줄기세포에 자기력을 가하는 단계;를 포함하는 줄기세포 유래 엑소좀의 생성 촉진 방법.2) The step of applying a magnetic force to the stem cells of step 1); Stem cell-derived exosome production promotion method comprising a.
제1항에 있어서, The method of claim 1,
상기 단계 1)의 고분자는 생체적합성 고분자인 것을 특징으로 하는, 방법.The method, characterized in that the polymer of step 1) is a biocompatible polymer.
제2항에 있어서,The method of claim 2,
상기 생체적합성 고분자는,The biocompatible polymer,
폴리(베타-히드록시 에틸 메타아크릴레이트)(Poly(beta-hydroxyethyl Methacrylate), PHEMA); 폴리아크릴아미드(Polyacrylamide, PA); 폴리비닐 알코올(Polyvinyl Alcohol, PVA); 폴리아크릴산(Polyacrylic Acid, PAA), 그 염 및 그 유도체; 폴리메타아크릴산(Poly(metha acrylic Acid), PMAA) 또는 그 유도체; 폴리아크릴아미드(Poly(acrylic amide)) 또는 그 유도체; 폴리언데세노산(Poly(undecenoic acid)) 또는 그 유도체, 또는 상기 고분자의 공중합체; 덱스트란(Dextran) 또는 그 유도체, 폴리비닐피롤리돈(Polyvinylpyrrolidone, PVP); 폴리에틸렌 옥사이드(Polyethyleneoxide, PEO); 폴리에틸렌글리콜(Polyethyleneglycol, PEG) 또는 그 유도체; 폴리프로필렌글리콜(Polypropylene Glycol, PPG) 또는 그 유도체; 상기 폴리에틸렌글리콜 및 폴리프로필렌글리콜의 공중합체, 또는 이들의 모노에스테르화 유도체; 폴리(에틸렌옥시드-b-프로필렌 옥사이드(Poly(ethylene oxide-b-propylene oxide), PEO-PPO); 폴리-l-라이신(Polyl-l-lysine); 폴리에틸렌이민(Polyethylenimine); 폴리락트산(Poly lactic acid, PLA); 폴리글리콜산(Polyglycolic acid, PGA); 폴리(락트산-co-글리콜산)(Poly(lactic- co -glycolic acid), PLGA) 공중합체; 키토산(Chitosan); 히알루론산(Hyaluronic acid); 및 이들의 혼합물로 이루어진 군으로부터 선택된 1종 이상인 것을 특징으로 하는, 방법.Poly(beta-hydroxyethyl methacrylate) (PHEMA); Polyacrylamide (PA); Polyvinyl alcohol (PVA); Polyacrylic acid (PAA), its salt and its derivative; Polymethacrylic acid (Poly (metha acrylic acid), PMAA) or a derivative thereof; Poly(acrylic amide) or a derivative thereof; Poly(undecenoic acid) or a derivative thereof, or a copolymer of the polymer; Dextran or a derivative thereof, polyvinylpyrrolidone (PVP); Polyethylene oxide (PEO); Polyethyleneglycol (PEG) or a derivative thereof; Polypropylene Glycol (PPG) or a derivative thereof; A copolymer of the polyethylene glycol and polypropylene glycol, or a monoesterified derivative thereof; Poly(ethylene oxide-b-propylene oxide, PEO-PPO); Polyl-l-lysine; Polyethylenimine; Polylactic acid lactic acid, PLA); Polyglycolic acid (PGA); Poly(lactic-co-glycolic acid) (PLGA) copolymer; Chitosan; Hyaluronic acid (Hyaluronic acid) acid); And a mixture thereof, characterized in that at least one selected from the group consisting of.
제1항에 있어서, The method of claim 1,
상기 단계 1)의 자성은 초상자성인 것을 특징으로 하는, 방법.The method, characterized in that the magnetism of step 1) is superparamagnetic.
제1항에 있어서, The method of claim 1,
상기 단계 1)의 자성 나노입자는,The magnetic nanoparticles of step 1),
산화 제2철(iron(Ⅱ) oxide: FeO), 산화 제3철(iron(Ⅲ) oxide, magnemite; Fe2O3, α-Fe2O3, β-Fe2O3, γ-Fe2O3, ε-Fe2O3), 마그네타이트(magnetite; Fe3O4) 및 산화 제2, 3철(iron(Ⅱ, Ⅲ) oxides; Fe4O5, Fe5O6, Fe5O7, Fe13O19, Fe25O32)로 이루어진 군으로부터 선택된 1종 이상인 것을 특징으로 하는, 방법.Ferric oxide (iron(Ⅱ) oxide: FeO), ferric oxide (iron(Ⅲ) oxide, magnemite; Fe 2 O 3 , α-Fe 2 O 3 , β-Fe 2 O 3 , γ-Fe 2 O 3 , ε-Fe 2 O 3 ), magnetite (Fe 3 O 4 ) and iron(II, Ⅲ) oxides; Fe 4 O 5 , Fe 5 O 6 , Fe 5 O 7 , Fe 13 O 19 , Fe 25 O 32 ), characterized in that at least one selected from the group consisting of.
제1항에 있어서,The method of claim 1,
상기 단계 1)의 고분자-자성 나노입자 클러스터는 양전하로 표면 개질된 고분자-자성 나노입자 클러스터인 것을 특징으로 하는, 방법.The polymer-magnetic nanoparticle cluster of step 1) is a polymer-magnetic nanoparticle cluster surface-modified with a positive charge.
제6항에 있어서, The method of claim 6,
상기 양전하로 표면 개질된 고분자-자성 나노입자 클러스터는 고분자-자성 나노입자 클러스터 표면에 양이온성 형질 전환제가 부착된 것을 특징으로 하는, 방법.The positively charged polymer-magnetic nanoparticle cluster is characterized in that the cationic transformant is attached to the polymer-magnetic nanoparticle cluster surface.
제7항에 있어서, The method of claim 7,
상기 양이온성 형질 전환제는,The cationic transformant,
