WO2018088821A1 - Composition for promoting stem cell activity, comprising histone deacetylase inhibitor and priming factor as active ingredients - Google Patents

Composition for promoting stem cell activity, comprising histone deacetylase inhibitor and priming factor as active ingredients Download PDF

Info

Publication number
WO2018088821A1
WO2018088821A1 PCT/KR2017/012675 KR2017012675W WO2018088821A1 WO 2018088821 A1 WO2018088821 A1 WO 2018088821A1 KR 2017012675 W KR2017012675 W KR 2017012675W WO 2018088821 A1 WO2018088821 A1 WO 2018088821A1
Authority
WO
WIPO (PCT)
Prior art keywords
disease
stem cells
vpa
stem cell
pulmonary
Prior art date
Application number
PCT/KR2017/012675
Other languages
French (fr)
Korean (ko)
Inventor
신동명
임지선
오연목
Original Assignee
울산대학교 산학협력단
재단법인 아산사회복지재단
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 KR1020170148019A external-priority patent/KR102097191B1/en
Application filed by 울산대학교 산학협력단, 재단법인 아산사회복지재단 filed Critical 울산대학교 산학협력단
Priority to CN201780069520.XA priority Critical patent/CN110462023B/en
Priority to JP2019520846A priority patent/JP6934516B2/en
Publication of WO2018088821A1 publication Critical patent/WO2018088821A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/465Nicotine; Derivatives thereof
    • 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
    • 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

Definitions

  • the present invention relates to a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
  • MSCs Mesenchymal stem-cells
  • BM bone-marrow
  • UC umbilical-cord
  • UB umbilical cord blood
  • MSCs Multipotent progenitor cells.
  • Administration of MSCs repairs organ tissues of damaged organs to provide a therapeutic effect.
  • the efficacy of MSCs is due to cytoprotective effects regulated by cytokines and paracrine factors such as pro-angiogenic and pro-arteriogenic effects.
  • current MSC treatments are limited due to their low therapeutic effectiveness, particularly in vivo engraftment and survival of transplanted cells. Most ( ⁇ 99%) intravenously inject MSCs near the lungs, of which only 2-3% are released into the circulation. Thus, there is a need to understand the exact mechanisms for the migration and engraftment of MSCs during tissue repair.
  • chemokines such as stromal cell derived factor-1 (SDF-1) and growth factors such as vascular endothelial growth-factor (VEGF), platelet- Platelet-derived growth-factor (PDGF) or hepatocyte growth-factor (HGF) plays an important role in the migration and engraftment of adult MSCs.
  • SDF-1 stromal cell derived factor-1
  • growth factors such as vascular endothelial growth-factor (VEGF), platelet- Platelet-derived growth-factor (PDGF) or hepatocyte growth-factor (HGF) plays an important role in the migration and engraftment of adult MSCs.
  • VEGF vascular endothelial growth-factor
  • PDGF platelet- Platelet-derived growth-factor
  • HGF hepatocyte growth-factor
  • HSPCs After chemotherapy () in the bone marrow.
  • the sensitivity / responsiveness of HSPCs to the SDF-1 gradient is positively influenced (“primed”) by a rich set of molecules in the injured tissue.
  • bioactive lipids eg, sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P)
  • neutrophil-derived cationic peptide caseycides eg, sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P)
  • neutrophil-derived cationic peptide caseycides eg, neutrophil-derived cationic peptide caseycides.
  • Cathelicidin LL-37
  • sMAC soluble membrane attack complex
  • primed MSCs promote cell migration, colony-forming activity and anti-inflammatory capacity in cell culture conditions, which enhances the effect of treating pulmonary arterial hypertension (PAH).
  • PAH pulmonary arterial hypertension
  • primed MSCs still exhibit limited in vivo engraftment in injured tissue.
  • An object of the present invention is to provide a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient, and a method for promoting stem cell activity through the same.
  • Another object of the present invention is to provide a pharmaceutical composition for preventing or treating hypertension, including a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture solution thereof as an active ingredient.
  • Another object of the present invention to provide a pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases, including stem cells treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient. Is in.
  • the present invention provides a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
  • the present invention provides a method for promoting stem cell activity comprising the step of treating a histone deacetylation inhibitor and priming factor to the isolated stem cells.
  • the present invention also provides a pharmaceutical composition for preventing or treating hypertension, comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
  • the present invention also provides the use of histone deacetylation inhibitors and priming factors for promoting stem cell activity.
  • a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof there is provided the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof.
  • the present invention also provides the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof in the manufacture of a medicament for preventing or treating an inflammatory disease or an immune disease.
  • the present invention relates to a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
  • a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
  • the activity of the stem cells is increased. Excellent improvement.
  • primed MSCs promote cell migration, colony-forming activity, and anti-inflammatory capacity in cell culture conditions, which may be useful therapeutic strategies because they enhance the effect of treating hypertension, inflammatory or immunological diseases. do.
  • FIG. 2 shows the results for the effect of VPA on UC-MSCs primed with S1P.
  • a and B Flow cytometry and pluripotency (B) for surface antigen (A) of UC-MSCs primed for 24 hours with 0.5-mM VPA alone (VPA) or in combination with 50-nM S1P (VPA + S1P) The analysis results are shown.
  • FIG. 3 shows the results for improved reactivity to SDF-1 in UC-MSCs primed with VPA + S1P.
  • a and B 150-ng / ml SDF-1 of MSCs exposed for 24 hours to 0.5-mM VPA alone (VPA), 50-nM S1P alone (S1P) or combination with 50-nM S1P (VPA + S1P) The results of the chemotaxis analysis for.
  • A Representative images of transwell insets are shown in the migration assay.
  • (B) Relative amounts for migration were quantified as a change in magnification for the number of migrated cells and expressed as means ⁇ SEM (n 6).
  • VPA + S1P upregulated genes involved in the therapeutic effect of MSCs VPA + S1P upregulated genes involved in the therapeutic effect of MSCs.
  • MMP12 MMP12 in UC-MSCs primed for 24 h with 0.5-mM VPA alone (VPA), 50-nM S1P alone (S1P) or in combination with 50-nM S1P (VPA + S1P) (A), growth factors (B), angiogenesis (C) and anti-inflammatory (D) related gene expression levels. Expression levels were calculated as the numerical ratio of primed UC-MSCs to unprimed UC-MSCs (NT).
  • FIG. 7 shows the results of improving the treatability of VPA + S1P primed UC-MSCs with respect to bladder function recovery in the HCl-IC rat model.
  • (a) Results of awake cystometry under non-anesthetic.
  • FIG. 9 shows the effect of UCB-MSCs and VPA + S1P primed MSCs (VPA + S1P-MSCs) in MCT induced PAH rat model.
  • MCT induced an increase in RVSP levels and significantly reduced this elevation as a result of administration of VPA + S1P primed UC-MSC two weeks after MCT infusion (left panel).
  • MCT increased the weight ratio of RV / (LV + S) and VPA + S1P-UC-MSC significantly reduced this effect (right panel).
  • MCT increased lung tissue inflammation and medial wall thickness index (vascular wall thickness per vessel diameter), and VPA + S1P-MSC significantly reduced this increase (top panel).
  • Unprimed UCB-MSC showed little improvement in RV / (LV + S) and media thickness index. It shows an immunofluorescence staining of ⁇ -SMA + smooth muscle cells in the pulmonary artery (lower panel) (magnification ⁇ 400).
  • RVSP right ventricular systolic pressure
  • CTL control
  • MSC human cord blood mesenchymal stem cell
  • S1P-MSC sphingosine-1-phosphate primed human umbilical cord mesenchymal stem cells
  • RV right ventricle
  • LV + S left ventricle and interventricular septum.
  • CXCR4 a major target of stem cell priming, is upregulated by DNA-demethylating agent 5-azacytidine (5-Aza) and histone deacetylation inhibitor valproic acid (VPA). do.
  • 5-azacytidine 5-Aza
  • VPA histone deacetylation inhibitor valproic acid
  • the present invention provides a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
  • the histone deacetylation inhibitor may be valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) or sirtinol, but is not limited thereto. It doesn't happen.
  • VPA valproic acid
  • NaB sodium butyrate
  • NAD nicotinamide
  • sirtinol sirtinol
  • the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), cathelicidin (LL-37) or It may be a pioglitazone, but is not limited thereto.
  • the VPA may be included in a concentration of 0.1 to 1.0 mM
  • the S1P may be included in a concentration of 10 to 50 nM, but is not limited thereto.
  • the term "priming” refers to a phenomenon in which reactivity (activity) is improved to enhance the therapeutic efficacy of stem cells, and in the present invention, a histone deacetylation inhibitor and a priming factor that induce the priming. (priming factor) to promote the activity of the stem cells.
  • stem cell activity refers to cell mobility, cell proliferation ability, pluripotency (in vitro differentiation into chondrocytes, osteocytes or adipocyte lineage), colony forming ability or anti-inflammatory activity of stem cells, etc. .
  • the activity promotion or priming induction is not only induced priming of stem cells when the stem cells are directly treated with histone deacetylation inhibitors and priming factors, but also by the priming treatment (pre-treatment). Other differentiation may be induced by using stem cells with improved stem cell activity.
  • composition for promoting activity can be enhanced by injecting in vivo by mixing with a cell therapy agent for treatment, to enhance the in vivo effect of the cell therapy, and after treatment of the present composition to the stem cells themselves cell therapy agent increased function It can also be used as a method for implanting in vivo.
  • the priming of the present invention is to enhance stem cell function, including cell mobility, colony forming ability, and anti-inflammatory activity of stem cells.
  • stem cell refers to a cell having the ability to differentiate into two or more cells while having a self-replicating ability, totipotent stem cells, pluripotent stem cells It can be classified into multipotent stem cells.
  • Stem cells of the present invention may be selected without appropriate limitation depending on the purpose, and may be derived from adult cells, such as all known tissues, cells, etc. derived from mammals, including humans, preferably humans, for example, Bone marrow, umbilical cord blood, placenta (or placental tissue cells), fat (or adipose tissue cells) and the like.
  • the stem cells are restricted from bone marrow, adipose tissue, muscle tissue, ex vivo cultured autologous mesenchymal stem cells, allogeneic mesenchymal stem cells, umbilical cord blood, embryonic yolk sac, placenta, umbilical cord, periosteum, fetal and adolescent skin, and blood It may be a stem cell obtained without, and may be a stem cell derived from the fetus or shortly after birth or adult.
  • the stem cells are neural stem cells, liver stem cells, hematopoietic stem cells, cord blood stem cells, epidermal stem cells, gastrointestinal stem cells, endothelial stem cells, muscle stem cells, mesenchymal stem cells and It is selected from the group consisting of pancreatic stem cells, and more preferably may be selected from the group consisting of liver stem cells, hematopoietic stem cells, cord blood stem cells and mesenchymal stem cells, but is not limited thereto.
  • the present invention provides a method for promoting stem cell activity comprising the step of treating a histone deacetylation inhibitor and priming factor to the isolated stem cells.
  • the histone deacetylation inhibitor may be valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) or sirtinol, but is not limited thereto. It doesn't happen.
  • VPA valproic acid
  • NaB sodium butyrate
  • NAD nicotinamide
  • sirtinol sirtinol
  • the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), cathelicidin (LL-37) or It may be a pioglitazone, but is not limited thereto.
  • the VPA may be included in a concentration of 0.1 to 1.0 mM
  • the S1P may be included in a concentration of 10 to 50 nM, but is not limited thereto.
  • the present invention also provides a pharmaceutical composition for preventing or treating hypertension, comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
  • the present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
  • stem cells when stem cells are activated through priming, they can be applied to various inflammatory diseases and immune diseases by participating in immune regulation and inflammatory response, and there are various reports on the mechanisms of immune control and inflammatory response of primed stem cells. (J Orthop Res. 2016 Apr 6, 1-12; Stem Cell Rev and Rep (2014) 10: 351-375; Stem Cells International 2016 Article ID 9364213).
  • the histone deacetylation inhibitor may be valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) or sirtinol, but is not limited thereto. It doesn't happen.
  • VPA valproic acid
  • NaB sodium butyrate
  • NAD nicotinamide
  • sirtinol sirtinol
  • the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), cathelicidin (LL-37) or It may be a pioglitazone, but is not limited thereto.
  • the "culture medium” includes a culture medium capable of supporting stem cell growth and survival in vitro, secretion of cultured stem cells contained in the medium, and the like.
  • the medium used for culturing includes all conventional mediums used in the art suitable for culturing stem cells. Depending on the type of cells, medium and culture conditions can be selected.
  • the medium used for the culturing is preferably a cell culture minimum medium (CCMM), and generally includes a carbon source, a nitrogen source and a trace element component.
  • CCMM cell culture minimum medium
  • Such cell culture minimal media include, for example, Dulbecco's Modified Eagle's Medium (DMEM), Minimal Essential Medium (MEM), Basic Medium Eagle (BME), RPMI1640, F-10, F-12, ⁇ Minimal Essential Medium (GMEM) (Glasgow's Minimal essential Medium) and Iscove's Modified Dulbecco's Medium, but are not limited to these.
  • DMEM Dulbecco's Modified Eagle's Medium
  • MEM Minimal Essential Medium
  • BME Basic Medium Eagle
  • RPMI1640 RPMI1640
  • F-10 F-12
  • GMEM ⁇ Minimal Essential Medium
  • Iscove's Modified Dulbecco's Medium Iscove's Modified Dulbecco's Medium
  • the medium may include antibiotics such as penicillin, streptomycin, gentamicin, and the like.
  • the present invention is the form containing all of the stem cells, their secretions, media components, forms containing only secretions and media components, only secretion to separate or used in combination with stem cells, or by administering only stem cells It is also possible to use it in a form that produces secretions in the body.
  • the stem cells can be obtained using any method known in the art.
  • Stem cells treated with histone deacetylation inhibitors and priming factors of the present invention can be used as a cell therapy for the treatment of certain diseases, and the treatment can be direct or pre-treatment of the molecules. .
  • cell therapy means proliferating, screening, or otherwise altering the biological properties of cells in vitro to autologous, allogenic, and xenogenic cells to restore the function of cells and tissues. It refers to medicines used for the purpose of treatment, diagnosis and prevention through a series of actions.
  • the cell therapeutic agent may be administered to the human body via any general route as long as it can reach the desired tissue.
  • the hypertension is idiopathic pulmonary arterial hypertension; Familial pulmonary arterial hypertension; Pulmonary arterial hypertension associated with collagen vascular disease, congenital pulmonary shortness, portal hypertension, HIV infection, drugs or toxins; Pulmonary hypertension associated with thyroid disorders, glycogen storage disease, Gaucher disease, hereditary hemorrhagic capillary dilatation, hemochromatosis, myeloproliferative disorder or splenectomy; Pulmonary arterial hypertension associated with pulmonary capillary angiomatosis; Persistent pulmonary hypertension in newborns; Pulmonary hypertension associated with chronic obstructive pulmonary disease, interstitial lung disease, hypoxia induced alveolar hypoventilatory disorder, hypoxia induced sleep disorder breathing or chronic exposure to high altitudes; Pulmonary hypertension associated with developmental abnormalities; And pulmonary hypertension by thromboembolic obstruction of distal pulmonary artery, more preferably idiopathic pulmonary arterial hypertension, familial
  • compositions of the present invention may be prepared using pharmaceutically suitable and physiologically acceptable auxiliaries in addition to the active ingredients, which may include excipients, disintegrants, sweeteners, binders, coatings, swelling agents, lubricants, lubricants. Or solubilizers such as flavoring agents can be used.
  • the pharmaceutical composition of the present invention may be preferably formulated into a pharmaceutical composition by containing one or more pharmaceutically acceptable carriers in addition to the active ingredient for administration.
  • Acceptable pharmaceutical carriers in compositions formulated in liquid solutions are sterile and physiologically compatible, including saline, sterile water, Ringer's solution, buffered saline, albumin injectable solutions, dextrose solution, maltodextrin solution, glycerol, ethanol and One or more of these components may be mixed and used, and other conventional additives such as antioxidants, buffers and bacteriostatic agents may be added as necessary. Diluents, dispersants, surfactants, binders and lubricants may also be added in addition to formulate into injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like.
  • compositions of the present invention may be granules, powders, coated tablets, tablets, capsules, suppositories, syrups, juices, suspensions, emulsions, drops or injectable solutions and sustained release formulations of the active compounds, and the like.
  • the pharmaceutical compositions of the present invention may be administered in a conventional manner via intravenous, intraarterial, intraperitoneal, intramuscular, intraarterial, intraperitoneal, sternum, transdermal, nasal, inhalation, topical, rectal, oral, intraocular or intradermal routes.
  • An effective amount of the active ingredient of the pharmaceutical composition of the present invention means an amount required to prevent or treat a disease.
  • the type of disease the severity of the disease, the type and amount of the active and other ingredients contained in the composition, the type of formulation and the age, weight, general health, sex and diet, sex and diet, time of administration, route of administration and composition of the patient. It can be adjusted according to various factors including the rate of secretion, the duration of treatment, and the drug used concurrently.
  • the present invention also provides the use of histone deacetylation inhibitors and priming factors for promoting stem cell activity.
  • a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof there is provided the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof.
  • the present invention also provides the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof in the manufacture of a medicament for preventing or treating an inflammatory disease or an immune disease.
  • UC-derived MSCs are 2-mM L-glutamine, 20-mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) (pH 7.3), minimal-required medium ( minimum-essential medium (MEM) non-essential amino acid solution, penicillin / streptomycin (Corning Cellgro, Pittsburgh, PA), 1-mg / ml ascorbic acid (Sigma-Aldrich), 10% heat-inactivated fetal bovine serum ( fetal bovine serum; FBS) (HyClone), 5-ng / mL human epidermal growth factor (Sigma-Aldrich, St.
  • DMEM low-glucose Dulbecco's modified Eagle's medium
  • UC-MSCs extended to 5 times or less were used. Expression of surface proteins was analyzed as previously reported (Stem Cells and Dev. 24 (2015) 1658-1671).
  • UC-MSCs were primed for 1 day with VPA (0.5-mM; Sigma-Aldrich) or 5-Aza (1- ⁇ M; Sigma-Aldrich) alone or mixed with 50-nM S1P.
  • VPA 0.5-mM
  • Sigma-Aldrich 5-Aza (1- ⁇ M; Sigma-Aldrich
  • UC-MSCs are TrypLE solution (Thermo Scientific, Pittsburgh PA) as was separated, washed and resuspended in DMEM containing 0.5% bovine serum albumin (BSA) , 3 X 10 4 trans-well inserts with cells / well density ( Transwell inserts) (Corning Costar, Pittsburgh, Pa.) Were inoculated into the upper chamber.
  • BSA bovine serum albumin
  • the lower chamber was filled with 150-ng / ml SDF-1 (R & D Systems) in DMEM containing 0.5% BSA. After 1 day, the inserts were removed from the transwell plates. Cells remaining in the upper chamber were scraped with cotton wool, and the migrated cells were fixed in 4% paraformaldehyde (PFA) solution dissolved in phosphate-buffered saline (PBS), followed by 0.5% crystal violet ( Sigma-Aldrich). Digital images were analyzed with Image Pro 5.0 software (Media Cybernetics, Rockville, MD, USA) to quantify the stained cells on the underside of the membrane.
  • PFA paraformaldehyde
  • PBS phosphate-buffered saline
  • crystal violet Sigma-Aldrich
  • UC-MSCs primed with 50-nM S1P and 0.5-mM VPA were starvated for 12 hours at 37 ° C. in DMEM containing 0.5% BSA and 5, 10, 20 with 150-ng / ml SDF-1. Or after 30 min of stimulation, phosphorylation of mitogen-activated protein kinase (MAPK) p42 / 44 and AKT (Ser473) using a 30 ⁇ g-cell extract was analyzed via Western blot. For gene expression analysis, total RNA from indicated cells was reverse transcribed and the indicated transcripts were quantified via RQ-PCR.
  • MAPK mitogen-activated protein kinase
  • An object of the present invention is to develop an umbilical cord-derived MSCs derived from human umbilical cord-derived MSCs primed with V1P + S1P to treat interstitial cystitis / bladder pain syndrome (IC / BPS) in a rat model.
  • V1P + S1P primed UC-MSCs were administered to rats with bladder injuries and affected the effects on bladder urination function, urothelium denudation, mast cell infiltration, tissue fibrosis, apoptosis and tumor formation. Measured. For all experiments, two independent experiments were performed on 5 independent animals per group.
  • HCl infused IC / BPS rat models have been previously reported (Stem Cells and Development 24, 2015, 1648-1657).
  • HCl week after injury the abdomen was cut, using a 500 ⁇ m syringe and 26-gauge needle and a 2.5 ⁇ 10 5 (250 K) UC-MSCs or PBS was administered directly to the front wall outer layer and the ceiling of the bladder.
  • indomethacin PMG Pharm Co., Ltd.
  • gefitinib to block Wnt or IGF-mediated signaling
  • Santa Cruz Biotechnology Santa Cruz, CA, USA; every day at 5 mg / kg was injected subcutaneously.
  • Cystometrograms were performed in non-anesthesia and non-suppressed mice in metabolic cages. Catheter was inserted simultaneously 3 days prior to bladder pressure measurement for measurement of intravesical pressure (IVP) and intra-abdominal pressure (IAP) recordings. Briefly, after anesthesia induction, a cuffed polyethylene catheter (PE-50; Becton-Dickinson, Parsippany, NJ, USA) was inserted into the bladder ceiling via an intraperitoneal incision. To record the IAP, an abdominal balloon (Latex; Daewoo Medical, Incheon, Korea) near the cuff of the catheter tip was placed proximal to the bladder and tied to another catheter with a silk thread.
  • PE-50 Becton-Dickinson, Parsippany, NJ, USA
  • the polyethylene catheter (PE-50) was heated in warm water and the tip of the insert was extended to ⁇ 1.5 times its original length and filled with heparinized saline (100 IU / mL).
  • the extended catheter was inserted into the femoral vein. The catheter then penetrated through the subcutaneous space, exited through the back of the animal, and attached to the back skin. After surgery, each rat was raised individually and maintained in the same way.
  • a T-tube connected to a pressure transducer (Research Grade Blood Pressure Transducer; Harvard Apparatus, Holliston, Mass., USA) and a microinjection pump (PHD22 / 2000 pump; Harvard Apparatus)
  • An indwelling catheter was connected to the bladder via a double valve.
  • another indwelling catheter with a fluid-filled abdominal balloon was connected to another pressure transducer.
  • Sterile saline was injected into the bladder at a rate of 0.4 mL / min and urine volume was continuously recorded through a fluid collector connected to a force displacement transducer (Research Grade Isometric Transducer; Harvard Apparatus).
  • IVP, IAP and urination volume were continuously recorded using an MP150 data acquisition system (Biopac Systems, Goleta, CA, USA) equipped with Acq Knowledge 3.8.1 software at a sampling rate of 50 Hz. All urination cycle values measured for 8 minutes from each animal were used for the evaluation.
  • NVC non-voiding contraction
  • MCT Monocrotaline
  • Male specific-pathogen-free Lewis rats (8 weeks old, 250-280 g) were reared without restriction of food and water, under the control of breeding temperature and light (12 hour contrast cycle).
  • PAH was induced through subcutaneous administration of MCT (60 mg / kg, Sigma).
  • Mice in the control group received the same volume of PBS.
  • priming-induced UC-MSCs or UC-MSCs pretreated with VPA + S1P were administered via the tail vein at a density of 2.5 ⁇ 10 5 cells per 200 ⁇ l PBS.
  • a solvent-only vehicle control was administered with 200 ⁇ l PBS without cells. Prior to injection, cells were washed twice with warm PBS and viability of the administered cells was observed using FACS analysis via 7-AAD (BD Biosciences) exclusion staining.
  • RVSP right ventricular systolic pressure
  • RVH right ventricular hypertrophy
  • ⁇ -SMA ⁇ -smooth muscle actin
  • UC-MSCs primed with 5-Aza + S1P in transwell migration assays reduced migration activity for SDF-1 responses by about 30% compared to unprimed cells.
  • FIG. 1E 5-Aza + S1P priming severely impaired the activity of clonal CFU-F, which indicates the frequency of self-renewal into clonal progenitor cells (FIG. 1F).
  • Cell proliferation of MSCs was little changed by the priming of 5-Aza + S1P (FIG. 1D).
  • the present inventors investigated the effect of VPA on the priming of UC-MSCs at low concentrations of S1P treatment.
  • 50-nM S1P four times less than the optimal dose (200-nM) used for priming of fat- and UCB-derived MSCs.
  • UC-MSCs priming increased VT + S1P treatment two to three times higher chemotactic activity for SDF-1 than unprimed cells (FIGS. 3A and 3A). 3B).
  • the response to SDF-1 enhanced by VPA + S1P priming appeared completely different from that of 5-Aza + S1P priming.
  • UC-MSCs exposed to VPA + S1P increased the phosphorylation of MAPK p42 / 44 and AKT proteins, indicating activation of signaling pathways (FIG. 3C).
  • VPA + S1P priming we tested the effect of VPA + S1P priming on other cellular activities related to the therapeutic effect of MSCs. Unlike 5-Aza + S1P (FIG. 1), UC-MSCs primed with VPA + S1P improved both cell proliferation (FIG. 4A) and clonal CFU-F activity (FIG. 4B). However, no increase in cell proliferation and CFU-F activity was observed in UC-MSCs treated with VPA alone and S1P alone (FIGS. 4A and 4B). Since MSCs affect anti-inflammatory and immunomodulatory capacity, we tested whether VPA + S1P priming affected the anti-inflammatory effects of UC-MSCs.
  • the present inventors prepared a conditioned medium collected from unprimed UC-MSCs or VPA alone or from UC-MSCs primed with VPA + S1P, and the conditioned medium stimulated with lipopolysaccharide (LPS).
  • LPS lipopolysaccharide
  • TNF- ⁇ Tumor necrosis factor- ⁇
  • Conditional medium collected from UC-MSCs was effective in reducing the secretion of TNF- ⁇ from LPS stimulated MH-S cells (FIG. 4C).
  • the conditioned media collected from VPA + S1P primed UC-MSCs inhibited TNF- ⁇ secretion more than unprimed UC-MSCs or UC-MSCs primed with VPA alone.
  • CM collected with IMR90, a human lung fibroblast, as a control which was shown to further increase TNF- ⁇ secretion in vitro anti-inflammatory analysis.
  • UC-MSCs primed with VPA + S1P showed growth factors and their receptors [eg, PDGFB, PDGFRB, cMET] (FIG. 5B), pre-angiogenesis- [eg , VEGFB, VEGFC, ANGPT1, ANGPT2] (FIG. 5C), anti-inflammatory- [eg, LIF, TSG6, IDO1, IDO2] (FIG.
  • the present inventors attempted to determine the lowest concentration of VPA and S1P by performing a chemotaxis analysis on SDF-1a according to various VPA and S1P concentrations.
  • VPA was fixed at 0.5 mM, and the S1P concentration was tested at 5-50 nM. As a result, the lowest concentration of S1P was determined to be 10 nM (FIG. 6A). In addition, S1P was fixed at 50 nM and the VPA concentration was 0.05-0.5 mM. As a result, the lowest concentration of VPA was determined to be 0.1 nM (FIG. 6B).
  • RVSP increased significantly after 4 weeks of MCT administration.
  • administration of UC-MSC primed with VPA + S1P significantly reduced the elevation of MCT-induced RVSP (FIG. 9a).
  • RV / (LV + S) was also increased, with VPA + S1P-MSC significantly reducing RV hypertrophy, and unpriming UC-MSC was ineffective (FIG. 9A).
  • VPA + S1P-MSC administration reduced lung tissue inflammation, medial thickness index, and ⁇ -SMA + smooth muscle cell induced by MCTs (FIG. 9B).
  • VPA + S1P-MSC administration reduced lung tissue inflammation, medial thickness index, and ⁇ -SMA + smooth muscle cell induced by MCTs (FIG. 9B).

