WO2017074043A1 - Composition containing cells treated with 15-pgdh inhibitor or culture thereof and use thereof - Google Patents
Composition containing cells treated with 15-pgdh inhibitor or culture thereof and use thereof Download PDFInfo
- Publication number
- WO2017074043A1 WO2017074043A1 PCT/KR2016/012125 KR2016012125W WO2017074043A1 WO 2017074043 A1 WO2017074043 A1 WO 2017074043A1 KR 2016012125 W KR2016012125 W KR 2016012125W WO 2017074043 A1 WO2017074043 A1 WO 2017074043A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- inhibitor
- cells
- pgdh
- pge2
- cell
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K8/00—Cosmetics or similar toiletry preparations
- A61K8/18—Cosmetics or similar toiletry preparations characterised by the composition
- A61K8/30—Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
- A61K8/33—Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing oxygen
- A61K8/34—Alcohols
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/045—Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
- A61K31/05—Phenols
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/34—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/335—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
- A61K31/35—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
- A61K31/352—Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/41—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
- A61K31/415—1,2-Diazoles
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K8/00—Cosmetics or similar toiletry preparations
- A61K8/18—Cosmetics or similar toiletry preparations characterised by the composition
- A61K8/30—Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
- A61K8/49—Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds containing heterocyclic compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61Q—SPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
- A61Q19/00—Preparations for care of the skin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61Q—SPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
- A61Q5/00—Preparations for care of the hair
Definitions
- the present invention relates to a composition comprising a cell cultured by adding a 15-PGDH inhibitor to a cell expressing PGE2 or a culture thereof and a use thereof. More specifically, the present invention relates to a pharmaceutical composition for the prevention or treatment of immune diseases, inflammatory diseases, or wounds, comprising cells cultured with the addition of a 15-PGDH inhibitor to PGE2 expressing cells or a culture thereof.
- a pharmaceutical composition, a method of inhibiting an immune response, an inflammatory response or a wound in a subject comprising administering the cell or a culture thereof to the subject, an immunosuppressive agent, an anti-inflammatory agent using the cell or its culture Or a method for preparing a wound therapeutic agent, a method of preparing PGE2 comprising culturing by adding a 15-PGDH inhibitor to a cell expressing PGE2, a transplant comprising a cell expressing PGE2 and a 15-PGDH inhibitor, the transplant Method for preparing a sieve, a complex comprising a PGE2 expressing cell and a 15-PGDH inhibitor and a culture obtained by adding a 15-PGDH inhibitor to a PGE2 expressing cell It relates.
- Prostaglandin E2 (hereinafter PGE2) is prostaglandin E2 (5Z, 11 (alpha) 13E, 15S) -11, 15-dihydroxy-9-oxo-prostar-5, 13-diene-1-oic acid [ (5Z, 11 (alpha), 13E, 15S) -11, 15-Dihydroxy-9-oxo-prosta-5, 13-dien-1-oic acid], and the physiological and pathological environment Is the most widely produced prostaglandin (Ushikubi F et al., J. Pharmacol. Sci. 83: 279, 2000).
- PGE2 has traditionally been used in the cervix to prepare for birth and is actually pharmacologically used to induce birth and is sold in the form of vaginal suppositories under the following trade names: Cervidil (Forest Corporation), Prostin E2 (Pfizer Corporation), Propess (Ferring Pharmaceutical Company), Glandin (Nabiqasim Pharmaceuticals, Pakistan).
- Cervidil Formest Corporation
- Prostin E2 Pfizer Corporation
- Propess Frerring Pharmaceutical Company
- Glandin Naabiqasim Pharmaceuticals, Pakistan
- immunosuppressive and modulators has been in the spotlight, which plays a role in inhibiting the secretion of cytokines such as interleukin-1 beta and TNF alpha produced by macrophages, and inhibiting the differentiation of helper T1 cells.
- TGF- ⁇ 1 Transforming growth factor beta 1
- PGE2 has been widely available as an immunomodulator, and thus an economical and convenient production method has been required.
- immunosuppression includes specific inhibitors and nonspecific inhibitors that inhibit only the response, and theoretically, the action of specific inhibitors should be excellent, but nonspecific inhibitors are mainly used.
- the most commonly used immunosuppressive agents are cyclosporine (Neoral, Cipol A), azacioprine (imuran) and prednisolone (a type of steroid). The three were found to have fewer side effects and the highest immunosuppressive effects when used together when used separately.
- various immunosuppressive agents such as FK 506, RATG, OKT3, and Cellcept have been developed and used.
- immunosuppressive agents inhibit immunosuppression by inhibiting several processes such as phagocytosis of antigen by macrophages, antigen recognition by lymphocytes, cell division, T cell and B cell division, and antibody production. Cause. Most of them have antitumor activity because they inhibit cell division through the mediation of DNA disorders and the inhibition of DNA synthesis.
- the typical side effects are hypertension and nephrotoxicity (decrease in kidney function), and the incidence of these side effects has been high, so that there has been a problem of observing the course when used sufficiently.
- Other side effects rarely include tremor, seizures, hepatitis, biliary retention, increased blood uric acid, muscle weakness, hypertrichosis, and gingival hypertrophy.
- azachioprine inhibits bone marrow function, such as decreased white blood cell count, anemia, and platelet reduction.
- pancreatitis, hepatitis, and biliary retention may rarely cause hair loss and fever.
- Prednisolone one of the steroid agents, was the first to be used among immunosuppressants and has the broadest inhibitory action. It may increase appetite, increase muscles in the shoulders and back, and temporarily bring happiness, but these steroids not only promote atherosclerosis, but also cause high blood pressure, gastric ulcers, diabetes, growth inhibition, osteoporosis, cataracts, and glaucoma. The drug to watch out for.