폴리에틸렌이민(Polyethyleneimine, PEI), 페길레이티드 (Pegylated) 폴리에틸렌이민, 히스티딜레이티드 (Histidylated) 폴리에틸렌이민, 락토실레이티드 (Lactosylated) 폴리에틸렌이민, 엽산 공유결합형(Folate-conjugated) 폴리에틸렌이민, 멜리틴 공유결합형 (Melittin-conjugated) 폴리에틸린이민 등의 폴리에틸렌이민 유도체; 폴리라이신(Polylysine), 스퍼민(Spermine), 프로타민 설페이트(Protamine sulfate), 폴리아미도아민 (Polyamidoamine, PAMAM), 폴리 프로필렌이민 (Polypropyleneimine), 폴리브렌 (Polybrene) 및 디이에이이 덱스트란 (DEAE-dextran)으로 이루어진 군으로부터 선택된 1종 이상인 것을 특징으로 하는, 방법.Polyethyleneimine (PEI), Pegylated polyethyleneimine, Histidylated polyethyleneimine, Lactosylated polyethyleneimine, Folate-conjugated polyethyleneimine, Mellie Polyethyleneimine derivatives, such as a tin covalent bond type (Melittin-conjugated) polyethylinimine; Polylysine, Spermine, Protamine sulfate, Polyamidoamine (PAMAM), Polypropyleneimine, Polybrene, and DEAE-dextran Characterized in that at least one selected from the group consisting of, the method.
제1항에 있어서, The method of claim 1,
상기 단계 1)에서 고분자-자성 나노입자 클러스터를 1 내지 1000 ㎍/㎖로 처리하는 것을 특징으로 하는, 방법.In the step 1), the polymer-magnetic nanoparticle cluster is treated with 1 to 1000 μg/ml.
제1항에 있어서, The method of claim 1,
상기 줄기세포는 골수, 지방조직, 말초혈액, 간, 근육, 폐, 양수, 태반의 융모막 및 제대혈로 이루어진 군으로부터 선택된 1종 이상의 조직으로부터 유래된 것을 특징으로 하는, 방법.The stem cell is characterized in that derived from one or more tissues selected from the group consisting of bone marrow, adipose tissue, peripheral blood, liver, muscle, lung, amniotic fluid, placental chorionic membrane and umbilical cord blood.
제8항에 있어서, The method of claim 8,
상기 줄기세포는 배아 줄기세포 또는 성체 줄기세포인 것을 특징으로 하는, 방법.The method, characterized in that the stem cells are embryonic stem cells or adult stem cells.
제9항에 있어서, The method of claim 9,
상기 성체 줄기세포는 중간엽줄기세포, 인간 조직 유래 중간엽 기질세포, 인간 조직 유래 중간엽줄기세포 및 다분화능 줄기세포로 구성된 군에서 선택된 1종 이상의 성체 줄기세포인 것을 특징으로 하는, 방법.The adult stem cells are at least one adult stem cell selected from the group consisting of mesenchymal stem cells, human tissue-derived mesenchymal stromal cells, human tissue-derived mesenchymal stem cells, and multipotent stem cells.
제1항에 있어서, The method of claim 1,
상기 단계 2)에서 자기력을 0.1 내지 2.0 T로 가하는 것을 특징으로 하는, 방법.The method, characterized in that applying a magnetic force of 0.1 to 2.0 T in step 2).
1) 고분자-자성 나노입자 클러스터(cluster)를 줄기세포에 처리하는 단계;1) treating a macromolecular-magnetic nanoparticle cluster on stem cells;
2) 상기 단계 1)의 줄기세포에 자기력을 가하는 단계; 및2) applying a magnetic force to the stem cells of step 1); And
3) 상기 단계 2)의 줄기세포로부터 엑소좀을 분리하는 단계;를 포함하는, 엑소좀 제조방법.3) separating the exosomes from the stem cells of step 2); containing, exosomes manufacturing method.
제14항의 제조방법에 따라 제조된 엑소좀.An exosome prepared according to the manufacturing method of claim 14.
고분자-자성 나노입자 클러스터(cluster)를 포함하는 줄기세포 유래 엑소좀 생성 촉진용 조성물.A composition for promoting generation of exosomes derived from stem cells comprising a polymer-magnetic nanoparticle cluster.
제16항에 있어서, The method of claim 16,
상기 조성물은 DMEM (Dulbecco's Modified Eagle's Medium), MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10), DMEM/F-12 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12), α-MEM(α-Minimal essential Medium), G-MEM(Glasgow's Minimal Essential Medium), IMDM(Isocove's Modified Dulbecco's Medium), 및 KnockOut DMEM으로 이루어진 군에서 선택된 1종 이상의 배지에서 배양되는 것을 특징으로 하는, 조성물.The composition is DMEM (Dulbecco's Modified Eagle's Medium), MEM (Minimal Essential Medium), BME (Basal Medium Eagle), RPMI 1640, DMEM/F-10 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-10), DMEM/F- 12 (Dulbecco's Modified Eagle's Medium: Nutrient Mixture F-12), α-MEM (α-Minimal Essential Medium), G-MEM (Glasgow's Minimal Essential Medium), IMDM (Isocove's Modified Dulbecco's Medium), and KnockOut DMEM The composition, characterized in that cultivated in one or more selected medium.
PCT/KR2020/015093 2019-10-30 2020-10-30 Method for promoting stem cell-derived exosome production by means of magnetic nanoparticle cluster WO2021086139A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR1020190136417A KR102236399B1 (en) 2019-10-30 2019-10-30 Method for promoting production of stem cell-derived exosome using magnetic nanoparticle cluster
KR10-2019-0136417 2019-10-30