Abstract

The present invention relates to a composition for promoting stem cell activity, comprising a histone deacetylase inhibitor and a priming factor as active ingredients. When stem cells were treated with a low-concentration histone deacetylase inhibitor and priming factor, the activity of the stem cells significantly improved. In particular, primed MSCs promote a cell movement, colony-forming activity, and anti-inflammatory capability in terms of cell culturing conditions and thereby improve hypertension, inflammatory disease, or immune disease treatment effects and thus are expected to become a useful treatment strategy.

Description

히스톤 탈아세틸화 저해제 및 프라이밍 인자를 유효성분으로 포함하는 줄기세포 활성 촉진용 조성물Stem cell activity promoting composition comprising a histone deacetylation inhibitor and a priming factor as an active ingredient
본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍(priming) 인자를 유효성분으로 포함하는 줄기세포 활성 촉진용 조성물에 관한 것이다.The present invention relates to a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
중간엽 줄기세포(Mesenchymal stem-cells, MSCs)는 골수(bone-marrow; BM), 지방, 치수(dental pulp), 탯줄(umbilical-cord; UC) 및 제대혈(UC-blood; UCB)로부터 분리된, 다능성 전구 세포(multipotent progenitor cell)이다. MSC의 투여는 손상된 기관의 장기 조직을 복구하여 치료 효과를 제공한다. MSC의 효능은 사이토카인에 의해 조절되는 세포보호효과(cytoprotective effect)와 전혈관형성(pro-angiogenic) 및 전동맥형성(pro-arteriogenic) 효과와 같은 측분비인자(paracrine factor)에 기인한다. 하지만, 현재 MSC 치료는 치료 유효성, 특히 생체 내 생착(engraftment) 및 이식된 세포의 생존율이 낮아 한계가 있다. 대부분(≥99%)은 MSCs를 폐 부근에 정맥 주사하는데, 이들 중 단지 2-3% 만이 순환계로 방출된다. 따라서, 조직 복구 과정에서 MSCs의 이동 및 생착에 대한 정확한 기작을 이해할 필요가 있다. Mesenchymal stem-cells (MSCs) are isolated from bone-marrow (BM), fat, dental pulp, umbilical-cord (UC) and umbilical cord blood (UCB). , Multipotent progenitor cells. Administration of MSCs repairs organ tissues of damaged organs to provide a therapeutic effect. The efficacy of MSCs is due to cytoprotective effects regulated by cytokines and paracrine factors such as pro-angiogenic and pro-arteriogenic effects. However, current MSC treatments are limited due to their low therapeutic effectiveness, particularly in vivo engraftment and survival of transplanted cells. Most (≥99%) intravenously inject MSCs near the lungs, of which only 2-3% are released into the circulation. Thus, there is a need to understand the exact mechanisms for the migration and engraftment of MSCs during tissue repair.
몇몇 케모카인들, 예를 들면, 기질세포 유래 인자-1(stromal cell derived factor-1; SDF-1) 및 성장인자, 예를 들면, 혈관내피성장인자(vascular endothelial growth-factor; VEGF), 혈소판-유래 성장인자(platelet-derived growth-factor; PDGF) 또는 간세포 성장인자(hepatocyte growth-factor; HGF)는 성체 MSCs의 이동 및 생착에 중요한 역할을 한다. 이들 중, SDF-1 및 이의 수용체인 CXCR4 축(axis)은 줄기 세포의 호밍(homing)/생착에 중요한 인자이다. 사실, 골수에 있어서 SDF-1 구배(gradient)에 따라 조혈 줄기/전구 세포(hematopoietic stem/progenitor cells; HSPCs) 생착은 이식을 위한 전처치(conditioning)(예를 들면, 전신 방사선 조사 및 골수 절제식 항암치료) 후에 골수에서 상향 조절된다. SDF-1 구배에 대한 HSPCs의 민감성/반응성은 손상된 조직에 풍부한 분자 집합에 의해 긍정적인 영향을 미친다("primed"). 여기에는 생체활성 지질[예를 들면, 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P) 및 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P)], 호중구-유래 양이온성 펩타이드 카세리시딘(cathelicidin)(LL-37), β2-디펜신(defensin) 및 수용성 막 공격 복합체(soluble membrane attack complex; sMAC)(C5b-9)를 포함한다. Some chemokines such as stromal cell derived factor-1 (SDF-1) and growth factors such as vascular endothelial growth-factor (VEGF), platelet- Platelet-derived growth-factor (PDGF) or hepatocyte growth-factor (HGF) plays an important role in the migration and engraftment of adult MSCs. Of these, the SDF-1 and its receptor, the CXCR4 axis, are important factors for homing / engraftment of stem cells. In fact, the engraftment of hematopoietic stem / progenitor cells (HSPCs) according to the SDF-1 gradient in the bone marrow is a prerequisite for transplantation (e.g. systemic irradiation and bone marrow ablation). After chemotherapy) in the bone marrow. The sensitivity / responsiveness of HSPCs to the SDF-1 gradient is positively influenced (“primed”) by a rich set of molecules in the injured tissue. These include bioactive lipids (eg, sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P)), neutrophil-derived cationic peptide caseycides. Cathelicidin (LL-37), β2-defensin and soluble membrane attack complex (sMAC) (C5b-9).
본 발명자들은 HSPCs 과정에서 관찰되는 프라이밍 현상이 S1P 및 CP1 생체활성 지질 및 양이온성 펩타이드인 LL-37로 프라이밍된 MSCs에서도 유사하게 일어난다는 것을 최근에 확인하였다. 특히, 프라이밍된 MSCs는 세포 배양 조건에 있어서 세포 이동, 콜로니-형성 활성 및 항-염증 능력을 촉진시키는데, 이는 폐동맥성 고혈압(pulmonary arterial hypertension; PAH) 치료 효과를 향상시킨다. 하지만, 프라이밍된 MSCs는 여전히 손상된 조직에서 제한된 생체 내 생착을 나타낸다. 더구나, MSC 투여 전 집중적인 세척 과정에도 불구하고, 부정적인 염증 반응을 촉발시키는 잔여 프라이밍 인자들을 완전히 제거하는데 실패하였다. 이에, 저농도에서의 프라이밍 전략 개발이 필요하다. We recently confirmed that the priming phenomena observed in HSPCs process similarly occurs in MSCs primed with LL-37, S1P and CP1 bioactive lipids and cationic peptides. In particular, primed MSCs promote cell migration, colony-forming activity and anti-inflammatory capacity in cell culture conditions, which enhances the effect of treating pulmonary arterial hypertension (PAH). However, primed MSCs still exhibit limited in vivo engraftment in injured tissue. Moreover, despite the intensive washing process prior to MSC administration, it failed to completely eliminate residual priming factors that trigger a negative inflammatory response. Therefore, it is necessary to develop a priming strategy at low concentration.
본 발명의 목적은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 유효성분으로 포함하는 줄기세포 활성 촉진용 조성물, 이를 통한 줄기세포 활성 촉진 방법을 제공하는 데에 있다.Disclosure of Invention An object of the present invention is to provide a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient, and a method for promoting stem cell activity through the same.
또한, 본 발명의 다른 목적은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 고혈압 예방 또는 치료용 약학조성물을 제공하는 데에 있다.Another object of the present invention is to provide a pharmaceutical composition for preventing or treating hypertension, including a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture solution thereof as an active ingredient.
또한, 본 발명의 또 다른 목적은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 염증질환 또는 면역질환 예방 또는 치료용 약학조성물을 제공하는 데에 있다.In addition, another object of the present invention to provide a pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases, including stem cells treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient. Is in.
본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 유효성분으로 포함하는 줄기세포 활성 촉진용 조성물을 제공한다.The present invention provides a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
또한, 본 발명은 분리된 줄기세포에 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 처리하는 단계를 포함하는 줄기세포 활성 촉진 방법을 제공한다. In addition, the present invention provides a method for promoting stem cell activity comprising the step of treating a histone deacetylation inhibitor and priming factor to the isolated stem cells.
또한, 본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 고혈압 예방 또는 치료용 약학조성물을 제공한다.The present invention also provides a pharmaceutical composition for preventing or treating hypertension, comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
또한, 본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 염증질환 또는 면역질환 예방 또는 치료용 약학조성물을 제공한다.In another aspect, the present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
또한, 본 발명은 줄기세포 활성 촉진을 위한 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)의 용도를 제공한다.The present invention also provides the use of histone deacetylation inhibitors and priming factors for promoting stem cell activity.
또한, 고혈압 예방 또는 치료를 위한 약물의 제조에 있어서, 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액의 용도를 제공한다.In addition, in the manufacture of a medicament for preventing or treating hypertension, there is provided the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof.
또한, 염증질환 또는 면역질환 예방 또는 치료를 위한 약물의 제조에 있어서, 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액의 용도를 제공한다.The present invention also provides the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof in the manufacture of a medicament for preventing or treating an inflammatory disease or an immune disease.
본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍 인자를 유효성분으로 포함하는 줄기세포 활성 촉진용 조성물에 관한 것으로서, 줄기세포에 저농도의 히스톤 탈아세틸화 저해제 및 프라이밍 인자를 처리한 경우, 줄기세포의 활성이 탁월하게 향상되었다. 특히, 프라이밍된 MSCs는 세포 배양 조건에 있어서 세포 이동, 콜로니-형성 활성 및 항-염증 능력을 촉진시키는데, 이는 고혈압, 염증질환 또는 면역질환 치료 효과를 향상시키므로, 유용한 치료전략이 될 수 있을 것으로 예상된다.The present invention relates to a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient. When the stem cells are treated with a low concentration of histone deacetylation inhibitor and a priming factor, the activity of the stem cells is increased. Excellent improvement. In particular, primed MSCs promote cell migration, colony-forming activity, and anti-inflammatory capacity in cell culture conditions, which may be useful therapeutic strategies because they enhance the effect of treating hypertension, inflammatory or immunological diseases. do.
도 1은 S1P로 프라이밍된 UC-MSCs에서 5-아자시티딘(5-azacytidine; 5-Aza)의 부정적 영향에 대한 결과를 나타낸다. (A) 1-μM 5-Aza 또는 0.5-mM VPA로 24시간 동안 처리한 인간 UC-MSCs에서의 CXCR4에 대한 RQ-PCR 분석 결과이다. 표시된 유전자들의 상대적 발현 수준은 미처리된 MSCs(NT)의 수치와 비교하여 배율 변화로서 나타냈고, means±SEM (n=4), ***p<0.001, one-way ANOVA test로 나타냈다. (B) 1-μM 5-Aza 및 50-nM S1P로 24시간 동안 프라이밍된 UC-MSCs에서, 표시된 MSC 표면(CD29, CD73 및 CD90) 및 조혈 계통(CD14, CD34, 및 CD45) 단백질들의 발현을 유세포 분석한 결과이다. (C) 5-Aza+S1P로 프라이밍된 UC-MSCs를 사용하여 연골세포, 골세포 또는 지방세포 분화 분석한 대표적인 이미지를 나타낸다. 연골세포 생성, 골세포 생성 및 지방세포 생성은 알시안 블루(Alcian Blue), 알리자린 레드 S(Alizarin Red S) 및 오일 레드 O(Oil Red O) 염색을 통해 각각 측정하였다. 5-Aza+S1P로 24시간 동안 프라이밍된 UC-MSCs의 (D) 세포 증식 (n=3), (E) SDF-1에 대한 화학주성 (n=6) 및 (F) CFU-F 분석 (n≥6) 결과를 나타낸다. 모든 결과는 means±SEM으로 나타냈다. **p<0.01, ***p<0.001, compared to non-primed cells (non-parametric Mann Whitney test). 이동된 세포 (E) 또는 염색된 콜로니 (F)에 대한 대표적인 이미지는 오른쪽 패널에 나타냈다.1 shows the results for the negative effect of 5-azacytidine (5-Aza) on UC-MSCs primed with S1P. (A) RQ-PCR analysis of CXCR4 in human UC-MSCs treated with 1-μM 5-Aza or 0.5-mM VPA for 24 hours. Relative expression levels of the indicated genes were expressed as change in magnification compared to the values of untreated MSCs (NT) and were indicated by means ± SEM (n = 4), *** p <0.001, one-way ANOVA test. (B) Expression of indicated MSC surfaces (CD29, CD73 and CD90) and hematopoietic lineage (CD14, CD34, and CD45) proteins in UC-MSCs primed for 24 hours with 1-μΜ 5-Aza and 50-nM S1P Flow cytometry results. (C) Representative images of chondrocyte, osteocytic or adipocyte differentiation analysis using UC-MSCs primed with 5-Aza + S1P are shown. Chondrocyte production, osteoblast production and adipocyte production were measured through Alcian Blue, Alizarin Red S and Oil Red O staining, respectively. (D) Cell proliferation (n = 3), (E) Chemotaxis (n = 6) and (F) CFU-F analysis of UC-MSCs primed with 5-Aza + S1P for 24 hours (n) n≥6) result is shown. All results are expressed as means ± SEM. ** p <0.01, *** p <0.001, compared to non-primed cells (non-parametric Mann Whitney test). Representative images of migrated cells (E) or stained colonies (F) are shown in the right panel.
도 2는 S1P로 프라이밍된 UC-MSCs에서 VPA의 효과에 대한 결과를 나타낸다. (A 및 B) 0.5-mM VPA 단독 (VPA) 또는 50-nM S1P와의 조합 (VPA+S1P)으로 24시간 동안 프라이밍된 UC-MSCs의 표면 항원 (A) 대한 유세포 분석 결과와 다능성 (B) 분석 결과를 나타낸다. 2 shows the results for the effect of VPA on UC-MSCs primed with S1P. (A and B) Flow cytometry and pluripotency (B) for surface antigen (A) of UC-MSCs primed for 24 hours with 0.5-mM VPA alone (VPA) or in combination with 50-nM S1P (VPA + S1P) The analysis results are shown.
도 3은 VPA+S1P로 프라이밍된 UC-MSCs에서 SDF-1에 대한 향상된 반응성에 대한 결과를 나타낸다. (A 및 B) 0.5-mM VPA 단독 (VPA), 50-nM S1P 단독 (S1P) 또는 50-nM S1P와의 조합 (VPA+S1P)에 24시간 동안 노출된 MSCs의 150-ng/ml SDF-1에 대한 화학주성 분석 결과를 나타낸다. (A) 이동 분석에서 트랜스웰 인셋(insets)의 대표적인 이미지를 나타낸다. (B) 이동에 대한 상대적인 양은 이동된 세포수에 대한 배율 변화로서 정량하였고, means±SEM (n=6)으로 나타냈다. **p<0.01, ***P<0.001 compared to nonprime cells (NT) (one-way ANOVA with Bonferroni post-test). (C) 인산화된-(p-) 또는 전체 AKT 및 MAPKp42 /44의 웨스턴 블랏 분석 결과를 나타낸다. VPA+S1P로 프라이밍된 MSCs는 혈청-결핍 상태에서 12시간 처리한 후 150-ng/ml SDF-1로 표시된 시간 동안 처리되었다.3 shows the results for improved reactivity to SDF-1 in UC-MSCs primed with VPA + S1P. (A and B) 150-ng / ml SDF-1 of MSCs exposed for 24 hours to 0.5-mM VPA alone (VPA), 50-nM S1P alone (S1P) or combination with 50-nM S1P (VPA + S1P) The results of the chemotaxis analysis for. (A) Representative images of transwell insets are shown in the migration assay. (B) Relative amounts for migration were quantified as a change in magnification for the number of migrated cells and expressed as means ± SEM (n = 6). ** p <0.01, *** P <0.001 compared to nonprime cells (NT) (one-way ANOVA with Bonferroni post-test). (C) Western blot analysis of phosphorylated- (p-) or total AKT and MAPK p42 / 44 is shown. MSCs primed with VPA + S1P were treated for 12 hours in serum-deficient state and then for the time indicated as 150-ng / ml SDF-1.
도 4는 S1P로 프라이밍된 UC-MSCs에서 VPA의 효과에 대한 결과를 나타낸다. (A 및 B) VPA, S1P 단독 또는 VPA+S1P로 24시간 동안 프라이밍된 UC-MSCs의 세포 증식 분석 (A, n=3) 및 CFU-F 분석 (B, n=6) 결과를 나타낸다. CFU-F 분석을 위해서, 60개의 세포들을 6-웰 배양 플레이트에 접종하였고, 14일 동안 배양하였으며, 콜로니 수를 정량하였다. 부착 세포들의 대표적인 염색된 콜로니는 오른쪽 패널에 나타냈다. (C) 표시된 세포로부터 수확된 조건 배지(conditioned medium; CM)의 존재 유무에 따라, LPS로 5시간 동안 자극된 쥐 폐포 대식세포주로부터 분비된 TNF-α 단백질 (n=4)의 정량 결과를 나타낸다. 모든 결과는 means±SEM, *p<0.05, **p<0.01, ***p<0.001 compared to nonprime cells (NT), #p<0.05, ###p<0.001 compared to VPA+S1P primed cells one-way ANOVA test로 나타냈다. 4 shows the results for the effect of VPA on UC-MSCs primed with S1P. (A and B) Cell proliferation assay of UC-MSCs primed for 24 hours with VPA, S1P alone or VPA + S1P (A, n = 3) and CFU-F analysis (B, n = 6) Results are shown. For CFU-F analysis, 60 cells were seeded in 6-well culture plates, incubated for 14 days, and colony numbers were quantified. Representative stained colonies of adherent cells are shown in the right panel. (C) shows quantitative results of TNF-α protein (n = 4) secreted from rat alveolar macrophage lines stimulated with LPS for 5 hours, depending on the presence of a conditioned medium (CM) harvested from the indicated cells. . All results are means ± SEM, * p <0.05, ** p <0.01, *** p <0.001 compared to nonprime cells (NT), #p <0.05, ### p <0.001 compared to VPA + S1P primed cells One-way ANOVA test.
도 5는 MSCs의 치료 효과와 관련되어 있는 VPA+S1P 상향 조절된 유전자들을 나타낸다. (A-D) 0.5-mM VPA 단독 (VPA), 50-nM S1P 단독 (S1P) 또는 50-nM S1P와의 조합 (VPA+S1P)으로 24시간 동안 프라이밍된 UC-MSCs에서의 MMP12 (A), 성장인자들 (B), 혈관신생 (C) 및 항-염증 (D) 관련 유전자들의 발현 수준을 나타낸다. 발현 수준은 프라이밍 되지 않은 UC-MSCs(NT)에 대한 프라이밍된 UC-MSCs의 수치 비율로서 계산되었다. 결과는 means±SEM (n≥4); *p<0.05, **p<0.01, ***p<0.001 compared to NT group, #p<0.05, ###p<0.001 compared to VPA+S1P (one-way ANOVA with Bonferroni post-test)으로 나타냈다.5 shows VPA + S1P upregulated genes involved in the therapeutic effect of MSCs. (MP) MMP12 in UC-MSCs primed for 24 h with 0.5-mM VPA alone (VPA), 50-nM S1P alone (S1P) or in combination with 50-nM S1P (VPA + S1P) (A), growth factors (B), angiogenesis (C) and anti-inflammatory (D) related gene expression levels. Expression levels were calculated as the numerical ratio of primed UC-MSCs to unprimed UC-MSCs (NT). The result is means ± SEM (n ≧ 4); * p <0.05, ** p <0.01, *** p <0.001 compared to NT group, #p <0.05, ### p <0.001 compared to VPA + S1P (one-way ANOVA with Bonferroni post-test) Indicated.