- Allogeneic transplantation such as organ transplantation and hematopoietic stem cell transplantation
- organ transplantation and hematopoietic stem cell transplantation is a breakthrough medical achievement made in the 21st century, and is being used as a fundamental treatment method in late stage diseases such as heart failure, chronic renal failure, and intractable blood diseases such as dilated cardiomyopathy.
- GVHD graft-versus-host-desease
- rejection of transplanted organs which are fatal complications after allogeneic transplantation.
- the immune response after transplantation is generated by cellular immunity by T cells which recognize allogeneic antigens, thereby treating the T cell immune response (Ikehara S, Exp Hematol., 31: 1142, 2003; First MR, Transplantation, 77:88, 2004), ie, the immune response by inhibiting the production of interleukin (IL) -2 in T cells using the immunosuppressive cyclosporin, FK506.
- T cell immune response Ikehara S, Exp Hematol., 31: 1142, 2003; First MR, Transplantation, 77:88, 2004
- IL interleukin
- FK506 immunosuppressive cyclosporin
- mesenchymal stem cells appear to have an effect of inhibiting APC (antigen presenting cell). Under certain conditions, the degree of immune response changes in proportion to the number of monocytes added during the culturing process, suggesting that monocytes may be involved in immunosuppressive effects.
- mesenchymal stem cells appear to exhibit immunosuppressive effects by controlling the proliferation rate of T cells. When T cells are incubated with mesenchymal stem cells, T cells stay at G0 / G1 phase of the cell cycle and no longer increase as cyclin D2 is inhibited. It has also been reported that proliferative capacity continues to decline even when mesenchymal stem cells are removed (Glennie S et al., Blood, 105: 2821, 2005).
- the present inventors have diligently tried to develop a method of more effectively controlling immunity or inflammation using cells.
- the 15-PGDH inhibitor is treated to cells, PGE2 is significantly produced to suppress the immune response more effectively.
- the present inventors confirmed that when treated with 15-PGDH inhibitor cells, PGE2 is significantly overproduced and used for wound treatment.
- the term "15-PGDH inhibitor (inhibitor)” refers to a substance (antagonist) that inhibits the activity of the 15-PGDH.
- the 15-PGDH inhibitor is a cyclooxygenase inhibitor, a flavonoid, a phytophenolic compound, and a peroxysome proliferator-activated receptor ⁇ (PPAR ⁇ ).
- PPAR ⁇ peroxysome proliferator-activated receptor ⁇
- One or more may be selected from the group consisting of organic compounds containing antagonists of d, but is not limited thereto.
- cell as used herein includes stem cells having the ability to differentiate into various tissues.
- prevention refers to any action that inhibits or delays the development of an immune disease or inflammatory disease by administration of a composition according to the present invention
- treatment refers to the administration of a pharmaceutical composition according to the present invention. Means any activity that improves or benefits the symptoms of an immune or inflammatory disease.
- graft refers to a material that can be implanted in a human or mammal as a support that isolates the damaged area from the outside or allows the transplanted cells or secreted therapeutic material to remain.
- implants include, without limitation, various materials used in the art such as synthetic polymers and natural materials having biodegradability as a support for tissue engineering.
- the present invention provides a pharmaceutical composition for the prevention or treatment of immune diseases or inflammatory diseases, including cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). To provide.
- 15-PGDH 15-hydroxyprostaglandin dehydrogenase
- the present invention provides a method for inhibiting an immune response or an inflammatory response in a subject other than a human, including administering a cell or a culture thereof to the subject by adding a 15-PGDH inhibitor to a PGE2 expressing cell. To provide.
- the present invention is to provide a method for producing an immunosuppressant or anti-inflammatory agent comprising the step of culturing by adding a 15-PGDH inhibitor to PGE2 expressing cells.
- the present invention includes the step of culturing PGE2 expressing cells in a medium to which 15-PGDH inhibitor is added, wherein the cells secrete PGE2 (Prostaglandin E2) or TGF- ⁇ 1 (Transforming growth factor beta 1). It is intended to provide a method for producing a characteristic PGE2 or TGF- ⁇ 1.
- the present invention seeks to provide an implant comprising a cell expressing PGE2 and a 15-PGDH inhibitor.
- the present invention is to provide a method for producing an implant comprising the step of culturing by adding a 15-PGDH inhibitor to the cells expressing PGE2 on the implant.
- the present invention seeks to provide a complex comprising a PGE2 expressing cell and a 15-PGDH inhibitor.
- the present invention seeks to provide a culture obtained by adding PGE2 expressing cells and a 15-PGDH inhibitor.
- the present invention also provides a pharmaceutical composition for preventing or treating wounds comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). I would like to.
- 15-PGDH 15-hydroxyprostaglandin dehydrogenase
- the present invention is to provide a cosmetic composition
- a cosmetic composition comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2).
- the pharmaceutical composition according to the present invention has anti-inflammatory effect, serum IgE reduction effect, NO and MDA reduction effect, activated T cell proliferation inhibitory effect, and dermal fibroblast and epithelial keratinocyte proliferation ability in DNCB-induced AD mouse model Can have an effect.
- FIG. 1 shows a test schedule according to an embodiment of the present invention.
- Figure 2 shows the results of macroscopic lesions of atopy-induced atopy-induced mice by day.
- Figure 3 shows the results of skin histology of DNCB-induced atopic dermatitis mice.
- Figure 5 shows the concentrations of NO and MDA in serum and skin tissue of DNCB induced atopic dermal mice.
- Figure 6 shows the activated T-cell immunosuppression of mesenchymal stem cells.
- FIG. 7 shows the results of activated T cell immunophenotype analysis.
- composition for immune disease or inflammatory disease 1.
- the present invention provides a pharmaceutical composition for the prevention or treatment of immune diseases or inflammatory diseases, including cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). To provide.
- 15-PGDH 15-hydroxyprostaglandin dehydrogenase
- the cells are human adult stem cells, human pluripotent stem cells, induced pluripotent stem cells, embryonic stem cells of animals Or adult stem cells of an animal.