Publications (1)

Publication Number Publication Date
WO2021086139A1 true WO2021086139A1 (en) 2021-05-06

Family

ID=75466362

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2020/015093 WO2021086139A1 (en) 2019-10-30 2020-10-30 Method for promoting stem cell-derived exosome production by means of magnetic nanoparticle cluster

Country Status (2)

Country Link
KR (1) KR102236399B1 (en)
WO (1) WO2021086139A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112877274A (en) * 2021-01-29 2021-06-01 南京大学 Extracellular vesicle magnetic imprinting material and preparation method and application thereof
CN114632480A (en) * 2022-03-22 2022-06-17 和携科技有限公司 Superparamagnetic nanoparticle and exosome with angiogenesis directional enhancement function, and preparation method and application thereof
CN116143934A (en) * 2023-03-21 2023-05-23 诺赛联合(北京)生物医学科技有限公司 Stem cell exosome extraction kit and application thereof
CN117187165A (en) * 2023-09-12 2023-12-08 知行健康产业(广东)有限责任公司 Method for separating exosomes

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102634542B1 (en) * 2021-06-15 2024-02-07 전남대학교 산학협력단 Method for manufacturing surface-modified magnetic nanoparticles and surface-modified magnetic nanoparticles manufactured therefrom

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20120023684A (en) * 2009-04-23 2012-03-13 소시에다지 베네피시엔찌 이스라엘리따 브라질레이라 오스삐따우 알버트 아인슈타인 Method for isolating exosomes from biological solutions using iron oxide nanoparticles
KR20180074123A (en) * 2016-12-23 2018-07-03 주식회사 제놉시 Magnetic nanostructure for detecting and isolating circulating cell-free DNA comprising conductive polymers containing magnetic nanoparticles and cationic polymers
KR20180111674A (en) * 2017-03-31 2018-10-11 (주)안트로젠 Media including mesenchymal stem cells derived high purity and high concentration exosome and the producing method thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20120023684A (en) * 2009-04-23 2012-03-13 소시에다지 베네피시엔찌 이스라엘리따 브라질레이라 오스삐따우 알버트 아인슈타인 Method for isolating exosomes from biological solutions using iron oxide nanoparticles
KR20180074123A (en) * 2016-12-23 2018-07-03 주식회사 제놉시 Magnetic nanostructure for detecting and isolating circulating cell-free DNA comprising conductive polymers containing magnetic nanoparticles and cationic polymers
KR20180111674A (en) * 2017-03-31 2018-10-11 (주)안트로젠 Media including mesenchymal stem cells derived high purity and high concentration exosome and the producing method thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHANG MING, YAW-JEN, CHAO PEI YU, YU QING: "Exosome purification based on PEG-coated Fe3O4 nanoparticles", PLOS ONE, vol. 13, no. 6, e0199438, 22 June 2018 (2018-06-22), pages 1 - 13, XP055808115, ISSN: 1932-6203, DOI: 10.1371/journal.pone.0199438 *
URSULA ALTANEROVA, BABINCOVA MELANIA, BABINEC PETER, BENEJOVA KATARINA, JAKUBECHOVA JANA, ALTANEROVA VERONIKA, ZDURIENCIKOVA MARTI: "Human mesenchymal stem cell-derived iron oxide exosomes allow targeted ablation of tumor cells via magnetic hyperthermia", INTERNATIONAL JOURNAL OF NANOMEDICINE, vol. 2017, no. 12, 1 January 2017 (2017-01-01), pages 7923 - 7936, XP055627984, DOI: 10.2147/IJN.S145096 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112877274A (en) * 2021-01-29 2021-06-01 南京大学 Extracellular vesicle magnetic imprinting material and preparation method and application thereof
CN112877274B (en) * 2021-01-29 2024-02-09 南京大学 Extracellular vesicle magnetic imprinting material and preparation method and application thereof