도 6은 다양한 VPA 및 S1P 농도에 따른 SDF-1a에 대한 주화성(Chemotaxis) 분석 결과를 나타낸다.6 shows the results of Chemotaxis analysis for SDF-1a according to various VPA and S1P concentrations.
도 7은 HCl-IC 쥐 모델에서 방광 기능 회복과 관련하여, VPA+S1P 프라이밍된 UC-MSC의 치료 가능성 향상을 나타내는 결과이다. (a) 비마취하 방광내압측정술(awake cystometry) 결과를 나타낸다. (b) 대조군(naive) 또는 VPA+S1P 프라이밍된 UC-MSCs(각 그룹당 독립적인 5마리 동물)을 2.5×105 (250 K) 세포수(K = 1,000)로 주입하고 1주일 후의 방광 배뇨 파라미터의 정량적 결과를 나타낸다. IVP; 방광 내압(intravesical pressure), IAP; 복강 내압(intra-abdominal pressure). Sham: 위-수술군(sham-operated). 모든 결과는 means±SEM; *p<0.05, **p<0.01, ***p<0.001 compared to the HCl-IC group; #p<0.05, ##p<0.001, ###p<0.001 compared to the naive UC-MSC group with Bonferroni post-test로 나타냈다. FIG. 7 shows the results of improving the treatability of VPA + S1P primed UC-MSCs with respect to bladder function recovery in the HCl-IC rat model. (a) Results of awake cystometry under non-anesthetic. (b) Bladder urination parameters one week after injection of control or VPA + S1P primed UC-MSCs (5 independent animals per group) with 2.5 × 10 5 (250 K) cell count (K = 1,000) The quantitative results of IVP; Bladder internal pressure, IAP; Intra-abdominal pressure. Sham: Sham-operated. All results are means ± SEM; * p <0.05, ** p <0.01, *** p <0.001 compared to the HCl-IC group; #p <0.05, ## p <0.001, ### p <0.001 compared to the naive UC-MSC group with Bonferroni post-test.
도 8은 HCl-유도된 방광 손상에 있어, UC-MSC 투여 효과에 대한 조직학적 분석 결과를 나타낸다. (a) PBS 또는 250 K 세포수를 투여하고 1주일 후, UC-MSCs HCl-IC 쥐의 방광 조직에서의 (i) 사이토케라틴(cytokeratin) 면역염색(배율 ×40, scale bar = 100 ㎛), (ii) 톨루이딘 블루(Toluidine blue)(배율 ×200, scale bar = 100 ㎛), (iii) 마손 삼색 염색(Masson's trichrome)(배율 ×200, scale bar = 100 ㎛) 및 (iv) TUNEL 분석 (배율 ×400, scale bar = 100 ㎛) 결과를 나타낸다. 화살표는 침윤된 비만 세포를 나타낸다. Sham: 위-수술군(sham-operated). 핵은 Mayer's hematoxylin (i, ii 및 iii) 또는 DAPI (blue, iv)로 염색하였다. (b) 조직학적 실험을 정량화한 결과이다. 데이터는 sham group (n=15)으로부터 표준화하였다. 결과는 mean ± SEM, *p<0.05, **p<0.01, ***p<0.001 compared to the HCl-IC group; #p<0.05, ##p<0.001, ###p<0.001 compared to the naive UC-MSC group with Bonferroni post-test로 나타냈다.8 shows the histological analysis of the effect of UC-MSC administration on HCl-induced bladder injury. (a) 1 week after administration of PBS or 250 K cell number, (i) cytokeratin immunostaining in bladder tissue of UC-MSCs HCl-IC mice (magnification × 40, scale bar = 100 μm), (ii) Toluidine blue (magnification × 200, scale bar = 100 μm), (iii) Masson's trichrome (magnification × 200, scale bar = 100 μm) and (iv) TUNEL analysis (magnification) X400, scale bar = 100 μm) The results are shown. Arrows indicate infiltrated mast cells. Sham: Sham-operated. Nuclei were stained with Mayer's hematoxylin (i, ii and iii) or DAPI (blue, iv). (b) Results of quantification of histological experiments. Data was normalized from sham group (n = 15). The results were mean ± SEM, * p <0.05, ** p <0.01, *** p <0.001 compared to the HCl-IC group; #p <0.05, ## p <0.001, ### p <0.001 compared to the naive UC-MSC group with Bonferroni post-test.
도 9는 MCT 유도된 PAH 쥐 모델에서, UCB-MSCs 및 VPA+S1P 프라이밍된 MSCs (VPA+S1P-MSC)의 효과를 나타낸다. (a) MCT는 RVSP 수준의 증가를 유도하였고, MCT 주입하고 2주 후에 VPA+S1P 프라이밍된 UC-MSC를 투여한 결과 이러한 상승을 상당히 감소시켰다(왼쪽 패널). MCT는 RV/(LV+S)의 무게 비율을 증가시켰고, VPA+S1P-UC-MSC는 상기 효과를 상당히 감소시켰다(오른쪽 패널). (b) MCT는 폐 조직 염증 및 중막(medial wall) 두께 지수(혈관 직경 당 혈관벽 두께)를 증가시켰고, VPA+S1P-MSC는 상기 증가를 상당히 감소시켰다(상부 패널). 프라이밍하지 않은 UCB-MSC는 RV/(LV+S) 및 중막 두께 지수의 개선 효과를 거의 나타내지 않았다. 폐동맥에서 α-SMA+ 평활근 세포의 면역 형광 염색 결과를 나타낸다(하부 패널)(배율 400×). RVSP, 수축기 우심실압(right ventricular systolic pressure); CTL, 대조군; MSC, 인간 탯줄 중간엽 줄기세포(human cord blood mesenchymal stem cell); S1P-MSC, 스핑고신-1-포스페이트 프라이밍된 인간 탯줄 중간엽 줄기세포; RV, 우심실(right ventricle); LV+S, 좌심실 및 심실사이막(interventricular septum). *p<0.05, **p<0.01 compared to MCT alone (one-way ANOVA with Bonferroni post-test). 9 shows the effect of UCB-MSCs and VPA + S1P primed MSCs (VPA + S1P-MSCs) in MCT induced PAH rat model. (a) MCT induced an increase in RVSP levels and significantly reduced this elevation as a result of administration of VPA + S1P primed UC-MSC two weeks after MCT infusion (left panel). MCT increased the weight ratio of RV / (LV + S) and VPA + S1P-UC-MSC significantly reduced this effect (right panel). (b) MCT increased lung tissue inflammation and medial wall thickness index (vascular wall thickness per vessel diameter), and VPA + S1P-MSC significantly reduced this increase (top panel). Unprimed UCB-MSC showed little improvement in RV / (LV + S) and media thickness index. It shows an immunofluorescence staining of α-SMA + smooth muscle cells in the pulmonary artery (lower panel) (magnification × 400). RVSP, right ventricular systolic pressure; CTL, control; MSC, human cord blood mesenchymal stem cell; S1P-MSC, sphingosine-1-phosphate primed human umbilical cord mesenchymal stem cells; RV, right ventricle; LV + S, left ventricle and interventricular septum. * p <0.05, ** p <0.01 compared to MCT alone (one-way ANOVA with Bonferroni post-test).
줄기세포 프라이밍의 주요 표적인 CXCR4의 발현은 DNA-탈메틸화제인 5-아자시티딘(5-azacytidine; 5-Aza) 및 히스톤 탈아세틸화 저해제인 발프로익산(valproic acid; VPA)에 의해 상향 조절된다. 이에 본 발명자들은 MSC 프라이밍을 향상시켜 치료 효과를 촉진시키기 위한 전략에 있어서, 상기 후성유전적(epigenetic) 조절자들의 역할을 확인하고 본 발명을 완성하였다.Expression of CXCR4, a major target of stem cell priming, is upregulated by DNA-demethylating agent 5-azacytidine (5-Aza) and histone deacetylation inhibitor valproic acid (VPA). do. The present inventors have identified the role of the epigenetic modulators in a strategy for promoting MSC priming to promote a therapeutic effect and completed the present invention.
본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 유효성분으로 포함하는 줄기세포 활성 촉진용 조성물을 제공한다. The present invention provides a composition for promoting stem cell activity comprising a histone deacetylation inhibitor and a priming factor as an active ingredient.
바람직하게는, 상기 히스톤 탈아세틸화 저해제는 발프로익산(valproic acid; VPA), 소듐 뷰티레이트(sodium butyrate; NaB), 니코틴아미드(nicotinamide; NAD) 또는 서티놀(sirtinol)일 수 있으나, 이에 제한되는 것은 아니다. Preferably, the histone deacetylation inhibitor may be valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) or sirtinol, but is not limited thereto. It doesn't happen.
바람직하게는, 상기 프라이밍 인자는 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P), 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P), 카세리시딘(cathelicidin; LL-37) 또는 피오글리타존(pioglitazone)일 수 있으나, 이에 제한되는 것은 아니다.Preferably, the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), cathelicidin (LL-37) or It may be a pioglitazone, but is not limited thereto.
바람직하게는, 상기 VPA는 0.1 내지 1.0 mM 농도로 포함될 수 있고, 상기 S1P는 10 내지 50 nM 농도로 포함될 수 있으나, 이에 제한되는 것은 아니다.Preferably, the VPA may be included in a concentration of 0.1 to 1.0 mM, the S1P may be included in a concentration of 10 to 50 nM, but is not limited thereto.
본 명세서에서 사용되는 용어 "프라이밍(Priming)"은 줄기세포의 치료 효능을 증진시키기 위하여 반응성(활성)이 향상되는 현상을 의미하며, 본 발명에서는 상기 프라이밍을 유도하는 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)을 이용하여 해당 줄기세포의 활성을 촉진시킨다.As used herein, the term "priming" refers to a phenomenon in which reactivity (activity) is improved to enhance the therapeutic efficacy of stem cells, and in the present invention, a histone deacetylation inhibitor and a priming factor that induce the priming. (priming factor) to promote the activity of the stem cells.
본 발명에 있어서, "줄기세포 활성"은 줄기세포의 세포 이동성, 세포 증식능, 다능성(연골세포, 골세포 또는 지방세포 계통으로의 생체 외 분화), 콜로니 형성능 또는 항-염증 활성 등을 의미한다. In the present invention, "stem cell activity" refers to cell mobility, cell proliferation ability, pluripotency (in vitro differentiation into chondrocytes, osteocytes or adipocyte lineage), colony forming ability or anti-inflammatory activity of stem cells, etc. .
본 발명에서 활성 촉진 또는 프라이밍 유도는 줄기세포에 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 직접 처리한 경우 줄기세포의 프라이밍이 유도되는 것뿐만 아니라, 상기 프라이밍 처리(전-처리)에 의하여 줄기세포의 활성이 향상된 줄기세포를 이용하여 다른 분화가 유도되는 것도 포함한다.In the present invention, the activity promotion or priming induction is not only induced priming of stem cells when the stem cells are directly treated with histone deacetylation inhibitors and priming factors, but also by the priming treatment (pre-treatment). Other differentiation may be induced by using stem cells with improved stem cell activity.
또한, 상기 활성 촉진용 조성물은 치료를 위한 세포치료제와 혼합하여 생체 내 주입함으로써 세포치료제의 생체 내 효과를 증진시킬 수 있을 뿐만 아니라, 줄기세포 자체에 본 조성물을 처리한 후 기능이 증가된 세포치료제를 생체 내 이식하는 방법으로도 사용될 수 있다.In addition, the composition for promoting activity can be enhanced by injecting in vivo by mixing with a cell therapy agent for treatment, to enhance the in vivo effect of the cell therapy, and after treatment of the present composition to the stem cells themselves cell therapy agent increased function It can also be used as a method for implanting in vivo.
예컨대, 본 발명의 프라이밍은 줄기세포의 세포 이동성, 콜로니 형성능 및 항-염증 활성을 비롯한 줄기세포성에 관련된 기능을 향상시키는 것이다.For example, the priming of the present invention is to enhance stem cell function, including cell mobility, colony forming ability, and anti-inflammatory activity of stem cells.
본 명세서에서 사용되는 용어 "줄기세포"는, 자기복제능력을 가지면서 두 개 이상의 세포로 분화하는 능력을 갖는 세포를 말하며, 만능줄기세포(totipotent stem cell), 전분화능 줄기세포(pluripotent stem cell), 다분화능 줄기세포(multipotent stem cell)로 분류할 수 있다.As used herein, the term "stem cell" refers to a cell having the ability to differentiate into two or more cells while having a self-replicating ability, totipotent stem cells, pluripotent stem cells It can be classified into multipotent stem cells.
본 발명의 줄기세포는 목적에 따라 적절히 제한 없이 선택될 수 있으며, 인간을 포함한 포유동물, 바람직하게는 인간으로부터 유래된 공지된 모든 조직, 세포 등의 성체 세포로부터 유래할 수 있으며, 예를 들어, 골수, 제대혈, 태반(또는 태반 조직세포), 지방(또는 지방조직 세포) 등으로부터 유래할 수 있다.Stem cells of the present invention may be selected without appropriate limitation depending on the purpose, and may be derived from adult cells, such as all known tissues, cells, etc. derived from mammals, including humans, preferably humans, for example, Bone marrow, umbilical cord blood, placenta (or placental tissue cells), fat (or adipose tissue cells) and the like.
예컨대, 상기 줄기세포는 골수, 지방 조직, 근육 조직, ex vivo 배양된 자기조직 간엽 줄기 세포, 동종 이계 간엽 줄기 세포, 제대혈, 배 난황낭, 태반, 제대, 골막, 태아 및 사춘기 피부, 그리고 혈액으로부터 제한없이 얻어지는 줄기세포일 수 있으며, 태아 또는 출생직후 또는 성인으로부터 유래된 줄기세포일 수 있다.For example, the stem cells are restricted from bone marrow, adipose tissue, muscle tissue, ex vivo cultured autologous mesenchymal stem cells, allogeneic mesenchymal stem cells, umbilical cord blood, embryonic yolk sac, placenta, umbilical cord, periosteum, fetal and adolescent skin, and blood It may be a stem cell obtained without, and may be a stem cell derived from the fetus or shortly after birth or adult.
본 발명의 바람직한 구현예에 있어서, 상기 줄기세포는 신경 줄기세포, 간 줄기세포, 조혈 줄기세포, 제대혈 줄기세포, 표피 줄기세포, 위장관 줄기세포, 내피 줄기세포, 근육 줄기세포, 중간엽 줄기세포 및 췌장 줄기세포로 이루어진 군으로부터 선택되며, 보다 바람직하게는 간 줄기세포, 조혈 줄기세포, 제대혈 줄기세포 및 중간엽 줄기세포로 이루어진 군으로부터 선택될 수 있으나, 이에 제한되는 것은 아니다.In a preferred embodiment of the present invention, the stem cells are neural stem cells, liver stem cells, hematopoietic stem cells, cord blood stem cells, epidermal stem cells, gastrointestinal stem cells, endothelial stem cells, muscle stem cells, mesenchymal stem cells and It is selected from the group consisting of pancreatic stem cells, and more preferably may be selected from the group consisting of liver stem cells, hematopoietic stem cells, cord blood stem cells and mesenchymal stem cells, but is not limited thereto.
또한, 본 발명은 분리된 줄기세포에 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 처리하는 단계를 포함하는 줄기세포 활성 촉진 방법을 제공한다.In addition, the present invention provides a method for promoting stem cell activity comprising the step of treating a histone deacetylation inhibitor and priming factor to the isolated stem cells.
바람직하게는, 상기 히스톤 탈아세틸화 저해제는 발프로익산(valproic acid; VPA), 소듐 뷰티레이트(sodium butyrate; NaB), 니코틴아미드(nicotinamide; NAD) 또는 서티놀(sirtinol)일 수 있으나, 이에 제한되는 것은 아니다. Preferably, the histone deacetylation inhibitor may be valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) or sirtinol, but is not limited thereto. It doesn't happen.
바람직하게는, 상기 프라이밍 인자는 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P), 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P), 카세리시딘(cathelicidin; LL-37) 또는 피오글리타존(pioglitazone)일 수 있으나, 이에 제한되는 것은 아니다.Preferably, the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), cathelicidin (LL-37) or It may be a pioglitazone, but is not limited thereto.
바람직하게는, 상기 VPA는 0.1 내지 1.0 mM 농도로 포함될 수 있고, 상기 S1P는 10 내지 50 nM 농도로 포함될 수 있으나, 이에 제한되는 것은 아니다.Preferably, the VPA may be included in a concentration of 0.1 to 1.0 mM, the S1P may be included in a concentration of 10 to 50 nM, but is not limited thereto.
또한, 본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 고혈압 예방 또는 치료용 약학조성물을 제공한다.The present invention also provides a pharmaceutical composition for preventing or treating hypertension, comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
한편, 프라이밍을 통해 줄기세포가 활성화되면 고혈압을 비롯한 심혈관계 질환에 효과를 나타내는데, 프라이밍된 줄기세포의 심혈관계 질환 관련 작용 기작들에 대해서는 여러 보고가 있다(Stem Cell Research & Therapy (2015) 6:218; Stroke. 2011;42:2932-2939; J. Cell. Mol. Med. Vol 17, No 5, 2013 pp.617-625; Stem Cells International 2015 Article ID 685383).On the other hand, the activation of stem cells through priming has an effect on cardiovascular diseases including hypertension, and there are several reports on the mechanisms of cardiovascular disease related priming stem cells (Stem Cell Research & Therapy (2015) 6: 218; Stroke. 2011; 42: 2932-2939; J. Cell. Mol. Med. Vol 17, No 5, 2013 pp. 617-625; Stem Cells International 2015 Article ID 685383).
또한, 본 발명은 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 염증질환 또는 면역질환 예방 또는 치료용 약학조성물을 제공한다.In another aspect, the present invention provides a pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
한편, 프라이밍을 통해 줄기세포가 활성화되면 면역 조절 및 염증 반응에 관여하여 다양한 염증질환 및 면역질환에 응용될 수 있으며, 프라이밍된 줄기세포의 면역 조절 및 염증 반응 관련 작용 기작들에 대해서는 여러 보고가 있다(J Orthop Res. 2016 Apr 6, 1-12; Stem Cell Rev and Rep (2014) 10:351-375; Stem Cells International 2016 Article ID 9364213).On the other hand, when stem cells are activated through priming, they can be applied to various inflammatory diseases and immune diseases by participating in immune regulation and inflammatory response, and there are various reports on the mechanisms of immune control and inflammatory response of primed stem cells. (J Orthop Res. 2016 Apr 6, 1-12; Stem Cell Rev and Rep (2014) 10: 351-375; Stem Cells International 2016 Article ID 9364213).
바람직하게는, 상기 히스톤 탈아세틸화 저해제는 발프로익산(valproic acid; VPA), 소듐 뷰티레이트(sodium butyrate; NaB), 니코틴아미드(nicotinamide; NAD) 또는 서티놀(sirtinol)일 수 있으나, 이에 제한되는 것은 아니다. Preferably, the histone deacetylation inhibitor may be valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) or sirtinol, but is not limited thereto. It doesn't happen.
바람직하게는, 상기 프라이밍 인자는 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P), 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P), 카세리시딘(cathelicidin; LL-37) 또는 피오글리타존(pioglitazone)일 수 있으나, 이에 제한되는 것은 아니다.Preferably, the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), cathelicidin (LL-37) or It may be a pioglitazone, but is not limited thereto.