- the adult stem cells may be mesenchymal stem cells, human tissue-derived mesenchymal stromal cells, human tissue-derived mesenchymal stem cells, multipotent stem cells or amniotic epithelial cells.
- the adult stem cells are umbilical cord-derived mesenchymal stem cells, umbilical cord blood-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, fat-derived mesenchymal stem cells, muscle-derived mesenchymal stem cells, nerve-derived mesenchymal stem cells, skin It may be derived mesenchymal stem cells, amnion derived mesenchymal stem cells or placental derived mesenchymal stem cells.
- the immune disease or inflammatory disease may be autoimmune disease, transplant rejection, arthritis, graft-versus-host disease, bacterial infection, sepsis or inflammation.
- the autoimmune diseases include Crohn's disease, erythema, atopy, rheumatoid arthritis, Hashimoto's thyroiditis, pernicious anemia, Edison's disease, type 1 diabetes, lupus, chronic fatigue syndrome, fibromyalgia, hypothyroidism and hypertension, scleroderma, Behcet's disease, Inflammatory bowel disease, multiple sclerosis, myasthenia gravis, Meniere's syndrome, Guilian-Barre syndrome, Sjogren's syndrome, vitiligo, endometriosis, psoriasis, vitiligo, systemic scleroderma, asthma or It may be ulcerative colitis.
- the present invention provides a method for inhibiting an immune response or an inflammatory response in a subject other than a human, including administering a cell or a culture thereof to the subject by adding a 15-PGDH inhibitor to a PGE2 expressing cell. To provide.
- the method may inhibit the immune response or an inflammatory response, thereby performing the prevention or treatment of an immune disease or an inflammatory disease in a subject.
- the administration may be intraperitoneal or vascular administration, direct administration to the lesion, or administration in the synovial cavity of the joint.
- the subject may include all animals except cattle, dogs, pigs, chickens, sheep, horses or humans.
- the present invention is to provide a method for producing an immunosuppressant or anti-inflammatory agent comprising the step of culturing by adding a 15-PGDH inhibitor to PGE2 expressing cells.
- the concentration of the 15-PGDH inhibitor is 0.01 to 100 ⁇ g / ml with respect to the medium, and the time of addition and incubation may be 0.1 to 200 hours.
- the present invention includes the step of culturing PGE2 expressing cells in a medium to which 15-PGDH inhibitor is added, wherein the cells secrete PGE2 (Prostaglandin E2) or TGF- ⁇ 1 (Transforming growth factor beta 1). It is intended to provide a method for producing a characteristic PGE2 or TGF- ⁇ 1.
- the present invention seeks to provide an implant comprising a cell expressing PGE2 and a 15-PGDH inhibitor.
- the cells expressing PGE2 can be removed after being incubated with the 15-PGDH inhibitor.
- the present invention is to provide a method for producing an implant comprising the step of culturing by adding a 15-PGDH inhibitor to the cells expressing PGE2 on the implant.
- the method may further comprise the step of removing the cells after the culture.
- the present invention seeks to provide a complex comprising a PGE2 expressing cell and a 15-PGDH inhibitor.
- 15-PGDH inhibitor may be bound to PGE2 of the cells.
- a 15-PGDH inhibitor may be bound to PGE2 of the cells to activate PGE2.
- the present invention seeks to provide a culture obtained by adding PGE2 expressing cells and a 15-PGDH inhibitor.
- compositions for the prevention or treatment of immunological and inflammatory diseases is a cell therapy that can be used economically without side effects and replace the existing immunosuppressive agents and inflammatory inhibitors known to have side effects, such as Clone's disease, rheumatoid arthritis, atopy It can be usefully used for the prevention or treatment of immune diseases such as autoimmune diseases and inflammatory diseases.
- the present invention is to provide a pharmaceutical composition for preventing or treating wounds comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). .
- 15-PGDH 15-hydroxyprostaglandin dehydrogenase
- PGE2 prostaglandin E2
- the cells are human adult stem cells, human pluripotent stem cells, induced pluripotent stem cells, embryonic stem cells of animals or adults of animals Stem cells.
- the adult stem cells may be mesenchymal stem cells, human tissue-derived mesenchymal stromal cells, human tissue-derived mesenchymal stem cells, multipotent stem cells or amniotic epithelial cells.
- the adult stem cells are umbilical cord-derived mesenchymal stem cells, umbilical cord blood-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, fat-derived mesenchymal stem cells, muscle-derived mesenchymal stem cells, nerve-derived mesenchymal stem cells, skin It may be derived mesenchymal stem cells, amnion derived mesenchymal stem cells or placental derived mesenchymal stem cells.
- the wound is contusion or bruise, non-healing traumatic wound, destruction of tissue by irradiation, abrasion, bone necrosis, laceration ), Avulsion, penetrated wounds, gunshot wounds, cuts, burns, frostbite, skin ulcers, dry skin, keratosis, cracking, rupture, dermatitis, pain caused by dermatophytosis, surgery, Vascular disease wounds, corneal wounds, bedsores, swellings, conditions related to diabetes and circulatory failure such as diabetic skin erosion, chronic ulcers, sutures after plastic surgery, spinal injury wounds, gynecological wounds, chemical wounds and acne It may include.
- the wound may include, but is not limited to, abrasions, lacerations, cuts, cuts, crystallizations, penetrating wounds or skin ulcers.
- the present invention is to provide a cosmetic composition comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2).
- 15-PGDH 15-hydroxyprostaglandin dehydrogenase
- PGE2 prostaglandin E2
- the cosmetic composition may be used for the purpose of skin improvement, wrinkle improvement, hair growth, hair loss prevention, rinse improvement.
- the cosmetic composition may be in the form of gel, lipstick, aerosol or emulsion in the form of a solution, lotion, water dispersed in oil or oil dispersed in water.