CN114632480A (en) * 2022-03-22 2022-06-17 和携科技有限公司 Superparamagnetic nanoparticle and exosome with angiogenesis directional enhancement function, and preparation method and application thereof
CN114632480B (en) * 2022-03-22 2024-02-09 和携科技有限公司 Superparamagnetic nanoparticle with directional angiogenesis enhancing function, exosome, preparation method and application thereof
CN116143934A (en) * 2023-03-21 2023-05-23 诺赛联合(北京)生物医学科技有限公司 Stem cell exosome extraction kit and application thereof
CN116143934B (en) * 2023-03-21 2023-07-25 诺赛联合(北京)生物医学科技有限公司 Stem cell exosome extraction kit and application thereof
CN117187165A (en) * 2023-09-12 2023-12-08 知行健康产业(广东)有限责任公司 Method for separating exosomes

Also Published As

Publication number Publication date
KR102236399B1 (en) 2021-04-02

Similar Documents

Publication Publication Date Title
WO2021086139A1 (en) Method for promoting stem cell-derived exosome production by means of magnetic nanoparticle cluster
Liao et al. Exosomes: the next generation of endogenous nanomaterials for advanced drug delivery and therapy
CN111603454A (en) Multi-targeting fusion cell membrane modified bionic nano delivery system and preparation method and application thereof
US20100055167A1 (en) Stem cell delivery of anti-neoplastic medicine
CN108553650B (en) Bionic nano erythrocyte gene vector and preparation method and application thereof
Duan et al. Superparamagnetic iron oxide-loaded cationic polymersomes for cellular MR imaging of therapeutic stem cells in stroke
WO2009134866A2 (en) Cell membrane engineering
CN111670029A (en) Nanoprosomes derived from adult stem cells and their use in targeted therapy
US20110008304A1 (en) Use of cells to facilitate targeted delivery of nanoparticle therapies
WO2012148130A1 (en) Method for preparing induced pluripotent stem cells using microvesicles derived from embryonic stem cells
WO2014054588A1 (en) Nanogel/exosome complex and dds
WO2022134246A1 (en) Method for extracting neural tissue-derived exosomes
WO2016141162A1 (en) Methods for making neural stem cells and uses thereof
CN113136362A (en) Vesicle and application thereof
Xia et al. Development of a microdevice-based human mesenchymal stem cell-mediated drug delivery system
Zhang et al. Smart graphene-based hydrogel promotes recruitment and neural-like differentiation of bone marrow derived mesenchymal stem cells in rat skin
Thalakiriyawa et al. Regenerative potential of mesenchymal stem cell-derived extracellular vesicles
WO2021147923A1 (en) Vesicle and use thereof
CN115463107A (en) Preparation method and application of engineered vesicle
WO2024045404A1 (en) Bone marrow supernatant and use thereof in cell culture
Cheng et al. A new strategy for stem cells therapy for erectile dysfunction: adipose-derived stem cells transfect Neuregulin-1 gene through superparamagnetic iron oxide nanoparticles
CN116036042A (en) Stable exosome nanoparticle targeting M2 type tumor-associated macrophage and preparation method and application thereof
Nguyen et al. Harnessing the versatility of PLGA nanoparticles for targeted Cre-mediated recombination
WO2007105171A2 (en) Killing of selected cells
CN114452266A (en) Nucleic acid drug delivery system based on recombinant ribosomal protein and preparation method and application thereof

Legal Events

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

Ref document number: 20880800

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20880800

Country of ref document: EP

Kind code of ref document: A1