본 발명에 있어서, "배양액"은 생체 외에서 줄기세포 성장 및 생존을 지지할 수 있게 하는 배지, 상기 배지에 포함된 배양된 줄기세포의 분비물 등을 포함한다. 배양에 사용되는 배지는 줄기세포의 배양에 적절한 당 분야에서 사용되는 통상의 배지를 모두 포함한다. 세포의 종류에 따라 배지와 배양 조건을 선택할 수 있다. 배양에 사용되는 배지는 바람직하게는 세포 배양 최소 배지(cell culture minimum medium: CCMM)로, 일반적으로 탄소원, 질소원 및 미량원소 성분을 포함한다. 이런 세포 배양 최소 배지에는 예들 들어, DMEM(Dulbecco's Modified Eagle's Medium), MEM(Minimal essential Medium), BME(Basal Medium Eagle), RPMI1640, F-10, F-12, αMEM(α Minimal essential Medium), GMEM(Glasgow's Minimal essential Medium), Iscove's Modified Dulbecco's Medium 등이 있으나, 이로 제한되지 않는다.In the present invention, the "culture medium" includes a culture medium capable of supporting stem cell growth and survival in vitro, secretion of cultured stem cells contained in the medium, and the like. The medium used for culturing includes all conventional mediums used in the art suitable for culturing stem cells. Depending on the type of cells, medium and culture conditions can be selected. The medium used for the culturing is preferably a cell culture minimum medium (CCMM), and generally includes a carbon source, a nitrogen source and a trace element component. Such cell culture minimal media include, for example, Dulbecco's Modified Eagle's Medium (DMEM), Minimal Essential Medium (MEM), Basic Medium Eagle (BME), RPMI1640, F-10, F-12, α Minimal Essential Medium (GMEM) (Glasgow's Minimal essential Medium) and Iscove's Modified Dulbecco's Medium, but are not limited to these.
또한 상기 배지는 페니실린(penicillin), 스트렙토마이신(streptomycin), 겐타마이신(gentamicin) 등의 항생제를 포함할 수 있다.In addition, the medium may include antibiotics such as penicillin, streptomycin, gentamicin, and the like.
한편, 본 발명은 상기 줄기세포, 이의 분비물, 배지 성분을 모두 포함하는 형태, 분비물 및 배지성분만을 포함하는 형태, 분비물만을 분리하여 단독으로 또는 줄기세포와 함께 사용하는 형태, 또는 줄기세포만을 투여하여 체내에서 분비물을 생성하는 형태로 사용하는 것도 모두 가능하다.On the other hand, the present invention is the form containing all of the stem cells, their secretions, media components, forms containing only secretions and media components, only secretion to separate or used in combination with stem cells, or by administering only stem cells It is also possible to use it in a form that produces secretions in the body.
상기 줄기세포는 통상적으로 당업계에 공지된 어떠한 방법을 이용하여 획득할 수 있다.The stem cells can be obtained using any method known in the art.
본 발명의 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포는 특정 질환의 치료를 위한 세포치료제로 이용될 수 있으며, 상기 처리는 상기 분자들의 직접적인 처리 또는 전-처리일 수 있다.Stem cells treated with histone deacetylation inhibitors and priming factors of the present invention can be used as a cell therapy for the treatment of certain diseases, and the treatment can be direct or pre-treatment of the molecules. .
상기 "세포치료제"란, 세포와 조직의 기능을 복원하기 위하여 살아 있는 자가(autologous), 동종(allogenic), 이종(xenogenic) 세포를 체외에서 증식, 선별하거나 여타 방법으로 세포의 생물학적 특성을 변화시키는 등 일련의 행위를 통하여 치료, 진단, 예방 목적으로 사용되는 의약품을 의미한다.The term "cell therapy" means proliferating, screening, or otherwise altering the biological properties of cells in vitro to autologous, allogenic, and xenogenic cells to restore the function of cells and tissues. It refers to medicines used for the purpose of treatment, diagnosis and prevention through a series of actions.
상기 세포 치료제는 목적 조직에 도달할 수 있는 한 어떠한 일반적인 경로를 통하여 인체에 투여될 수 있다.The cell therapeutic agent may be administered to the human body via any general route as long as it can reach the desired tissue.
본 발명의 바람직한 구현예에 있어서, 상기 고혈압은 특발성 폐동맥성 고혈압; 가족성 폐동맥성 고혈압; 콜라겐 혈관 질환, 선천성 체폐 단락(短絡), 문맥 고혈압, HIV 감염, 약물 또는 독소와 관련된 폐동맥성 고혈압; 티로이드 장애, 글리코겐 저장 질환, 고셔(Gaucher) 질환, 유전성 출혈성 모세혈관 확장, 혈색소병증, 골수증식장애 또는 비장절제술과 관련된 폐동맥성 고혈압; 폐 모세혈관 혈관종증과 관련된 폐동맥성 고혈압; 신생아의 지속성 폐고혈압; 만성 폐쇄성 폐 질환, 간질성 폐 질환, 저산소증 유도 폐포 저환기 장애, 저산소증 유도 수면장애 호흡 또는 높은 고도에의 만성 노출과 관련된 폐고혈압; 발달 이상과 관련된 폐고혈압; 및 원위 폐동맥의 혈전색전성 폐쇄에 의한 폐고혈압으로 이루어지는 군으로부터 선택되며, 보다 바람직하게는 특발성 폐동맥성 고혈압, 가족성 폐동맥성 고혈압 또는 만성 폐쇄성 폐 질환이고, 가장 바람직하게는 특발성 폐동맥성 고혈압이다.In a preferred embodiment of the present invention, the hypertension is idiopathic pulmonary arterial hypertension; Familial pulmonary arterial hypertension; Pulmonary arterial hypertension associated with collagen vascular disease, congenital pulmonary shortness, portal hypertension, HIV infection, drugs or toxins; Pulmonary hypertension associated with thyroid disorders, glycogen storage disease, Gaucher disease, hereditary hemorrhagic capillary dilatation, hemochromatosis, myeloproliferative disorder or splenectomy; Pulmonary arterial hypertension associated with pulmonary capillary angiomatosis; Persistent pulmonary hypertension in newborns; Pulmonary hypertension associated with chronic obstructive pulmonary disease, interstitial lung disease, hypoxia induced alveolar hypoventilatory disorder, hypoxia induced sleep disorder breathing or chronic exposure to high altitudes; Pulmonary hypertension associated with developmental abnormalities; And pulmonary hypertension by thromboembolic obstruction of distal pulmonary artery, more preferably idiopathic pulmonary arterial hypertension, familial pulmonary arterial hypertension or chronic obstructive pulmonary disease, most preferably idiopathic pulmonary arterial hypertension.
본 발명의 바람직한 구현예에 있어서, 상기 염증질환 또는 면역질환은 골관절염, 류마티스 관절염(Rheumatoid Arthritis), 방광염, 간질성방광염, 천식(Asthma), 피부염(Dermititis), 아토피, 건선(Psoriasis), 낭섬유증(Cystic Fibrosis), 고형장기 이식 후기 및 만성 거부증(Post transplantation late and chronic solid organ rejection), 이식편대숙주질환(graft-versus-host disease), 이식거부질환, 다발성 경화증(Multiple Sclerosis), 전신성 홍반성 루푸스(systemic lupus erythematosus), 쇼그렌 증후군(Sjogren syndrome), 하시모토 갑상선(Hashimoto thyroiditis), 다발성근염(polymyositis), 경피증(scleroderma), 아디슨병(Addison disease), 백반증(vitiligo), 악성빈혈(pernicious anemia), 사구체신염(glomerulonephritis) 및 폐섬유증(pulmonary fibrosis), 염증성장질환(Inflammatory Bowel Diesese), 크론 병(Crohns disease), 자가면역성 당뇨(Autoimmune Diabetes), 당뇨 망막증(Diabetic retinopathy), 비염(Rhinitis), 허혈-재관류 손상(Ischemia-reperfusion injury), 혈관성형술후 재협착(Post-angioplasty restenosis), 만성 폐색성 심장 질환(Chronic obstructive pulmonary disease; COPD), 그레이브병(Graves disease), 위장관 알러지(Gastrointestinal allergy), 결막염(Conjunctivitis), 죽상경화증(Atherosclerosis), 관상동맥질환(Coronary artery disease), 협심증(Angina), 암 전이, 소동맥 질환 또는 미토콘드리아 질환(mitochondrial disease)일 수 있으나, 이에 제한되는 것은 아니다.In a preferred embodiment of the present invention, the inflammatory disease or immune disease is osteoarthritis, rheumatoid arthritis, cystitis, interstitial cystitis, asthma, dermititis, atopy, psoriasis, cystic fibrosis (Cystic Fibrosis), Post transplantation late and chronic solid organ rejection, Graft-versus-host disease, Graft rejection disease, Multiple Sclerosis, Systemic lupus erythematosus Systemic lupus erythematosus, Sjogren syndrome, Hashimoto thyroiditis, polymyositis, scleroderma, Addison disease, vitiligo, pernicious anemia , Glomerulonephritis and pulmonary fibrosis, Inflammatory Bowel Diesese, Crohns disease, Autoimmune Diabetes, Diabetes Diabetic retinopathy, rhinitis, ischemia-reperfusion injury, post-angioplasty restenosis, chronic obstructive pulmonary disease (COPD), Graves disease (Graves disease), gastrointestinal allergy, conjunctivitis, atherosclerosis, coronary artery disease, angina, cancer metastasis, arterial disease or mitochondrial disease May be, but is not limited thereto.
본 발명의 약학 조성물은 유효 성분 이외에 약제학적으로 적합하고 생리학적으로 허용되는 보조제를 사용하여 제조될 수 있으며, 상기 보조제로는 부형제, 붕해제, 감미제, 결합제, 피복제, 팽창제, 윤활제, 활택제 또는 향미제 등의 가용화제를 사용할 수 있다. 본 발명의 약학 조성물은 투여를 위해서 유효 성분 이외에 추가로 약제학적으로 허용 가능한 담체를 1 종 이상 포함하여 의약 조성물로 바람직하게 제제화할 수 있다. 액상 용액으로 제제화되는 조성물에 있어서 허용 가능한 약제학적 담체로는, 멸균 및 생체에 적합한 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. The pharmaceutical compositions of the present invention may be prepared using pharmaceutically suitable and physiologically acceptable auxiliaries in addition to the active ingredients, which may include excipients, disintegrants, sweeteners, binders, coatings, swelling agents, lubricants, lubricants. Or solubilizers such as flavoring agents can be used. The pharmaceutical composition of the present invention may be preferably formulated into a pharmaceutical composition by containing one or more pharmaceutically acceptable carriers in addition to the active ingredient for administration. Acceptable pharmaceutical carriers in compositions formulated in liquid solutions are sterile and physiologically compatible, including saline, sterile water, Ringer's solution, buffered saline, albumin injectable solutions, dextrose solution, maltodextrin solution, glycerol, ethanol and One or more of these components may be mixed and used, and other conventional additives such as antioxidants, buffers and bacteriostatic agents may be added as necessary. Diluents, dispersants, surfactants, binders and lubricants may also be added in addition to formulate into injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like.
본 발명의 의약 조성물의 약제 제제 형태는 과립제, 산제, 피복정, 정제, 캡슐제, 좌제, 시럽, 즙, 현탁제, 유제, 점적제 또는 주사 가능한 액제 및 활성 화합물의 서방출형 제제 등이 될 수 있다. 본 발명의 의약 조성물은 정맥내, 동맥내, 복강내, 근육내, 동맥내, 복강내, 흉골내, 경피, 비측내, 흡입, 국소, 직장, 경구, 안구내 또는 피내 경로를 통해 통상적인 방식으로 투여할 수 있다. 본 발명의 의약 조성물의 유효성분의 유효량은 질환의 예방 또는 치료 요구되는 양을 의미한다. 따라서, 질환의 종류, 질환의 중증도, 조성물에 함유된 유효 성분 및 다른 성분의 종류 및 함량, 제형의 종류 및 환자의 연령, 체중, 일반 건강 상태, 성별 및 식이, 투여 시간, 투여 경로 및 조성물의 분비율, 치료 기간, 동시 사용되는 약물을 비롯한 다양한 인자에 따라 조절될 수 있다. Pharmaceutical formulation forms of the pharmaceutical compositions of the present invention may be granules, powders, coated tablets, tablets, capsules, suppositories, syrups, juices, suspensions, emulsions, drops or injectable solutions and sustained release formulations of the active compounds, and the like. Can be. The pharmaceutical compositions of the present invention may be administered in a conventional manner via intravenous, intraarterial, intraperitoneal, intramuscular, intraarterial, intraperitoneal, sternum, transdermal, nasal, inhalation, topical, rectal, oral, intraocular or intradermal routes. Can be administered. An effective amount of the active ingredient of the pharmaceutical composition of the present invention means an amount required to prevent or treat a disease. Thus, the type of disease, the severity of the disease, the type and amount of the active and other ingredients contained in the composition, the type of formulation and the age, weight, general health, sex and diet, sex and diet, time of administration, route of administration and composition of the patient. It can be adjusted according to various factors including the rate of secretion, the duration of treatment, and the drug used concurrently.
또한, 본 발명은 줄기세포 활성 촉진을 위한 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)의 용도를 제공한다.The present invention also provides the use of histone deacetylation inhibitors and priming factors for promoting stem cell activity.
또한, 고혈압 예방 또는 치료를 위한 약물의 제조에 있어서, 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액의 용도를 제공한다.In addition, in the manufacture of a medicament for preventing or treating hypertension, there is provided the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof.
또한, 염증질환 또는 면역질환 예방 또는 치료를 위한 약물의 제조에 있어서, 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액의 용도를 제공한다.The present invention also provides the use of a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture thereof in the manufacture of a medicament for preventing or treating an inflammatory disease or an immune disease.
이하, 본 발명의 이해를 돕기 위하여 실시예를 들어 상세하게 설명하기로 한다. 다만 하기의 실시예는 본 발명의 내용을 예시하는 것일 뿐 본 발명의 범위가 하기 실시예에 한정되는 것은 아니다. 본 발명의 실시예는 당업계에서 평균적인 지식을 가진 자에게 본 발명을 보다 완전하게 설명하기 위해 제공되는 것이다.Hereinafter, examples will be described in detail to help understand the present invention. However, the following examples are merely to illustrate the content of the present invention is not limited to the scope of the present invention. The embodiments of the present invention are provided to more completely explain the present invention to those skilled in the art.
<실험예>Experimental Example
하기의 실험예들은 본 발명에 따른 각각의 실시예에 공통적으로 적용되는 실험예를 제공하기 위한 것이다.The following experimental examples are intended to provide experimental examples that are commonly applied to each embodiment according to the present invention.
1. 인간 탯줄 유래 MSCs의 배양1. Culture of Human Umbilical Cord-derived MSCs
아산병원의 생명윤리심의위원회에 의해 승인된 가이드라인에 따라 부모의 서면 동의를 받아, 건강한 정상 만삭 신생아로부터 인간 UC를 얻었다. UC 수집 전에 모든 산모로부터 동의서를 받았다. UC-유래 MSCs(UC-derived MSCs; UC-MSCs)는 2-mM L-글루타민, 20-mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) (pH 7.3), 최소-필수 배지(minimum-essential medium; MEM) 비필수 아미노산 용액, 페니실린/스트렙토마이신(Corning Cellgro, Pittsburgh, PA), 1-mg/ml 아스코르빅산(Sigma-Aldrich), 10% 열-불활성화된 우태아혈청(fetal bovine serum; FBS) (HyClone), 5-ng/mL 인간 상피세포 성장인자(Sigma-Aldrich, St. Louis, MO), 10-ng/ml 기본 섬유아세포 성장인자 및 50-mg/ml long-R3 인슐린 유사 성장인자-1(ProSpec, Rehovot, Israel)가 첨가된 low-glucose Dulbecco's modified Eagle's medium (DMEM) (HyClone, Pittsburgh, PA)에서 5% CO2 대기 조건, 37℃로 배양하였다. 다능성을 유지하기 위해서 5번 이하로 계대하여 확장한 UC-MSCs을 사용하였다. 표면 단백질의 발현은 이전에 보고한 대로 분석하였다(Stem Cells and Dev. 24 (2015) 1658-1671). Human UCs were obtained from healthy normal term newborns with written informed parental consent in accordance with guidelines approved by the Bioethics Review Board of Asan Hospital. All mothers received informed consent prior to UC collection. UC-derived MSCs (UC-MSCs) are 2-mM L-glutamine, 20-mM 4- (2-hydroxyethyl) -1-piperazineethanesulfonic acid (HEPES) (pH 7.3), minimal-required medium ( minimum-essential medium (MEM) non-essential amino acid solution, penicillin / streptomycin (Corning Cellgro, Pittsburgh, PA), 1-mg / ml ascorbic acid (Sigma-Aldrich), 10% heat-inactivated fetal bovine serum ( fetal bovine serum; FBS) (HyClone), 5-ng / mL human epidermal growth factor (Sigma-Aldrich, St. Louis, MO), 10-ng / ml basic fibroblast growth factor and 50-mg / ml long- R3 insulin-like growth factor -1 (ProSpec, Rehovot, Israel) are added to the low-glucose Dulbecco's modified Eagle's medium (DMEM) (HyClone, Pittsburgh, PA) 5% CO 2 at atmospheric conditions, were incubated in 37 ℃. In order to maintain pluripotency, UC-MSCs extended to 5 times or less were used. Expression of surface proteins was analyzed as previously reported (Stem Cells and Dev. 24 (2015) 1658-1671).
2. 세포 이동 분석2. Cell migration assay
8-μm 폴리카보네이트 막을 1.0% 젤라틴(Sigma) 50 ㎕로 1 시간 동안 코팅하였다. UC-MSCs는 VPA (0.5-mM; Sigma-Aldrich) 또는 5-Aza (1-μM; Sigma-Aldrich) 단독 또는 50-nM S1P와 혼합하여 1일 동안 프라이밍시켰다. UC-MSCs는 TrypLE solution (Thermo Scientific, Pittsburgh PA)으로 분리시킨 후, 세척하고 0.5% bovine serum albumin(BSA)를 함유한 DMEM에 재현탁하여, 3 X 104 세포/웰 밀도로 트렌스웰 인서트(Transwell inserts)(Corning Costar, Pittsburgh, PA)의 상부 챔버에 접종하였다. 하부 챔버에는 0.5% BSA를 함유한 DMEM에 넣은 150-ng/ml SDF-1 (R&D Systems)로 채웠다. 1일 후, 트랜스웰 플레이트로부터 인서트를 제거하였다. 상부 챔버에 남아있는 세포들을 탈지면으로 긁어냈고, 이동한 세포들을 포스페이트 완충된 식염수(phosphate-buffered saline; PBS)에 녹인 4% 파라포름알데히드(paraformaldehyde; PFA) 용액으로 고정한 후, 0.5 % 크리스탈 바이올렛(Sigma-Aldrich)으로 염색하였다. Image Pro 5.0 software (Media Cybernetics, Rockville, MD, USA)로 디지털 이미지를 분석하여, 막의 하부면에 있는 염색된 세포들을 정량하였다. An 8-μm polycarbonate membrane was coated with 50 μl of 1.0% gelatin (Sigma) for 1 hour. UC-MSCs were primed for 1 day with VPA (0.5-mM; Sigma-Aldrich) or 5-Aza (1-μM; Sigma-Aldrich) alone or mixed with 50-nM S1P. UC-MSCs are TrypLE solution (Thermo Scientific, Pittsburgh PA) as was separated, washed and resuspended in DMEM containing 0.5% bovine serum albumin (BSA) , 3 X 10 4 trans-well inserts with cells / well density ( Transwell inserts) (Corning Costar, Pittsburgh, Pa.) Were inoculated into the upper chamber. The lower chamber was filled with 150-ng / ml SDF-1 (R & D Systems) in DMEM containing 0.5% BSA. After 1 day, the inserts were removed from the transwell plates. Cells remaining in the upper chamber were scraped with cotton wool, and the migrated cells were fixed in 4% paraformaldehyde (PFA) solution dissolved in phosphate-buffered saline (PBS), followed by 0.5% crystal violet ( Sigma-Aldrich). Digital images were analyzed with Image Pro 5.0 software (Media Cybernetics, Rockville, MD, USA) to quantify the stained cells on the underside of the membrane.