- the cosmetic composition is an oil, a fat, an emulsifier, a humidifier, a humectant, a softener, a preservative, a surfactant, a thickening agent, a perfume, a pigment, a dye and an alcohol, a polyol, an electrolyte, a silicone derivative, and the like. It can be prepared by combining commonly used components such as additives.
- ADSC adipose stem cells
- mice were 5 weeks old. BALB / c mice (18–20 g initial weight) were used in the experiment after adapting the males in the laboratory for one week. During the experiment, water was freely ingested. The room temperature (22 ⁇ 2 °C) and relative humidity (60 ⁇ 5%) were properly maintained, and the contrast (06:00 AM ⁇ 18: 00 PM) was adjusted every 12 hours. .
- DNCB to be used for primary and secondary sensitization was dissolved in acetone: olive oil mixed solution (4: 1) at concentrations of 1% (primary sensitization) and 0.2% (secondary sensitization).
- Adipose stem cell culture was approved by public IRB, and the adipose tissue was supplied from Diete Hospital (Seoul). The separation of adipose stem cells from fat was carried out as follows. First, collagenase is used to decompose tissues, and to isolate only a single cell with a cell strainer and incubate it with a culture medium in a culture flask while continuously growing cells through passage. All cells used in this test were 5 passage cells.
- Human bone marrow-derived mesenchymal stem cells hereinafter referred to as bone marrow stem cells, BMSC
- human umbilical cord-derived mesenchymal stem cells hereinafter called umbilical cord stem cells, UCSC
- the 15-PGDH inhibitor was supplied from Cho Hun's laboratory and used at a final concentration of 5 uM.
- each stem cell was treated with a culture medium with 15-PGDH inhibitor for 4 to 12 hours (priming), and the cells washed three times with physiological saline were collected and used for testing (fat stem cells priming with 15-PGDH inhibitor).
- Umbilical cord stem cells priming with PADSC, 15-PGDH inhibitor; Bone marrow stem cells priming with PUCSC, 15-PGDH inhibitor; PBMSC The culture medium used to test 15-PGDH inhibitor in each stem cell with culture medium After the time treatment, the culture was replaced with fresh culture and then used for 24 hours.
- adipose stem cells The injection of adipose stem cells was injected into the tail of the mouse by intravenous injection.
- the fibroblast (fibroblast) administration group and 15-PGDH inhibitor treated adipose stem cells were tested in the same manner as the adipose stem cells.
- the schedule of administration of adipose stem cells (FIG. 1) was administered on the 12th day of the test day as a single dose and confirmed the gross lesion for 2 weeks after the administration.
- the experimental group was as follows and the group was separated without statistical significance (Table 1). After one day of secondary sensitization, the group was newly separated, and the cell group was administered (1X10 6 cells / head) to the vein and the same amount of physiological saline was administered to the Negative control group and the DNCB control group.
- the macroscopic evaluation assessed the gross relief effect for each mouse once daily from the time of group separation. Severity of disease from dryness, excoriation, erythema and edema, depending on symptoms, from 0 to 3 points (0, none; 1, mild; 2, moderate) 3, severe), and added and evaluated the values. Every three days after administration, the values were summed up and the mean change of each group was compared.
- Serum IgE, PGE2 and TGFb1 levels were measured in a microplate reader using a mouse ELISA kit, respectively.
- NO synthase increases NO production and is known to increase NO content at sites showing pathological tissue damage.
- Increasing NO in dermal cells also promotes an increase in ROS that induces lipid peroxidation.
- NO is a factor that causes inflammation by expressing a large amount in inflammatory diseases such as allergy and asthma, and the variety of functions expressed by NO is known to vary depending on the concentration and activation of target cells.
- the NO (nitric oxide) content was measured using Griess reaction of NO2- (nitrite), a stable NO oxide.
- MDA oxidative damage (inflammation) of tissue cells as an oxide of polyunsaturated fatty acids by oxidation.
- TBA thiobarbituric acid
- T cells isolated from human peripheral blood were activated by treatment with several mitogens (anti-CD3, PHA, IL-2) and co-cultured with ADSC, UCSC, BMSC, PADSC, PUCSC, and PBMSC for 5 days each. ) And the proliferation rate of T cells was measured using the ADAM cell counter at the end of the culture.
- Mesenchymal stem cell concentration in co-cuture was 1 ⁇ 10 4 (10: 1), 5 ⁇ 10 4 (5: 1), 1 ⁇ 10 5 (1: 1) compared to T cells.
- use a transwell plate to prevent cell contact between T cells and mesenchymal stem cells. Culture was also conducted. The number of cells counted by the cell counter was tabulated by converting each cell number to% based on the activated T cells without co-culture (100% cell number).
- T cells and mesenchymal stem cells treated with 15-PGDH inhibitor were co-cultured in Transwell for 5 days, such as T cells isolated from human peripheral blood and culture medium treated with various mitogens. Surface antibodies of T cells were analyzed using (FACS).
- Adipose stem cells, umbilical cord stem cells, and bone marrow stem cells were treated with or without 15-PGDH inhibitor for 4 to 12 hours, and then replaced with fresh culture medium without 15-PGDH inhibitor added and cultured for 24 hours. Collected. Dermal fibroblasts (HDF) and epidermal keratinocytes (HEK) were inoculated with 20,000 cells in 6 wells one day before the test and replaced with the cultures collected above on the test day. Then, the cells were incubated in a 5% CO2 incubator, and after 3 days, cell numbers were counted with a cell counter to confirm viable cell numbers. All tests were triplicated to calculate standard deviation values.
- HDF Dermal fibroblasts
- HEK epidermal keratinocytes
- FIG. 2A Anti-inflammatory effects of adipose stem cells were observed in a mouse model of DNCB-induced allergic dermatitis (AD) (FIG. 2A). Every 3 days after MSC administration, dryness, abrasion, erythema, and edema were observed and the severity was assigned to 0-3 points to compare the significance between the groups (Fig. 2B).