3. MSCs의 생체 외(in vitro) 특성 분석3. In vitro Characterization of MSCs
MSCs의 세포 증식, 섬유아세포 콜로니-형성 단위(colony-forming unit-fibroblast; CFU-F), 다능성(연골세포, 골세포 또는 지방세포 계통으로의 생체 외 분화) 및 생체 외 항염증 분석은 이전에 보고한 대로 수행하였다(Stem Cells and Dev. 24 (2015) 1658-1671). Cell proliferation of MSCs, colony-forming unit-fibroblast (CFU-F), pluripotency (in vitro differentiation into chondrocytes, osteocytes or adipocyte lineages) and in vitro anti-inflammatory assays This was done as reported in Stem Cells and Dev. 24 (2015) 1658-1671.
4. 웨스턴 블랏 및 실시간 정량적 PCR(real-time quantitative PCR; RQ-PCR)4. Western blot and real-time quantitative PCR (RQ-PCR)
50-nM S1P 및 0.5-mM VPA로 프라이밍된 UC-MSCs는 0.5% BSA를 함유하는 DMEM에서 37℃로 12시간 동안 결핍(starvation)시켰고, 150-ng/ml SDF-1으로 5, 10, 20 또는 30분 동안 자극한 후, 30㎍-세포 추출물을 사용하여 미토젠-활성화된 단백질 키나아제(MAPK)p42/44 및 AKT (Ser473)의 인산화를 웨스턴 블랏을 통해 분석하였다. 유전자 발현 분석을 위해, 표시된 세포로부터 얻은 전체 RNA는 역전사시켰고, 표시된 전사체는 RQ-PCR을 통해 정량하였다. UC-MSCs primed with 50-nM S1P and 0.5-mM VPA were starvated for 12 hours at 37 ° C. in DMEM containing 0.5% BSA and 5, 10, 20 with 150-ng / ml SDF-1. Or after 30 min of stimulation, phosphorylation of mitogen-activated protein kinase (MAPK) p42 / 44 and AKT (Ser473) using a 30 μg-cell extract was analyzed via Western blot. For gene expression analysis, total RNA from indicated cells was reverse transcribed and the indicated transcripts were quantified via RQ-PCR.
5. 통계 분석5. Statistical Analysis
통계적 유의성 차이를 확인하기 위해서, 데이터는 non-parametric Mann Whitney test 또는 one-way ANOVA with the Bonferroni post-hoc test를 사용하여 분석하였다. 본 발명자들은 GraphPad Prism 6.0 software (GraphPad Software, La Jolla, CA)를 사용하여, 모든 분석을 수행하였고, 통계적 유의성은 p<0.05, 0.01, 또는 0.001로 정의하였다.To identify statistical significance differences, the data were analyzed using a non-parametric Mann Whitney test or one-way ANOVA with the Bonferroni post-hoc test. We performed all analyzes using GraphPad Prism 6.0 software (GraphPad Software, La Jolla, Calif.), And statistical significance was defined as p <0.05, 0.01, or 0.001.
6. 실험 디자인6. Experimental design
본 발명의 목적은 쥐 모델에서 간질성 방광염/방광통증증후군(Interstitial cystitis/bladder pain syndrome; IC/BPS)을 치료하기 위해, V1P+S1P로 프라이밍된 인간 탯줄 유래 MSCs(umbilical cord-derived MSCs; UC-MSCs)의 효과를 측정하는 것 뿐만 아니라 이식된 세포의 생체 내(in vivo) 세포적 특성도 광범위하게 관측하고자 하였다. 대조군 또는 V1P+S1P 프라이밍된 UC-MSCs를 방광이 손상된 쥐에 투여하였고, 방광 배뇨 기능, 요로 상피 박리(urothelium denudation), 비만 세포 침윤, 조직 섬유화, 세포 사멸(apoptosis) 및 종양 형성에 대한 영향을 측정하였다. 모든 실험에 있어, 그룹당 5마리의 독립적인 동물에 대해 2번의 독립적인 실험을 수행하였다. 이들은 손상 그룹, 세포 이식 또는 비히클(vehicle) 투여 그룹 및 방광내압측정술(Cystometry) 그룹으로 임의로 배분하여 처리하였다. 수술 과정에 관여한 연구자에게는 투여된 세포의 형태 및 투여량에 대한 정보를 알리지 않았다. 모든 방광내압측정, 조직학 및 유전자 발현 측정은 처리군에 대한 정보를 알지 못하는 연구자에 의해 수행되었다. 방광 손상 또는 카테터 삽입에 의해 예상치 않게 죽은 동물들은 모든 분석에서 모두 제외하였다. 모든 동물 실험은 울산대학교 의과대학의 동물실험윤리위원회의 가이드라인 및 규정에 따라 수행하였다(IACUC-2014-14-167).An object of the present invention is to develop an umbilical cord-derived MSCs derived from human umbilical cord-derived MSCs primed with V1P + S1P to treat interstitial cystitis / bladder pain syndrome (IC / BPS) in a rat model. In addition to measuring the effects of -MSCs), the in vivo cellular properties of the transplanted cells were also widely observed. Control or V1P + S1P primed UC-MSCs were administered to rats with bladder injuries and affected the effects on bladder urination function, urothelium denudation, mast cell infiltration, tissue fibrosis, apoptosis and tumor formation. Measured. For all experiments, two independent experiments were performed on 5 independent animals per group. They were randomly assigned to the injured group, cell transplant or vehicle administration group, and cystometry group for treatment. Researchers involved in the surgical procedure were not informed of the type and dosage of cells administered. All bladder pressure measurements, histology and gene expression measurements were performed by researchers who did not know information about treatment groups. All animals unexpectedly killed by bladder injury or catheter insertion were excluded from all analysis. All animal experiments were conducted in accordance with the guidelines and regulations of the Animal Experimental Ethics Committee of the University of Ulsan College of Medicine (IACUC-2014-14-167).
7. 동물 모델 및 UC-MSCs의 이식7. Transplantation of Animal Models and UC-MSCs
HCl 주입 IC/BPS 쥐 모델은 이전에 보고되었다(Stem Cells and Development 24, 2015, 1648-1657). HCl 손상 일주일 후, 하복부를 절개하였고, 500 ㎛ 실린지 및 26-게이지 바늘을 이용하여, 2.5×105 (250 K) UC-MSCs 또는 PBS를 방광의 전벽 바깥층 및 천장에 직접 투여하였다. 줄기세포 투여 하루 전부터, Wnt 또는 IGF-매개 신호전달을 차단하기 위해서 인도메타신(indomethacin; PMG Pharm Co., Ltd. Ansan, Korea; every 12 h at 2.5mg/kg) 또는 제피티닙(Gefitinib; Santa Cruz Biotechnology, Santa Cruz, CA, USA; every day at 5 mg/kg)을 각각 피하 주입하였다.HCl infused IC / BPS rat models have been previously reported (Stem Cells and Development 24, 2015, 1648-1657). HCl week after injury, the abdomen was cut, using a 500 ㎛ syringe and 26-gauge needle and a 2.5 × 10 5 (250 K) UC-MSCs or PBS was administered directly to the front wall outer layer and the ceiling of the bladder. From one day prior to administration of stem cells, indomethacin (PMG Pharm Co., Ltd. Ansan, Korea; every 12 h at 2.5 mg / kg) or gefitinib (Gefitinib; to block Wnt or IGF-mediated signaling); Santa Cruz Biotechnology, Santa Cruz, CA, USA; every day at 5 mg / kg) was injected subcutaneously.
8. 8. 비마취Non-anesthetic (( UnanesthetizedUnanesthetized ) 및 ) And 비억제Non-inhibition (unrestrained) (unrestrained) 방광내압측정도Bladder breakdown voltage chart 수집(비마취하 방광내압측정술) Acquisition (non-anesthetic bladder pressure measurement)
방광내압측정(Cystometrograms)은 대사 케이지 내의 비마취 및 비억제된 쥐에서 수행되었다. 방광 내압(intravesical pressure; IVP) 및 복강 내압(intra-abdominal pressure; IAP) 기록 측정을 위해, 방광내압측정 3일 전에 카테터를 동시에 삽입하였다. 간단히 설명하면, 마취 유도 후, 복강 절개를 통해 커프(cuff)가 달린 폴리에틸렌 카테터(PE-50; Becton-Dickinson, Parsippany, NJ, USA)를 방광 천장에 삽입하였다. IAP를 기록하기 위해서, 카테터 팁의 커프 부근 복강 벌룬(Latex; Daewoo Medical, Incheon, Korea)을 방광 근위부에 위치시켰고, 실크 스레드(thread)가 달린 다른 카테터에 묶었다. 폴리에틸렌 카테터(PE-50)를 따뜻한 물에서 가열하였고, 삽입부의 팁을 원래 길이의 ~ 1.5배 정도 늘렸고, 헤파린 처리한 식염수(100 IU/mL)로 채웠다. 방광 카테터가 이식되면, 늘어난 카테터가 대퇴정맥 내로 삽입되었다. 그 후, 상기 카테터들은 피하 공간을 통해 뚫고 지나갔고, 동물의 등을 통해 빠져나와, 등의 피부에 부착되었다. 수술 후, 각각의 쥐들은 개별적으로 사육되었고, 같은 방식으로 유지되었다.Cystometrograms were performed in non-anesthesia and non-suppressed mice in metabolic cages. Catheter was inserted simultaneously 3 days prior to bladder pressure measurement for measurement of intravesical pressure (IVP) and intra-abdominal pressure (IAP) recordings. Briefly, after anesthesia induction, a cuffed polyethylene catheter (PE-50; Becton-Dickinson, Parsippany, NJ, USA) was inserted into the bladder ceiling via an intraperitoneal incision. To record the IAP, an abdominal balloon (Latex; Daewoo Medical, Incheon, Korea) near the cuff of the catheter tip was placed proximal to the bladder and tied to another catheter with a silk thread. The polyethylene catheter (PE-50) was heated in warm water and the tip of the insert was extended to ~ 1.5 times its original length and filled with heparinized saline (100 IU / mL). When the bladder catheter was implanted, the extended catheter was inserted into the femoral vein. The catheter then penetrated through the subcutaneous space, exited through the back of the animal, and attached to the back skin. After surgery, each rat was raised individually and maintained in the same way.
비마취하 방광내압측정 분석(awake cystometric analysis)을 위하여, pressure transducer (Research Grade Blood Pressure Transducer; Harvard Apparatus, Holliston, MA, USA) 및 microinjection pump (PHD22/2000 pump; Harvard Apparatus)에 연결된 T-튜브를 통하여 2중 밸브로 방광에 유치 카테터(indwelling catheter)를 연결하였다. IAP를 기록하기 위해서, 체액이 채워진 복강 벌룬이 연결된 다른 유치 카테터는 다른 pressure transducer에 연결하였다. 멸균된 식염수를 방광 내에 0.4 mL/min의 속도로 주입하고, force displacement transducer (Research Grade Isometric Transducer; Harvard Apparatus)가 연결된 체액 수집기를 통해 배뇨량을 계속해서 기록하였다. 50 Hz의 샘플링 속도로 Acq Knowledge 3.8.1 software가 장착된 MP150 data acquisition system(Biopac Systems, Goleta, CA, USA)을 사용하여, IVP, IAP 및 배뇨량을 계속하여 기록하였다. 각 동물로부터 8분 동안 측정된 모든 배뇨 주기 수치를 평가에 사용하였다. For awake cystometric analysis, a T-tube connected to a pressure transducer (Research Grade Blood Pressure Transducer; Harvard Apparatus, Holliston, Mass., USA) and a microinjection pump (PHD22 / 2000 pump; Harvard Apparatus) An indwelling catheter was connected to the bladder via a double valve. To record the IAP, another indwelling catheter with a fluid-filled abdominal balloon was connected to another pressure transducer. Sterile saline was injected into the bladder at a rate of 0.4 mL / min and urine volume was continuously recorded through a fluid collector connected to a force displacement transducer (Research Grade Isometric Transducer; Harvard Apparatus). IVP, IAP and urination volume were continuously recorded using an MP150 data acquisition system (Biopac Systems, Goleta, CA, USA) equipped with Acq Knowledge 3.8.1 software at a sampling rate of 50 Hz. All urination cycle values measured for 8 minutes from each animal were used for the evaluation.
소변 배출 없이, 기준치로부터 15 cmH2O를 넘게 IVP가 증가하면, 비배뇨 수축(non-voiding contraction; NVC)으로 측정하였다. BP는 방광이 채워지는 동안에 최저 방광 압력을 의미하고, MP는 배뇨 주기 동안에 최고 방광 압력을 의미하며, MV는 소변 배출시 소변량을 의미하고, RV는 소변 배출 후 잔여 소변량을 의미한다. BC는 MV + RV로서 정의되고, MI는 배뇨 수축 사이의 간격을 의미한다. Without urine drainage, an increase in IVP above 15 cmH 2 O from baseline was measured by non-voiding contraction (NVC). BP means the lowest bladder pressure while the bladder is filled, MP means the highest bladder pressure during the urination cycle, MV means the urine volume during urine drainage, and RV means the amount of urine remaining after urine drainage. BC is defined as MV + RV, MI means the interval between urination contractions.
9. 폐동맥 고혈압(Pulmonary artery hypertension; PAH) 동물 모델 9. Pulmonary artery hypertension (PAH) animal model
모노크로탈린(monocrotaline; MCT) 유도된 PAH 쥐 모델은 이전 보고에 따라 확립하였다(Stem Cells and Development 24, 2015, 1658-1671). 수컷 특정-병원균-프리 루이스 쥐(8 주령, 250-280 g)는 사육 온도 및 빛(12시간 명암 주기)의 조절하에서, 음식 및 물의 제한 없이 사육되었다. MCT (60 mg/kg, Sigma)의 피하 투여를 통해 PAH를 유도하였다. 대조군의 쥐들은 동일 부피의 PBS를 투여하였다. MCT 또는 PBS 투여 2주 후, 프라이밍 유도하지 않은 UC-MSCs 또는 VPA+S1P로 전처리한 UC-MSCs를 200 ㎕ PBS 당 2.5×105 세포 밀도로 꼬리 정맥을 통해 투여하였다. 용매만 처리한 대조군(vehicle control)에는 세포 없이 200 ㎕ PBS를 투여하였다. 주입 전, 세포들을 따뜻한 PBS로 2번 씻어냈고, 7-AAD (BD Biosciences) 배제 염색을 통한 FACS 분석을 이용하여 투여한 세포의 생존능을 관측하였다. Monocrotaline (MCT) induced PAH rat models were established according to previous reports (Stem Cells and Development 24, 2015, 1658-1671). Male specific-pathogen-free Lewis rats (8 weeks old, 250-280 g) were reared without restriction of food and water, under the control of breeding temperature and light (12 hour contrast cycle). PAH was induced through subcutaneous administration of MCT (60 mg / kg, Sigma). Mice in the control group received the same volume of PBS. Two weeks after MCT or PBS administration, priming-induced UC-MSCs or UC-MSCs pretreated with VPA + S1P were administered via the tail vein at a density of 2.5 × 10 5 cells per 200 μl PBS. A solvent-only vehicle control was administered with 200 μl PBS without cells. Prior to injection, cells were washed twice with warm PBS and viability of the administered cells was observed using FACS analysis via 7-AAD (BD Biosciences) exclusion staining.
10. 수축기 10. Systolic 우심실압Right ventricular pressure (right ventricular systolic pressure; (right ventricular systolic pressure; RVSPRVSP ) 및 우심실 비대증(right ventricular hypertrophy; RVH) 측정) And right ventricular hypertrophy (RVH) measurements
MCT 또는 PBS 투여 4주 후, 인공호흡기(Harvard Apparatus, Holliston, MA)를 통한 호흡 유지와, 졸레틸(zoletil; 40 mg/kg) 및 럼푼(rompun; 10 mg/kg)에 의한 마취 하에서, 환자 모니터(MDE Escort II patient monitor, Arleta)가 연결된 26G 바늘을 사용한 횡격막의 직접 천자를 통해 수축기 우심실압을 측정하였다. 우심실압(Right ventricular pressure; RVP)은 인공호흡기를 통해 호흡을 유지시켰다. RVH를 측정하기 위해, 심장의 우심실을 심실사이막(interventricular septum)으로부터 분리시켰고, 우심실(RV) 및 심실사이막을 포함하는 좌심실(LV+S)의 무게를 측정하였다.  Four weeks after MCT or PBS administration, patients under breathing maintenance through a ventilator (Harvard Apparatus, Holliston, MA) and under anesthesia with zoletil (40 mg / kg) and rompun (10 mg / kg) The systolic right ventricular pressure was measured by direct puncture of the diaphragm using a 26G needle connected to an MDE Escort II patient monitor (Arleta). Right ventricular pressure (RVP) maintained breathing through the ventilator. To measure RVH, the right ventricle of the heart was isolated from the interventricular septum and the weight of the left ventricle (LV + S), including the right ventricle (RV) and the ventricular septum, was measured.
11. 조직학 및 유전자 발현 분석 11. Histology and Gene Expression Analysis
방광 조직의 조직학적 분석을 위하여, 사이토케라틴에 대한 면역염색을 통해 상피세포 박리, 톨루이딘 블루 염색(Toluidine blue staining; 8544-4125; Daejung Chemicals & Metals, Seoul, Korea)을 통해 비만세포 침윤, 마손 삼색 염색(Masson's trichrome staining; Junsei Chemical, Tokyo, Japan)을 통해 조직 섬유화 및 TUNEL 염색(1 684 795; Roche, Mannheim, Germany)을 통해 세포사멸을 측정하였다. 폐 조직에 대해서는, H&E- 또는 α-평활근 액틴(α-smooth muscle actin; α-SMA)-염색된 부위에서 4 ㎛ 두께 및 25-100 ㎛의 직경을 가진 임의로 선택된 혈관의 임의로 선택된 영역에 대해 적어도 5번 이상, 400×배율로 캡쳐하였다. 항-α-SMA (1:100, Abcam)를 제조사의 추천 프로토콜에 따라 적용하였고, 반응시켰다. 핵은 4',6'-디아미노-2-페닐인돌(4',6'-diamino-2-phenylindole; D9542; DAPI, Sigma-Aldrich)로 대비염색하였다. NIH ImageJ program (http://rsbweb.nih.gov/ij/)을 사용하여, 중막(Medial wall) 두께를 측정하였다. 중막 두께 지수는 외부 직경에 대한 외부 직경-내부 직경의 비율로 정의된다.For histological analysis of bladder tissues, epithelial cell detachment via cytokeratin immunostaining, mast cell infiltration and torsion tricolor through toluidine blue staining (8544-4125; Daejung Chemicals & Metals, Seoul, Korea) Apoptosis was measured through tissue fibrosis and TUNEL staining (1 684 795; Roche, Mannheim, Germany) via staining (Masson's trichrome staining; Junsei Chemical, Tokyo, Japan). For lung tissue, at least for an arbitrarily selected region of randomly selected vessels having a thickness of 4 μm and a diameter of 25-100 μm at H & E- or α-smooth muscle actin (α-SMA) -stained sites More than 5 times, capture at 400 × magnification. Anti-α-SMA (1: 100, Abcam) was applied according to the manufacturer's recommended protocol and reacted. Nuclei were counterstained with 4 ', 6'-diamino-2-phenylindole (4', 6'-diamino-2-phenylindole; D9542; DAPI, Sigma-Aldrich). Medial wall thickness was measured using the NIH ImageJ program (http://rsbweb.nih.gov/ij/). The media thickness index is defined as the ratio of the outer diameter to the inner diameter to the outer diameter.