- ADSC and PADSC reduced the severity of the disease compared to the DNCB group from 10 days later (Day 21).
- tissue structure Changes in tissue structure, mast cell infiltration and degranulation were observed in mice with anti-inflammatory effects caused by adipose stem cells.
- tissues such as mice were extracted to prepare paraffin blocks, and the tissues were sliced to a thickness of 6 ⁇ m to perform H & E staining and toluidin blue staining. Each performed H & E staining on tissues (FIG. 3).
- IgE, PGE2 and TGF- ⁇ 1 levels were performed using an Elisa kit to observe changes in inflammatory markers and inflammatory cytokine (FIG. 4).
- NO nitric oxide
- MDA malondialdehyde
- the ADSC and PADSC-administered groups showed lower NO levels in the serum than the control group, and the tissues showed a greater NO inhibitory effect than the PADSC (FIG. 5A).
- Serum MDA levels were similar to those of normal mice in the PADSC-administered group, but not significantly different from the negative control group (N.C) in the ADSC group. In the case of tissue, no significant change was found (FIG. 5B).
- mesenchymal stems were significantly increased in an environment where the ratio of T cells and mesenchymal stem cells was 1: 1.
- the cells suppressed the proliferation of activated T cells, and in the case of adipose stem cells and umbilical cord stem cells, mesenchymal stem cells treated with 15-PGDH were found to inhibit the proliferation of activated T cells more (FIG. 6A).
- the ratio of T cells and mesenchymal stem cells was significant in a 1: 1 environment.
- mesenchymal stem cells inhibited the proliferation of activated T cells and all mesenchymal stem cells treated with 15-PGDH inhibited the proliferation of activated T cells more (FIG. 6B).
- flow cytometry was performed by collecting mesenchymal stem cells and mesenchymal stem cells treated with 15-PGDH inhibitor and cocultured T cells, respectively. At this time, the antigens of CD4, CD25 and FoxP3 which are phenotypes of regulatory T cells were analyzed (FIG. 7).
- T cells co-cultured with umbilical stem cells treated with 15-PGDH inhibitor PUCSC
- T cells co-cultured with umbilical stem cells (UCSC) untreated with 15-PGDH inhibitor 44%)
- the Treg population of 92% was increased by 48% compared to the cells, and the T cells co-cultured with BMSC showed 39% of Treg population, while bone marrow stem cells treated with 15-PGDH inhibitor (PBMSC).
- PBMSC bone marrow stem cells treated with 15-PGDH inhibitor
- T cells co-cultured increased the Treg population by 98%.
- PADSC, PUCSC, and PBMSC priming ADSC, UCSC, BMSC, and 15-PGDH inhibitors were cultured for 24 hours, and then the cultures were harvested to compare the growth rate of human skin fibroblasts (HDF) and human epidermal keratinocytes (HEK). (FIG. 8).
- the growth rate of HDF was significantly increased when cultured with PADSC and PUCSC cultures priming 15-PGDH inhibitor.
- the growth rate was increased when the 15-PGDH inhibitor was incubated with PADSC and PUCSC cultures.
- the pharmaceutical composition according to the present invention has anti-inflammatory effect, serum IgE reduction effect, NO and MDA reduction effect, activated T cell proliferation inhibitory effect, and dermal fibroblast and epithelial keratinocyte proliferation ability in DNCB-induced AD mouse model Can have an effect. Therefore, the pharmaceutical composition according to the present invention is expected to be effectively used as an anti-inflammatory disease treatment agent, an inflammatory disease treatment agent or a wound treatment agent.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Epidemiology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Birds (AREA)
- Emergency Medicine (AREA)
- Dermatology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Description
본 발명은 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 조성물과 그의 용도에 관한 것이다. 보다 상세하게는, PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 면역질환, 염증질환, 또는 창상의 예방 또는 치료용과 약제학적 조성물에 관한 것으로, 상기 약제학적 조성물, 상기 세포 또는 그것의 배양물을 피험체에 투여하는 단계를 포함하는 피험체의 면역반응, 염증반응 또는 창상을 억제하는 방법, 상기 세포 또는 그것의 배양물을 이용하여 면역억제제, 항염증제 또는 창상 치료제를 제조하는 방법, PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양하는 단계를 포함하는 PGE2의 제조 방법, PGE2를 발현하는 세포 및 15-PGDH 억제제를 포함하는 이식체, 상기 이식체의 제조 방법, PGE2를 발현하는 세포 및 15-PGDH 억제제를 포함하는 복합체 및 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 얻은 배양물에 관한 것이다.The present invention relates to a composition comprising a cell cultured by adding a 15-PGDH inhibitor to a cell expressing PGE2 or a culture thereof and a use thereof. More specifically, the present invention relates to a pharmaceutical composition for the prevention or treatment of immune diseases, inflammatory diseases, or wounds, comprising cells cultured with the addition of a 15-PGDH inhibitor to PGE2 expressing cells or a culture thereof. A pharmaceutical composition, a method of inhibiting an immune response, an inflammatory response or a wound in a subject comprising administering the cell or a culture thereof to the subject, an immunosuppressive agent, an anti-inflammatory agent using the cell or its culture Or a method for preparing a wound therapeutic agent, a method of preparing PGE2 comprising culturing by adding a 15-PGDH inhibitor to a cell expressing PGE2, a transplant comprising a cell expressing PGE2 and a 15-PGDH inhibitor, the transplant Method for preparing a sieve, a complex comprising a PGE2 expressing cell and a 15-PGDH inhibitor and a culture obtained by adding a 15-PGDH inhibitor to a PGE2 expressing cell It relates.
프로스타글란딘 E2(이하 PGE2)는 프로스타글란딘 E2(prostaglandin E2: 5Z, 11(알파)13E, 15S)-11, 15-디하이드록시-9-옥소-프로스타-5, 13-디엔-1-오익산[(5Z, 11(alpha), 13E, 15S)-11, 15-Dihydroxy-9-oxo-prosta-5, 13-dien-1-oic acid])로 표시되는 화합물이며, 생리학적, 그리고 병리학적 환경에서 가장 널리 생산되는 프로스타글란딘이다(Ushikubi F etal., J. Pharmacol. Sci. 83:279, 2000). PGE2는 종래 출산 준비를 위해 자궁 경부에 사용되었으며 실제로 약학적으로 출산유도에 이용되어 온바, 다음 상표명으로 질 좌약의 형태로 판매 중이다: Cervidil(포레스트 주식 회사), Prostin E2 (화이자 주식 회사), Propess (Ferring 제약 회사), Glandin (Nabiqasim 제약, 파키스탄). 또한 최근 새롭게 면역억제, 조절제로서의 용도가 각광받고 있는데, 이는 대식세포에 의해 생산되는 인터류킨-1베타 및 TNF 알파와 같은 사이토카인의 분비를 억제하는 역할을 하며, helper T1 cell의 분화를 억제하는 것이 보고되어 있기 때문이다(Harris SG etal., TrendsImmunol., 23:144, 2002). 또한 invitro에서, PGE2가 인터류킨-2 및 IFN-감마와 같은 사이토카인의 생산을 억제하여, 인간 및 쥐의 T 세포의 분화를 억제하는 것 또한 보고되었다(Goodwin JS etal., J. clin. Immunol., 3:295, 1983). 또한, 위 실험 결과가 보고되어 온바, 면역 조절제로 이용 가능성이 풍부하여, 경제적이고 간편한 생산 방법이 요구되어 왔다. TGF-β1(Transforming growth factor beta 1)은 면역 억제 및 항염증제로 알려져 있으며, PGE2와 마찬가지로 면역 조절제로 이용 가능성이 풍부하여 경제적이고 간편한 생산 방법이 요구되어 왔다.Prostaglandin E2 (hereinafter PGE2) is prostaglandin E2 (5Z, 11 (alpha) 13E, 15S) -11, 15-dihydroxy-9-oxo-prostar-5, 13-diene-1-oic acid [ (5Z, 11 (alpha), 13E, 15S) -11, 15-Dihydroxy-9-oxo-prosta-5, 13-dien-1-oic acid], and the physiological and pathological environment Is the most widely produced prostaglandin (Ushikubi F et al., J. Pharmacol. Sci. 83: 279, 2000). PGE2 has traditionally been used in the cervix to prepare for birth and is actually pharmacologically used to induce birth and is sold in the form of vaginal suppositories under the following trade names: Cervidil (Forest Corporation), Prostin E2 (Pfizer Corporation), Propess (Ferring Pharmaceutical Company), Glandin (Nabiqasim Pharmaceuticals, Pakistan). In addition, recently, the use of immunosuppressive and modulators has been in the spotlight, which plays a role in inhibiting the secretion of cytokines such as interleukin-1 beta and TNF alpha produced by macrophages, and inhibiting the differentiation of helper T1 cells. (Harris SG et al., Trends Immunol., 23: 144, 2002). In invitro, it has also been reported that PGE2 inhibits the production of cytokines such as interleukin-2 and IFN-gamma, thereby inhibiting the differentiation of human and rat T cells (Goodwin JS et al., J. clin. Immunol. , 3: 295, 1983). In addition, since the above experimental results have been reported, the possibility of using as an immunomodulator is rich, and economical and simple production methods have been required. TGF-β1 (Transforming growth factor beta 1) is known as an immunosuppressive and anti-inflammatory agent, and like PGE2, has been widely available as an immunomodulator, and thus an economical and convenient production method has been required.
한편, 면역억제에는 그 반응만을 억제하는 특이적 억제제와 비특이적 억제제가 있으며, 이론적으로 특이적 억제제의 작용이 뛰어나야 하나, 비특이적 억제제가 주로 사용된다. 임상적으로 가장 흔히 사용되는 면역억제제로는 싸이클로스포린 (cyclosporine, Neoral, Cipol A), 아자치오프린(imuran), 프레드니솔론(일종의 스테로이드)이 있다. 위의 세 가지는 함께 병용했을 때가 따로 복용했을 때 보다 부작용이 적고 면역억제 효과도 가장 높은 것으로 판명되었다. 최근에는 FK 506, RATG, OKT3, Cellcept등 여러 가지 면역억제제가 개발되어 사용되고 있다. 이들 면역억제제는 항원자극에서 항체생성에 이르는 과정 중 대식세포에 의한 항원의 탐식, 림프구 등에 의한 항원 인식, 세포 분열, T세포와 B세포의 분열, 항체 생성 등 몇 가지 과정을 저해시킴으로써 면역 억제를 야기한다. 대부분 항종양 활성을 가지고 있는데, 그 이유는 DNA 장애, DNA 합성 저지 등을 매개로 하여 세포 분열을 저지하기 때문이다.On the other hand, immunosuppression includes specific inhibitors and nonspecific inhibitors that inhibit only the response, and theoretically, the action of specific inhibitors should be excellent, but nonspecific inhibitors are mainly used. The most commonly used immunosuppressive agents are cyclosporine (Neoral, Cipol A), azacioprine (imuran) and prednisolone (a type of steroid). The three were found to have fewer side effects and the highest immunosuppressive effects when used together when used separately. Recently, various immunosuppressive agents such as FK 506, RATG, OKT3, and Cellcept have been developed and used. These immunosuppressive agents inhibit immunosuppression by inhibiting several processes such as phagocytosis of antigen by macrophages, antigen recognition by lymphocytes, cell division, T cell and B cell division, and antibody production. Cause. Most of them have antitumor activity because they inhibit cell division through the mediation of DNA disorders and the inhibition of DNA synthesis.