유전자 발현 분석을 위해, 전체 RNA는 RNeasy Mini Kit (Qiagen Inc., Valencia, CA)을 사용하여 준비하였고, TaqMan Reverse Transcription Reagents (Applied Biosystems)를 사용하여 역전사를 수행하였고, PikoReal Real-Time PCR System (Thermo Scientific) 및 iQ SYBR Green PCR Master Mix (Bio-Rad, Hercules, CA)를 사용하여 실시간 정량 PCR(real-time quantitative PCR; RQ-PCR)을 수행하였다. 처리군 당 5 마리의 독립적인 동물에 있어, 각 슬라이드로부터 임의로 선택된 3부분의 영역(n=15)은 디지털 이미지를 정량하는데 사용되었다. 유사하게, 그룹당 임의로 선택된 5 마리 동물로부터 RQ-PCR을 2번 반복하여(n=10), 유전자 발현 데이터를 얻었다.For gene expression analysis, total RNA was prepared using the RNeasy Mini Kit (Qiagen Inc., Valencia, CA), reverse transcription was performed using TaqMan Reverse Transcription Reagents (Applied Biosystems), and PikoReal Real-Time PCR System ( Thermo Scientific) and iQ SYBR Green PCR Master Mix (Bio-Rad, Hercules, Calif.) Were used to perform real-time quantitative PCR (RQ-PCR). For five independent animals per treatment group, a randomly selected three-part area (n = 15) from each slide was used to quantify the digital image. Similarly, RQ-PCR was repeated twice (n = 10) from five randomly selected animals per group to obtain gene expression data.
<실시예 1> S1P에 의한 인간 UC-MSCs 프라이밍에 있어 5-Aza의 영향Example 1 Influence of 5-Aza on Priming of Human UC-MSCs by S1P
5-Aza 및 VPA의 저농도 처리(1-μM 및 0.5-mM)는 UC-MSCs에서 CXCR4의 발현을 매우 증가시킨다(도 1A). 다음으로, 본 발명자들은 UC-MSCs의 기본 특징에 대한 상기 후성유전적(epigenetic) 조절자의 효과를 확인하였다. 50-nM S1P의 프라이밍과는 독립적으로, 5-Aza 및 VPA는 모두 표면 마커 단백질(CD29, CD73 및 CD90 양성, CD34 및 CD45 음성)의 발현에는 그다지 영향을 미치지 않았다(도 1B 및 도 2A). 또한, 5-Aza (5-Aza+S1P) 또는 VPA (VPA+S1P)를 함께 처리한 S1P 프라이밍은 다계통 분화 능력에는 거의 영향을 미치지 않았는데, 다계통 분화 능력은 글리코스아미노글리칸스(glycosaminoglycans; 알시안 블루), 무기질 침착(mineral deposition; 알리자린 레드 S) 및 지질 축적(lipid accumulation; 오일 레드 O 염색)의 수준 증가를 각각 측정함으로써, 연골세포, 골세포 및 지방세포 계통의 in vitro 분화 분석를 기초로 하였다(도 1C 및 도 2B). Low concentration treatment (1-μM and 0.5-mM) of 5-Aza and VPA greatly increased the expression of CXCR4 in UC-MSCs (FIG. 1A). Next, the inventors confirmed the effect of the epigenetic modulator on the basic characteristics of UC-MSCs. Independent of the priming of 50-nM S1P, both 5-Aza and VPA did not significantly affect the expression of surface marker proteins (CD29, CD73 and CD90 positive, CD34 and CD45 negative) (FIG. 1B and FIG. 2A). In addition, S1P priming treated with 5-Aza (5-Aza + S1P) or VPA (VPA + S1P) had little effect on the multi-system differentiation capacity, but the multi-system differentiation capacity was determined by glycosaminoglycans; Based on in vitro differentiation analysis of chondrocytes, osteocytes and adipocyte lineages by measuring levels of alcian blue), mineral deposition (Alizarin Red S) and lipid accumulation (oil red O staining), respectively. (FIG. 1C and FIG. 2B).
CXCR4의 상향 조절과는 모순되게(도 1A), 트랜스웰 이동 분석에 있어서 5-Aza+S1P로 프라이밍된 UC-MSCs는 SDF-1 반응에 대한 이동 활성이 프라이밍되지 않은 세포보다 약 30% 정도 감소하였다(도 1E). 또한, 5-Aza+S1P 프라이밍은 클론원성(clonogenic) 전구세포로 자가재생되는 빈도를 나타내는 클론원성 CFU-F의 활성을 심하게 손상시켰다(도 1F). MSCs의 세포 증식은 5-Aza+S1P의 프라이밍에 의해 거의 변화가 없었다(도 1D). Contrary to upregulation of CXCR4 (FIG. 1A), UC-MSCs primed with 5-Aza + S1P in transwell migration assays reduced migration activity for SDF-1 responses by about 30% compared to unprimed cells. (FIG. 1E). In addition, 5-Aza + S1P priming severely impaired the activity of clonal CFU-F, which indicates the frequency of self-renewal into clonal progenitor cells (FIG. 1F). Cell proliferation of MSCs was little changed by the priming of 5-Aza + S1P (FIG. 1D).
<실시예 2> 저농도의 S1P 처리시 UC-MSCs 프라이밍을 강화시키는 VPAExample 2 VPA Enhancing the Priming of UC-MSCs at Low S1P Treatment
다음으로, 본 발명자들은 저농도의 S1P 처리시 UC-MSCs의 프라이밍에 VPA가 미치는 영향을 조사하였다. 이를 위해, 본 발명자들은 지방- 및 UCB-유래 MSCs의 프라이밍을 위해 사용되는 최적 용량(200-nM) 보다 4배 적은 50-nM S1P를 적용하였다. VPA 단독 처리와 S1P 단독 처리시와는 다르게, VPA+S1P 처리시 UC-MSCs 프라이밍은 SDF-1에 대한 화학주성(chemotactic activity)을 프라이밍 되지 않은 세포보다 2~3배 높게 증가시켰다(도 3A 및 도 3B). VPA+S1P 프라이밍에 의해 강화된 SDF-1에 대한 반응은 5-Aza+S1P 프라이밍의 경우와는 완전히 다르게 나타났다. 다음으로, 본 발명자들은 HSPCs의 이동과 관련된 신호전달 경로의 상태를 확인하였다. 화학주성 분석 결과와 일치하게, VPA+S1P에 노출된 UC-MSCs는 MAPKp42 /44 및 AKT 단백질의 인산화를 증가시켰는데, 이는 신호전달 경로의 활성화를 나타낸다(도 3C). 상기 결과는 5-Aza와는 다르게 VPA가 저농도의 S1P에서도 MAPKp42 /44 및 AKT 신호전달 경로를 활성화시켜 UC-MSCs의 프라이밍을 촉진할 수 있다는 것을 나타낸다. Next, the present inventors investigated the effect of VPA on the priming of UC-MSCs at low concentrations of S1P treatment. To this end, we applied 50-nM S1P four times less than the optimal dose (200-nM) used for priming of fat- and UCB-derived MSCs. Unlike VPA alone and S1P alone, UC-MSCs priming increased VT + S1P treatment two to three times higher chemotactic activity for SDF-1 than unprimed cells (FIGS. 3A and 3A). 3B). The response to SDF-1 enhanced by VPA + S1P priming appeared completely different from that of 5-Aza + S1P priming. Next, we identified the state of the signaling pathway associated with the movement of HSPCs. Consistent with the results of chemotaxis assay, UC-MSCs exposed to VPA + S1P increased the phosphorylation of MAPK p42 / 44 and AKT proteins, indicating activation of signaling pathways (FIG. 3C). The results indicate that 5-Aza unlike VPA is even lower concentrations of S1P MAPK p42 / 44 and activate the AKT signaling pathway can promote the priming of the UC-MSCs.
<실시예 3> VPA+S1P로 프라이밍된 UC-MSCs의 치료 능력 향상Example 3 Improvement of Treatment Capability of UC-MSCs Primed with VPA + S1P
본 발명자들은 MSCs의 치료 효과와 관련된 다른 세포 활성에 대한 VPA+S1P 프라이밍의 효과를 시험하였다. 5-Aza+S1P와 다르게(도 1), VPA+S1P로 프라이밍된 UC-MSCs는 세포 증식(도 4A) 및 클론원성 CFU-F 활성(도 4B) 모두 향상되었다. 하지만, VPA 단독 처리와 S1P 단독 처리한 UC-MSCs에서는 세포 증식능과 CFU-F 활성 증가가 관찰되지 않았다(도 4A 및 도 4B). MSCs는 항염증 및 면역조절 능력에 영향을 미치므로, 본 발명자들은 VPA+S1P 프라이밍이 UC-MSCs의 항염증 효과에 영향을 미치는지 시험하였다. 이에, 본 발명자들은 프라이밍 되지 않은 UC-MSCs 또는 VPA 단독이거나 VPA+S1P로 프라이밍된 UC-MSCs로부터 수집된 조건 배지를 준비하였고, 조건 배지가 지질다당류(lipopolysaccharide; LPS)로 자극된 폐포 대식 세포(alveolar macrophages cell)인 MH-S 세포로부터 종양 괴사 인자-α(tumor necrosis factor-α; TNF-α)의 분비를 억제할 수 있는지 시험하였다. UC-MSCs로부터 수집된 조건 배지는 LPS 자극된 MH-S 세포로부터 TNF-α의 분비를 감소시키는데 효과적이었다(도 4C). 특히, VPA+S1P 프라이밍된 UC-MSCs로부터 수집된 조건 배지는 프라이밍되지 않은 UC-MSCs 또는 VPA 단독으로 프라이밍된 UC-MSCs 보다 TNF-α 분비를 더욱 억제하였다. 본 발명자들은 인간 폐 섬유아세포인 IMR90으로 수집한 CM을 대조군으로 사용하였는데, 이는 in vitro 항염증 분석에서 TNF-α 분비를 더 증가시키는 것으로 나타났다.We tested the effect of VPA + S1P priming on other cellular activities related to the therapeutic effect of MSCs. Unlike 5-Aza + S1P (FIG. 1), UC-MSCs primed with VPA + S1P improved both cell proliferation (FIG. 4A) and clonal CFU-F activity (FIG. 4B). However, no increase in cell proliferation and CFU-F activity was observed in UC-MSCs treated with VPA alone and S1P alone (FIGS. 4A and 4B). Since MSCs affect anti-inflammatory and immunomodulatory capacity, we tested whether VPA + S1P priming affected the anti-inflammatory effects of UC-MSCs. Accordingly, the present inventors prepared a conditioned medium collected from unprimed UC-MSCs or VPA alone or from UC-MSCs primed with VPA + S1P, and the conditioned medium stimulated with lipopolysaccharide (LPS). Tumor necrosis factor-α (TNF-α) secretion was inhibited from MH-S cells, alveolar macrophages cells. Conditional medium collected from UC-MSCs was effective in reducing the secretion of TNF-α from LPS stimulated MH-S cells (FIG. 4C). In particular, the conditioned media collected from VPA + S1P primed UC-MSCs inhibited TNF-α secretion more than unprimed UC-MSCs or UC-MSCs primed with VPA alone. We used CM collected with IMR90, a human lung fibroblast, as a control, which was shown to further increase TNF-α secretion in vitro anti-inflammatory analysis.
VPA+S1P 프라이밍의 효과에 대한 분자학적 기전을 확인하기 위해서, 본 발명자들은 MSCs에 의해 분비되는 성장인자들, 전-염증 사이토카인들 및 항-염증 인자들의 발현을 시험하였다. 앞서 언급한 세포학적 특성 실험 결과와 일치하게, VPA+S1P로 프라이밍된 UC-MSCs는 성장인자들 및 이의 수용체들[e.g., PDGFB, PDGFRB, cMET] (도 5B), 전-혈관신생- [e.g., VEGFB, VEGFC, ANGPT1, ANGPT2] (도 5C), 항-염증- [e.g., LIF, TSG6, IDO1, IDO2] (도 5D) 및 줄기 세포 이동- [e.g., MMP12] (도 5A) 관련 인자들의 발현을 상당히 상향 조절하였다. 하지만, VPA 단독 처리와 S1P 단독 처리한 UC-MSCs에서는 앞서 언급한 유전자 발현에 유의적인 변화가 관찰되지 않았다(도 4A 및 도 4B). 또한, 5-Aza+S1P 프라이밍은 VPA+S1P 프라이밍에 의해 영향을 미친 앞서 언급한 유전자들을 하향조절하였다. 종합하면, 상기 결과들은 최소 농도의 VPA 및 S1P 처리를 통한 MSCs의 프라이밍이 MSCs의 이동, 증식, 자가재생 및 항-염증 능력을 효과적으로 촉진시킨다는 것을 나타내는데, 이는 MSCs의 치료 잠재력에 중요한 역할을 한다. To confirm the molecular mechanism for the effect of VPA + S1P priming, we tested the expression of growth factors, pro-inflammatory cytokines and anti-inflammatory factors secreted by MSCs. Consistent with the results of the above-mentioned cytological experiments, UC-MSCs primed with VPA + S1P showed growth factors and their receptors [eg, PDGFB, PDGFRB, cMET] (FIG. 5B), pre-angiogenesis- [eg , VEGFB, VEGFC, ANGPT1, ANGPT2] (FIG. 5C), anti-inflammatory- [eg, LIF, TSG6, IDO1, IDO2] (FIG. 5D) and stem cell migration-[eg, MMP12] (FIG. 5A) Expression was significantly upregulated. However, no significant changes were observed in the aforementioned gene expression in UC-MSCs treated with VPA alone and S1P alone (FIGS. 4A and 4B). In addition, 5-Aza + S1P priming downregulated the aforementioned genes affected by VPA + S1P priming. Taken together, the results indicate that priming of MSCs with minimal concentrations of VPA and S1P treatment effectively promotes the migration, proliferation, self-renewal and anti-inflammatory capacity of MSCs, which plays an important role in the therapeutic potential of MSCs.
<< 실시예Example 3> 다양한  3> various VPAVPA  And S1PS1P 농도에 따른  According to concentration SDFSDF -1a에 대한 주화성(Chemotaxis) 분석Chemotaxis Analysis of -1a
본 발명자들은 다양한 VPA 및 S1P 농도에 따른 SDF-1a에 대한 주화성(Chemotaxis) 분석을 수행하여, VPA 및 S1P의 최하 농도를 결정하고자 하였다.The present inventors attempted to determine the lowest concentration of VPA and S1P by performing a chemotaxis analysis on SDF-1a according to various VPA and S1P concentrations.
도 6에 나타낸 바와 같이, VPA는 0.5 mM로 고정하고, S1P 농도를 5-50 nM로 실험한 결과, S1P의 최하 농도는 10 nM로 결정되었다(도 6a). 또한, S1P는 50 nM 로 고정하고, VPA 농도를 0.05-0.5 mM로 실험한 결과, VPA의 최하 농도는 0.1 nM로 결정되었다(도 6b).As shown in FIG. 6, VPA was fixed at 0.5 mM, and the S1P concentration was tested at 5-50 nM. As a result, the lowest concentration of S1P was determined to be 10 nM (FIG. 6A). In addition, S1P was fixed at 50 nM and the VPA concentration was 0.05-0.5 mM. As a result, the lowest concentration of VPA was determined to be 0.1 nM (FIG. 6B).
<< 실시예Example 4> IC/BPS 치료에 있어,  4> In IC / BPS therapy, VPAVPA ++ S1PS1P 프라이밍된Primed UCUC -- MSC의MSc 생체 내(in vivo) 치료 가능성 확인 Confirmation of in vivo treatment potential
다음으로, 본 발명자들은 HCl 주입으로 확립된 IC/BPS 동물 모델을 사용하여 UC-MSCs의 생체 내 효과를 확인하였다. 비마취하 방광내압측정술(awake cystometry)를 사용한 방광 기능 분석 결과, HCl 주입 유도된 IC/BPS 쥐(HCl-IC group)는 대조군(sham-operated; sham) 쥐에 비하여, 불규칙한 배뇨를 보였고, 배뇨 간격(micturition interval; MI)이 줄어들었을 뿐만 아니라, 배뇨량(micturition volume; MV), 최대 압력, 배뇨 압력(micturition pressure; MP) 및 방광 용량(bladder capacity; BC)도 낮아진 것으로 나타났다(도 7a 및 도 7b). 2.5×105 세포수의 대조군(naive) UC-MSCs(UC-MSC 250K) 또는 VPA+S1P 프라이밍된 세포(UC-MSC 250K+VPA+S1P)의 단일 이식은 손상된 배뇨 파라미터들을 개선시켰다. 중요하게도, VPA+S1P 프라이밍된 UC-MSCs가 대조군 세포보다 우수한 효과를 나타냈다(도 7a 및 도 7b). 특히, HCl-IC 그룹의 동물들에서 비-배뇨 기간(non-voiding periods; NVC) 동안 수축 빈도가 증가하는 것으로 나타났는데, 이러한 증상은 VPA+S1P 프라이밍된 UC-MSCs에서 상당히 개선되었으나, 대조군 UC-MSCs에서는 그다지 개선되지 않았다(도 7a 및 도 7b). 비마취하 방광내압측정술(awake cystometry) 결과도 상기 기능 개선 효과와 일치하였는데, 대조군 또는 VPA+S1P 프라이밍된 2.5×105 UC-MSCs의 투여로 인해 쥐 모델에서 심각한 요로 상피 박리(urothelium denudation), 비만 세포 침윤, 섬유화, 세포 사멸과 같은 비정상적인 조직학적 현상들이 회복되는 것으로 나타났다(도 8a 및 도 8b). 이러한 비정상적인 조직학적 현상들은 인간 IC/BPS 방광에서도 나타나는 특징이다. 특히, 조직학적 변형의 회복은 대조군 세포에 비해 VPA+S1P 프라이밍된 UC-MSC가 이식된 방광 조직에서 더 뛰어난 개선 효과를 나타냈다. 종합하면, 상기 결과는 IC 방광 치료에 있어, VPA+S1P 프라이밍이 UC-MSCs의 치료 가능성을 상당히 개선시킬 수 있다는 것을 나타냈다. Next, we confirmed the in vivo effects of UC-MSCs using an IC / BPS animal model established by HCl injection. As a result of bladder function analysis using awake cystometry, the HCl-induced IC / BPS mice (HCl-IC group) showed irregular urination and urination compared to sham-operated (sham) mice. Not only did the MI (turturation interval; MI) decrease, but also the micturition volume (MV), maximum pressure, micturition pressure (MP), and bladder capacity (BC) were also shown to be lower (FIGS. 7A and FIG. 7b). Single transplantation of control UC-MSCs (UC-MSC 250K) or VPA + S1P primed cells (UC-MSC 250K + VPA + S1P) with 2.5 × 10 5 cell numbers improved impaired urination parameters. Importantly, VPA + S1P primed UC-MSCs showed a better effect than control cells (FIGS. 7A and 7B). In particular, in animals in the HCl-IC group, contraction frequency was increased during non-voiding periods (NVC), which was significantly improved in VPA + S1P primed UC-MSCs, but in control UC. Not so much improved in MSCs (FIGS. 7A and 7B). Were Bima take cystometry alcohol (awake cystometry) results are consistent with the functional effect of improving the control or VPA + S1P primed 2.5 × 10 5 UC-MSCs severe urinary tract epithelial detachment in a rat model because of the administration of (urothelium denudation), Abnormal histological phenomena such as mast cell infiltration, fibrosis, and cell death have been shown to recover (FIGS. 8A and 8B). These abnormal histological phenomena are also characteristic of human IC / BPS bladder. In particular, recovery of histological alterations showed a better improvement in bladder tissues implanted with VPA + S1P primed UC-MSCs compared to control cells. Taken together, the results showed that for IC bladder treatment, VPA + S1P priming can significantly improve the therapeutic potential of UC-MSCs.
<실시예 5> PAH 동물 모델에서 UC-MSC의 VPA+S1P-매개 치료 가능성 향상Example 5 Enhancement of VPA + S1P-Mediated Treatment Potential of UC-MSC in PAH Animal Model
다음으로, 본 발명자들은 MCT로 유도된 PAH 동물모델을 사용하여 생체 내 조건하에서 VPA+S1P 프라이밍의 효과를 확인하였다. 이전 보고와 유사하게, RVSP는 MCT 투여 4주 후에 상당히 증가하였다. 대조군(naive) UC-MSC (PAH+MSC 그룹)과 달리, VPA+S1P로 프라이밍된 UC-MSC (PAH+VPA+S1P-MSC 그룹)의 투여는 MCT-유도 RVSP의 상승을 상당히 감소시켰다(도 9a). 또한, MCT 투여 후, RV/(LV+S)도 증가되었는데, VPA+S1P-MSC는 RV 비대증(hypertrophy)를 상당히 감소시켰으며, 프라이밍 하지 않은 UC-MSC는 그다지 효과가 없었다(도 9a). 또한, VPA+S1P-MSC 투여는 MCT에 의해 유도된 폐 조직 염증, 중막 두께 지수 및 α-SMA+ 평활근 세포의 증가를 감소시켰다(도 9b). 종합하면, 상기 결과는 VPA+S1P에 의한 UC-MSC의 프라이밍이 신생혈관 형성을 촉진시키고, 염증반응을 억제하는 미세환경을 유도할 수 있다는 것을 나타냈다.Next, the inventors confirmed the effect of VPA + S1P priming under in vivo conditions using an MCT-induced PAH animal model. Similar to previous reports, RVSP increased significantly after 4 weeks of MCT administration. Unlike the control UC-MSC (PAH + MSC group), administration of UC-MSC primed with VPA + S1P (PAH + VPA + S1P-MSC group) significantly reduced the elevation of MCT-induced RVSP (FIG. 9a). In addition, after MCT administration, RV / (LV + S) was also increased, with VPA + S1P-MSC significantly reducing RV hypertrophy, and unpriming UC-MSC was ineffective (FIG. 9A). In addition, VPA + S1P-MSC administration reduced lung tissue inflammation, medial thickness index, and α-SMA + smooth muscle cell induced by MCTs (FIG. 9B). Taken together, the results indicated that priming of UC-MSCs with VPA + S1P can induce angiogenesis and induce a microenvironment that inhibits inflammatory responses.
이상으로 본 발명의 특정한 부분을 상세히 기술하였는 바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시예일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.Having described the specific parts of the present invention in detail, it will be apparent to those skilled in the art that such specific descriptions are merely preferred embodiments, and thus the scope of the present invention is not limited thereto. will be. Thus, the substantial scope of the present invention will be defined by the appended claims and their equivalents.