그러나, 이에 따른 대표적인 부작용으로 고혈압과 신독성(콩팥기능이 저하됨)이 있으며 이 부작용의 발생률이 높기 때문에 사용할 때 충분히 경과를 관찰해야 하는 등의 문제가 있어 왔다. 그 외 부작용으로 드물게 떨림, 발작, 간염, 담액 저류, 혈중 뇨산증가, 근육기력 저하, 조모증 (hypertrichosis), 치은 비대(gingival hypertrophy)등이 있다. 흔히 쓰이는 억제제 중 아자치오프린은 백혈구수치의 감소, 빈혈, 혈소판 감소 등 골수 기능을 억제하기도 하며 췌장염, 간염, 담즙저류와 함께 드물게 탈모, 발열 등을 보이는 합병증이 있을 수 있다. 스테로이드 제제의 하나인 프레드니솔론은 면역 억제제 중 가장 먼저 사용되기 시작하였으며 가장 광범위한 억제 작용을 보인다. 식욕을 증진시켜주며 어깨와 등의 근육을 증가시키고 일시적으로는 행복감을 가져다 주기도 하나 이러한 스테로이드제제는 동맥 경화증을 촉진시킬 뿐 아니라 고혈압, 위궤양, 당뇨, 성장 저해, 골다공증, 백내장, 녹내장 등을 일으키므로 주의해야 할 약물이다. However, the typical side effects are hypertension and nephrotoxicity (decrease in kidney function), and the incidence of these side effects has been high, so that there has been a problem of observing the course when used sufficiently. Other side effects rarely include tremor, seizures, hepatitis, biliary retention, increased blood uric acid, muscle weakness, hypertrichosis, and gingival hypertrophy. Among the commonly used inhibitors, azachioprine inhibits bone marrow function, such as decreased white blood cell count, anemia, and platelet reduction. In addition, pancreatitis, hepatitis, and biliary retention may rarely cause hair loss and fever. Prednisolone, one of the steroid agents, was the first to be used among immunosuppressants and has the broadest inhibitory action. It may increase appetite, increase muscles in the shoulders and back, and temporarily bring happiness, but these steroids not only promote atherosclerosis, but also cause high blood pressure, gastric ulcers, diabetes, growth inhibition, osteoporosis, cataracts, and glaucoma. The drug to watch out for.
장기이식과 조혈모세포이식 등과 같은 동종 이식은, 21세기에 이르러 이루어낸 획기적인 의료 성과로서, 확장성 심근병증과 같은 심부전, 만성 신부전 및 난치성 혈액질환 등의 말기 질환에서 근본적인 치료 방법으로 이용되고 있다. 그러나 동종 이식 후 발생되는 치명적인 합병증인 이식 장기의 거부 또는 이식편 대 숙주병(graft-versus-host-desease, GVHD)와 같은 면역학적 반응을 극복해야 하는 문제점이 있다. 현재 쓰이는 기술로는, 이러한 면역학적 반응을 최소화하기 위하여 이식 후 나타나는 면역반응은 동종항원을 인지한 T 세포에 의해서 세포성 면역으로 발생되어 T 세포의 면역반응을 조절하기 위한 치료법(Ikehara S, Exp. Hematol., 31:1142, 2003; First MR, Transplantation, 77:88, 2004), 즉, 면역억제제인 싸이클로스포린, FK506을 이용하여 T 세포의 interleukin(IL)-2의 생성을 억제하여 면역반응을 조절하는 치료법을 이용하고 있으나, 부작용이 없으면서도 경제적으로 사용될 수 있는 면역억제제의 개발이 필요하여 왔다.Allogeneic transplantation, such as organ transplantation and hematopoietic stem cell transplantation, is a breakthrough medical achievement made in the 21st century, and is being used as a fundamental treatment method in late stage diseases such as heart failure, chronic renal failure, and intractable blood diseases such as dilated cardiomyopathy. However, there is a problem in overcoming immunological reactions such as graft-versus-host-desease (GVHD) or rejection of transplanted organs, which are fatal complications after allogeneic transplantation. In current technology, in order to minimize these immunological reactions, the immune response after transplantation is generated by cellular immunity by T cells which recognize allogeneic antigens, thereby treating the T cell immune response (Ikehara S, Exp Hematol., 31: 1142, 2003; First MR, Transplantation, 77:88, 2004), ie, the immune response by inhibiting the production of interleukin (IL) -2 in T cells using the immunosuppressive cyclosporin, FK506. Although the treatment to control the use of, there is a need for the development of immunosuppressive agents that can be used economically without side effects.
한편, 중간엽 줄기세포(mesenchymal stem cells)의 면역조절능력에 대한 정확한 작용기전은 아직 상세히 알려지지 않았으며, 다만 최근 중간엽 줄기세포와 관련하여 다음과 같은 몇 가지 보고들만이 제시되었다. 첫째, 중간엽 줄기세포는 APC(antigen presenting cell)를 억제하는 효과가 있는 것으로 보인다. 특정 조건에서 배양과정 중 첨가된 단핵구 수에 비례하여 면역반응 정도가 변하는 것으로 보아 결국 단핵구가 면역억제효과에 관여하는 것으로 추정할 수 있다. 둘째, 중간엽 줄기세포가 T 세포의 증식속도를 조절함으로써, 면역억제 효과를 나타내는 것으로 보인다. T 세포를 중간엽 줄기세포와 함께 배양하는 경우, cyclin D2가 억제됨에 따라 T 세포가 세포주기 중 G0/G1 시기에 머물러 더 이상 증가하지 않는다. 또한 중간엽 줄기세포를 제거한다 하더라도 증식능력은 지속적으로 저하되는 것으로 보고되었다(Glennie S etal., Blood, 105:2821, 2005).On the other hand, the precise mechanism of action on the immunomodulatory capacity of mesenchymal stem cells is not yet known in detail, but only a few reports regarding the mesenchymal stem cells have recently been presented. First, mesenchymal stem cells appear to have an effect of inhibiting APC (antigen presenting cell). Under certain conditions, the degree of immune response changes in proportion to the number of monocytes added during the culturing process, suggesting that monocytes may be involved in immunosuppressive effects. Second, mesenchymal stem cells appear to exhibit immunosuppressive effects by controlling the proliferation rate of T cells. When T cells are incubated with mesenchymal stem cells, T cells stay at G0 / G1 phase of the cell cycle and no longer increase as cyclin D2 is inhibited. It has also been reported that proliferative capacity continues to decline even when mesenchymal stem cells are removed (Glennie S et al., Blood, 105: 2821, 2005).