Claims (20)

  1. 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 유효성분으로 포함하는 줄기세포 활성 촉진용 조성물.Stem cell activity promoting composition comprising a histone deacetylation inhibitor and a priming factor (priming factor) as an active ingredient.
  2. 제1항에 있어서, 상기 히스톤 탈아세틸화 저해제는 발프로익산(valproic acid; VPA), 소듐 뷰티레이트(sodium butyrate; NaB), 니코틴아미드(nicotinamide; NAD) 및 서티놀(sirtinol)로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 줄기세포 활성 촉진용 조성물.The method of claim 1, wherein the histone deacetylation inhibitor in the group consisting of valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) and sirtinol Stem cell activity promoting composition, characterized in that any one or more selected.
  3. 제1항에 있어서, 상기 프라이밍 인자는 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P), 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P), 카세리시딘(cathelicidin; LL-37) 및 피오글리타존(pioglitazone)으로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 줄기세포 활성 촉진용 조성물.The method according to claim 1, wherein the priming factor is sphingosine-1-phosphate (Sphingosine-1-phosphate (S1P), ceramide-1-phosphate (ceramide-1-phosphate (C1P), cathelicidin (LL-37) ) And pioglitazone (pioglitazone) composition for promoting stem cell activity, characterized in that any one or more selected from the group consisting of.
  4. 제2항에 있어서, 상기 VPA는 0.1 내지 1.0 mM 농도로 포함되는 것을 특징으로 하는 줄기세포 활성 촉진용 조성물.The composition for promoting stem cell activity according to claim 2, wherein the VPA is contained in a concentration of 0.1 to 1.0 mM.
  5. 제3항에 있어서, 상기 S1P는 10 내지 50 nM 농도로 포함되는 것을 특징으로 하는 줄기세포 활성 촉진용 조성물.The composition for promoting stem cell activity according to claim 3, wherein the S1P is included in a concentration of 10 to 50 nM.
  6. 제1항 내지 제5항 중 어느 한 항에 있어서, 상기 줄기세포는 신경 줄기세포, 간 줄기세포, 조혈 줄기세포, 제대혈 줄기세포, 표피 줄기세포, 위장관 줄기세포, 내피 줄기세포, 근육 줄기세포, 중간엽 줄기세포 및 췌장 줄기세포로 이루어진 군으로부터 선택되는 것을 특징으로 하는 줄기세포 활성 촉진용 조성물.The method according to any one of claims 1 to 5, wherein the stem cells are neural stem cells, liver stem cells, hematopoietic stem cells, cord blood stem cells, epidermal stem cells, gastrointestinal stem cells, endothelial stem cells, muscle stem cells, Stem cell activity promoting composition, characterized in that selected from the group consisting of mesenchymal stem cells and pancreatic stem cells.
  7. 제1항 내지 제5항 중 어느 한 항에 있어서, 상기 줄기세포 활성은 세포 이동성, 콜로니 형성능 및 항-염증 활성인 것을 특징으로 하는 줄기세포 활성 촉진용 조성물.The composition for promoting stem cell activity according to any one of claims 1 to 5, wherein the stem cell activity is cell mobility, colony forming ability and anti-inflammatory activity.
  8. 분리된 줄기세포에 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)를 처리하는 단계를 포함하는 줄기세포 활성 촉진 방법. Stem cell activity promotion method comprising the step of treating the isolated stem cells histone deacetylation inhibitor and priming factor (priming factor).
  9. 제8항에 있어서, 상기 히스톤 탈아세틸화 저해제는 발프로익산(valproic acid; VPA), 소듐 뷰티레이트(sodium butyrate; NaB), 니코틴아미드(nicotinamide; NAD) 및 서티놀(sirtinol)로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 줄기세포 활성 촉진 방법. The method of claim 8, wherein the histone deacetylation inhibitor in the group consisting of valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) and sirtinol Stem cell activity promoting method, characterized in that any one or more selected.
  10. 제8항에 있어서, 상기 프라이밍 인자는 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P), 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P), 카세리시딘(cathelicidin; LL-37) 및 피오글리타존(pioglitazone)으로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 줄기세포 활성 촉진 방법.The method according to claim 8, wherein the priming factor is sphingosine-1-phosphate (Sphingosine-1-phosphate (S1P), ceramide-1-phosphate (ceramide-1-phosphate (C1P), cathelicidin (LL-37) ) And pioglitazone (pioglitazone) is any one or more selected from the group consisting of stem cell activity promoting method.
  11. 제9항에 있어서, 상기 VPA는 0.1 내지 1.0 mM 농도로 처리되는 것을 특징으로 하는 줄기세포 활성 촉진 방법.The method of claim 9, wherein the VPA is treated at a concentration of 0.1 to 1.0 mM.
  12. 제10항에 있어서, 상기 S1P는 10 내지 50 nM 농도로 처리되는 것을 특징으로 하는 줄기세포 활성 촉진 방법.The method of claim 10, wherein the S1P is treated at a concentration of 10 to 50 nM.
  13. 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 고혈압 예방 또는 치료용 약학조성물.A pharmaceutical composition for preventing or treating hypertension, comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
  14. 제13항에 있어서, 상기 히스톤 탈아세틸화 저해제는 발프로익산(valproic acid; VPA), 소듐 뷰티레이트(sodium butyrate; NaB), 니코틴아미드(nicotinamide; NAD) 및 서티놀(sirtinol)로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 고혈압 예방 또는 치료용 약학조성물.The method of claim 13, wherein the histone deacetylation inhibitor in the group consisting of valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD) and sirtinol A pharmaceutical composition for preventing or treating hypertension, characterized in that any one or more selected.
  15. 제13항에 있어서, 상기 프라이밍 인자(priming factor)는 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P), 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P), 카세리시딘(cathelicidin; LL-37) 및 피오글리타존(pioglitazone)으로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 고혈압 예방 또는 치료용 약학조성물.The method of claim 13, wherein the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and cathelicidin LL-37) and pioglitazone (pioglitazone) is any one or more selected from the group consisting of a pharmaceutical composition for preventing or treating hypertension.
  16. 제13항에 있어서, 상기 고혈압은 특발성 폐동맥성 고혈압; 가족성 폐동맥성 고혈압; 콜라겐 혈관 질환, 선천성 체폐 단락(短絡), 문맥 고혈압, HIV 감염, 약물 또는 독소와 관련된 폐동맥성 고혈압; 티로이드 장애, 글리코겐 저장 질환, 고셔(Gaucher) 질환, 유전성 출혈성 모세혈관 확장, 혈색소병증, 골수증식장애 또는 비장절제술과 관련된 폐동맥성 고혈압; 폐 모세혈관 혈관종증과 관련된 폐동맥성 고혈압; 신생아의 지속성 폐고혈압; 만성 폐쇄성 폐 질환, 간질성 폐 질환, 저산소증 유도 폐포 저환기 장애, 저산소증 유도 수면장애 호흡 또는 높은 고도에의 만성 노출과 관련된 폐고혈압; 발달 이상과 관련된 폐고혈압; 및 원위 폐동맥의 혈전색전성 폐쇄에 의한 폐고혈압으로 이루어지는 군으로부터 선택되는 것을 특징으로 하는 고혈압 예방 또는 치료용 약학조성물.The method of claim 13, wherein the hypertension is idiopathic pulmonary arterial hypertension; Familial pulmonary arterial hypertension; Pulmonary arterial hypertension associated with collagen vascular disease, congenital pulmonary shortness, portal hypertension, HIV infection, drugs or toxins; Pulmonary hypertension associated with thyroid disorders, glycogen storage disease, Gaucher disease, hereditary hemorrhagic capillary dilatation, hemochromatosis, myeloproliferative disorder or splenectomy; Pulmonary arterial hypertension associated with pulmonary capillary angiomatosis; Persistent pulmonary hypertension in newborns; Pulmonary hypertension associated with chronic obstructive pulmonary disease, interstitial lung disease, hypoxia induced alveolar hypoventilatory disorder, hypoxia induced sleep disorder breathing or chronic exposure to high altitudes; Pulmonary hypertension associated with developmental abnormalities; And pulmonary hypertension due to thromboembolic closure of the distal pulmonary artery.
  17. 히스톤 탈아세틸화 저해제 및 프라이밍 인자(priming factor)로 처리된 줄기세포 또는 이의 배양액을 유효성분으로 포함하는 염증질환 또는 면역질환 예방 또는 치료용 약학조성물.A pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases, comprising a stem cell treated with a histone deacetylation inhibitor and a priming factor or a culture medium thereof as an active ingredient.
  18. 제17항에 있어서, 상기 히스톤 탈아세틸화 저해제는 발프로익산(valproic acid; VPA), 소듐 뷰티레이트(sodium butyrate; NaB), 니코틴아미드(nicotinamide; NAD) 및 서티놀(sirtinol)로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 염증질환 또는 면역질환 예방 또는 치료용 약학조성물.18. The method of claim 17, wherein the histone deacetylation inhibitor is selected from the group consisting of valproic acid (VPA), sodium butyrate (NaB), nicotinamide (NAD), and sirtinol. Pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases, characterized in that any one or more selected.
  19. 제17항에 있어서, 상기 프라이밍 인자(priming factor)는 스핑고신-1-포스페이트(sphingosine-1-phosphate; S1P), 세라마이드-1-포스페이트(ceramide-1-phosphate; C1P), 카세리시딘(cathelicidin; LL-37) 및 피오글리타존(pioglitazone)으로 이루어진 군에서 선택된 어느 하나 이상인 것을 특징으로 하는 염증질환 또는 면역질환 예방 또는 치료용 약학조성물.The method of claim 17, wherein the priming factor is sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and cathelicidin LL-37) and pioglitazone (pioglitazone) pharmaceutical composition for the prevention or treatment of inflammatory or immune diseases, characterized in that any one or more selected from the group consisting of.
  20. 제17항에 있어서, 상기 염증질환 또는 면역질환은 골관절염, 류마티스 관절염(Rheumatoid Arthritis), 방광염, 간질성방광염, 천식(Asthma), 피부염(Dermititis), 아토피, 건선(Psoriasis), 낭섬유증(Cystic Fibrosis), 고형장기 이식 후기 및 만성 거부증(Post transplantation late and chronic solid organ rejection), 이식편대숙주질환(graft-versus-host disease), 이식거부질환, 다발성 경화증(Multiple Sclerosis), 전신성 홍반성 루푸스(systemic lupus erythematosus), 쇼그렌 증후군(Sjogren syndrome), 하시모토 갑상선(Hashimoto thyroiditis), 다발성근염(polymyositis), 경피증(scleroderma), 아디슨병(Addison disease), 백반증(vitiligo), 악성빈혈(pernicious anemia), 사구체신염(glomerulonephritis) 및 폐섬유증(pulmonary fibrosis), 염증성장질환(Inflammatory Bowel Diesese), 크론 병(Crohns disease), 자가면역성 당뇨(Autoimmune Diabetes), 당뇨 망막증(Diabetic retinopathy), 비염(Rhinitis), 허혈-재관류 손상(Ischemia-reperfusion injury), 혈관성형술후 재협착(Post-angioplasty restenosis), 만성 폐색성 심장 질환(Chronic obstructive pulmonary disease; COPD), 그레이브병(Graves disease), 위장관 알러지(Gastrointestinal allergy), 결막염(Conjunctivitis), 죽상경화증(Atherosclerosis), 관상동맥질환(Coronary artery disease), 협심증(Angina), 암 전이, 소동맥 질환 및 미토콘드리아 질환(mitochondrial disease)으로 이루어진 군으로부터 선택되는 것을 특징으로 하는 염증질환 또는 면역질환 예방 또는 치료용 약학조성물.The method of claim 17, wherein the inflammatory disease or immune disease is osteoarthritis, Rheumatoid Arthritis, cystitis, interstitial cystitis, asthma, dermititis, atopic dermatitis, Psoriasis, cystic fibrosis ), Post transplantation late and chronic solid organ rejection, graft-versus-host disease, graft rejection disease, multiple sclerosis, systemic lupus erythematosus lupus erythematosus, Sjogren syndrome, Hashimoto thyroiditis, polymyositis, scleroderma, Addison disease, vitiligo, pernicious anemia, glomerulonephritis (glomerulonephritis) and pulmonary fibrosis, Inflammatory Bowel Diesese, Crohn's disease, Autoimmune Diabetes, Diabetic retinopathy ), Rhinitis, ischemia-reperfusion injury, post-angioplasty restenosis, chronic obstructive pulmonary disease (COPD), Graves disease Selected from the group consisting of gastrointestinal allergy, conjunctivitis, atherosclerosis, coronary artery disease, angina, cancer metastasis, arterial disease and mitochondrial disease Pharmaceutical composition for preventing or treating inflammatory diseases or immune diseases, characterized in that.
PCT/KR2017/012675 2016-11-10 2017-11-09 Composition for promoting stem cell activity, comprising histone deacetylase inhibitor and priming factor as active ingredients WO2018088821A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201780069520.XA CN110462023B (en) 2016-11-10 2017-11-09 Stem cell activity promoting composition comprising histone deacetylase inhibitor and activator as active ingredients
JP2019520846A JP6934516B2 (en) 2016-11-10 2017-11-09 A composition for promoting stem cell activity containing a histone deacetylation inhibitor and a priming factor as active ingredients.

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2016-0149375 2016-11-10
KR20160149375 2016-11-10
KR10-2017-0148019 2017-11-08
KR1020170148019A KR102097191B1 (en) 2016-11-10 2017-11-08 Composition for enforcing the priming effect of stem cell comprising histone deacetylase inhibitor and priming factors

Publications (1)

Publication Number Publication Date
WO2018088821A1 true WO2018088821A1 (en) 2018-05-17

Family

ID=62109863

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2017/012675 WO2018088821A1 (en) 2016-11-10 2017-11-09 Composition for promoting stem cell activity, comprising histone deacetylase inhibitor and priming factor as active ingredients

Country Status (1)

Country Link
WO (1) WO2018088821A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11266696B2 (en) * 2019-01-18 2022-03-08 Americord Registry Llc Method of treating stem cells with NAD

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000023567A2 (en) * 1998-10-16 2000-04-27 Novartis Ag Promotion of self-renewal and improved gene transduction of hematopoietic stem cells by histone deacetylase inhibitors

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000023567A2 (en) * 1998-10-16 2000-04-27 Novartis Ag Promotion of self-renewal and improved gene transduction of hematopoietic stem cells by histone deacetylase inhibitors

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
KANG, H. ET AL.: "The Therapeutic Effects of Human Mesenchymal Stem Cells Primed with Sphingosine-1 Phosphate on Pulmonary Artery Hypertension", STEM CELLS DEV., vol. 24, no. 14, 2015, pages 1658 - 1671, XP055493135 *
LIM, J. ET AL.: "Priming with Ceramide-l Phosphate Promotes the Therapeutic Effect of Mesenchymal Stern/stromal Cells on Pulmonary Artery Hypertension", BIOCHEM. BIOPHYS. RES. COMMUN., vol. 473, 2016, pages 35 - 41, XP055493127, DOI: doi:10.1016/j.bbrc.2016.03.046 *
LIM, J. ET AL.: "Valproic Acid Enforces the Priming Effect of Sphingosine-1 Phosphate on Human Mesenchymal Stem Cells", INT. J. MOL. MED., vol. 40, 3 July 2017 (2017-07-03), pages 739 - 747, XP055493143 *
MARQUEZ-CURTIS, L. A. ET AL.: "Migration, Proliferation, and Differentiation of Cord Blood Mesenchymal Stromal Cells Treated with Histone Deacetylase Inhibitor Valproic Acid", STEM CELLS INT., vol. 2014, 16 March 2014 (2014-03-16), pages 1 - 14, XP055493128 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11266696B2 (en) * 2019-01-18 2022-03-08 Americord Registry Llc Method of treating stem cells with NAD

Similar Documents

Publication Publication Date Title
Weil et al. Mesenchymal stem cells enhance the viability and proliferation of human fetal intestinal epithelial cells following hypoxic injury via paracrine mechanisms
Kwon et al. Multiple paracrine factors secreted by mesenchymal stem cells contribute to angiogenesis
Wang et al. Prolyl hydroxylase domain protein 2 silencing enhances the survival and paracrine function of transplanted adipose-derived stem cells in infarcted myocardium
US10632153B2 (en) Compositions and methods for cardiac tissue repair
Ohta et al. Intravenous infusion of adipose-derived stem/stromal cells improves functional recovery of rats with spinal cord injury
KR102097191B1 (en) Composition for enforcing the priming effect of stem cell comprising histone deacetylase inhibitor and priming factors
Zhang et al. Atorvastatin treatment improves the effects of mesenchymal stem cell transplantation on acute myocardial infarction: the role of the RhoA/ROCK/ERK pathway
WO2014181954A1 (en) Culture medium composition for improving regenerative capacity of stem cells, and stem cell culturing method using same
D'Avola et al. Phase 1–2 pilot clinical trial in patients with decompensated liver cirrhosis treated with bone marrow–derived endothelial progenitor cells
KR20090074044A (en) Expansion method for adult stem cells from blood, particularly peripheral blood, and relative application in medical field
Carreras et al. Obstructive apneas induce early activation of mesenchymal stem cells and enhancement of endothelial wound healing
Galie et al. Injection of mesenchymal stromal cells into a mechanically stimulated in vitro model of cardiac fibrosis has paracrine effects on resident fibroblasts
Ikhapoh et al. Sry-type HMG box 18 contributes to the differentiation of bone marrow-derived mesenchymal stem cells to endothelial cells
WO2017146468A1 (en) Composition and method for improving efficacy of stem cells
KR101777414B1 (en) Activating agent of stem cells and/or progenitor cells
Yin et al. Protective effects of CXCR3/HO‑1 gene‑modified BMMSCs on damaged intestinal epithelial cells: Role of the p38‑MAPK signaling pathway
US20210069177A1 (en) Method for destroying cellular mechanical homeostasis and promoting regeneration and repair of tissues and organs, and use thereof
WO2018088821A1 (en) Composition for promoting stem cell activity, comprising histone deacetylase inhibitor and priming factor as active ingredients
Wang et al. Reconstitute the damaged heart via the dual reparative roles of pericardial adipose-derived flk-1+ stem cells
Ye et al. Exendin-4 promotes proliferation of adipose-derived stem cells through PI3K/Akt-Wnt signaling pathways
WO2020189948A1 (en) Mesenchymal stem cell-based patch application technology for improving therapeutic efficiency and performance of induced pluripotent stem cell-derived cell therapeutic agent
Kaba et al. Peroxisome proliferator-activated receptor-γ agonist attenuates vocal fold fibrosis in rats via regulation of macrophage activation
WO2023182609A1 (en) Composition for promoting angiogenesis, comprising adipose tissue-derived mesenchymal stem cells and human stromal vascular fraction
WO2014051338A2 (en) Pharmaceutical composition for preventative or treatment use comprising peripheral blood mononuclear cell as active ingredient
WO2014042292A1 (en) Composition comprising protein kinase c activator for promoting stem cell adhesion, and method for promoting stem cell adhesion

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: 17870530

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019520846

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17870530

Country of ref document: EP

Kind code of ref document: A1