이에, 본 발명자들은 세포를 이용하여 더 효과적으로 면역 또는 염증을 조절하는 방법을 개발하고자 예의 노력한 결과, 15-PGDH 억제제를 세포에 처리하는 경우 PGE2가 유의적으로 과량 생산되어 면역반응을 보다 더 효과적으로 억제함으로써, 아토피 동물모델 또는 자가면역질환 동물모델에서 치료 효과가 있음을 확인하고 본 발명을 완성하였다. 또한, 본 발명자들은 15-PGDH 억제제를 세포에 처리하는 경우 PGE2가 유의적으로 과량 생산되어 창상치료에 이용될 수 있음을 확인하였다. Accordingly, the present inventors have diligently tried to develop a method of more effectively controlling immunity or inflammation using cells. As a result, when the 15-PGDH inhibitor is treated to cells, PGE2 is significantly produced to suppress the immune response more effectively. By doing so, it was confirmed that there is a therapeutic effect in an atopy animal model or an autoimmune disease animal model and completed the present invention. In addition, the present inventors confirmed that when treated with 15-PGDH inhibitor cells, PGE2 is significantly overproduced and used for wound treatment.
본 명세서에서 사용된 용어 “15-PGDH 억제제(inhibitor)”는 15-PGDH의 활성을 억제시키는 물질(길항제)을 의미한다. 예를 들면, 상기 15-PGDH 억제제는 사이클로옥시게나제 억제제(cyclooxygenase inhibitor), 플라보노이드, 피토페놀계(phytophenolic) 화합물 및 퍼옥시좀 증식자-활성화 수용체 감마(peroxisome proliferator-activated receptor λ: PPAR λ)의 길항제를 포함하는 유기화?d물로 이루어진 군으로부터 1종 이상 선택될 수 있으나, 이에 한정되는 것은 아니다. As used herein, the term "15-PGDH inhibitor (inhibitor)" refers to a substance (antagonist) that inhibits the activity of the 15-PGDH. For example, the 15-PGDH inhibitor is a cyclooxygenase inhibitor, a flavonoid, a phytophenolic compound, and a peroxysome proliferator-activated receptor λ (PPAR λ). One or more may be selected from the group consisting of organic compounds containing antagonists of d, but is not limited thereto.
본 명세서에서 사용된 용어 "세포"는 다양한 조직으로 분화할 수 있는 능력을 가진 줄기세포를 포함하고 있다.The term "cell" as used herein includes stem cells having the ability to differentiate into various tissues.
본 명세서에서 사용된 용어 "예방"은 본 발명에 따른 조성물의 투여로 면역질환 또는 염증질환의 발병을 억제 또는 지연시키는 모든 행위를 의미하며, "치료"는 본 발명에 따른 약제학적 조성물의 투여로 면역질환 또는 염증질환의 증세가 호전되거나 이롭게 되는 모든 행위를 의미한다.As used herein, the term "prevention" refers to any action that inhibits or delays the development of an immune disease or inflammatory disease by administration of a composition according to the present invention, and "treatment" refers to the administration of a pharmaceutical composition according to the present invention. Means any activity that improves or benefits the symptoms of an immune or inflammatory disease.
본 명세서에서 사용된 용어 "이식체"는 손상된 부위를 외부로부터 격리하거나 이식된 세포나 분비된 치료 물질이 머물러 있도록 하는 지지체로서, 인체 또는 포유동물에 이식될 수 이는 물질을 의미한다. 이와 같은 이식체는 조직공학용 지지체로서 생분해성을 가지는 합성고분자와 천연재료 등 당업계에 다양하게 사용되는 물질을 제한 없이 포함한다.As used herein, the term “graft” refers to a material that can be implanted in a human or mammal as a support that isolates the damaged area from the outside or allows the transplanted cells or secreted therapeutic material to remain. Such implants include, without limitation, various materials used in the art such as synthetic polymers and natural materials having biodegradability as a support for tissue engineering.
본 발명은 PGE2(prostaglandin E2)를 발현하는 세포에 15-PGDH(15-hydroxyprostaglandin dehydrogenase) 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 면역질환 또는 염증질환의 예방 또는 치료용 약제학적 조성물을 제공하고자 한다. The present invention provides a pharmaceutical composition for the prevention or treatment of immune diseases or inflammatory diseases, including cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). To provide.
본 발명은 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 피험체에 투여하는 단계를 포함하는 인간을 제외한 피험체의 면역 반응 또는 염증반응을 억제하는 방법을 제공하고자 한다. The present invention provides a method for inhibiting an immune response or an inflammatory response in a subject other than a human, including administering a cell or a culture thereof to the subject by adding a 15-PGDH inhibitor to a PGE2 expressing cell. To provide.
본 발명은 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양하는 단계를 포함하는 면역억제제 또는 항염증제의 제조 방법을 제공하고자 한다. The present invention is to provide a method for producing an immunosuppressant or anti-inflammatory agent comprising the step of culturing by adding a 15-PGDH inhibitor to PGE2 expressing cells.
본 발명은 PGE2를 발현하는 세포를 15-PGDH 억제제를 첨가한 배지에서 배양하는 단계를 포함하며, 배양시 상기 세포에서 PGE2(Prostaglandin E2) 또는 TGF-β1(Transforming growth factor beta 1)이 분비되는 것이 특징인 PGE2 또는 TGF- β1의 제조 방법을 제공하고자 한다. The present invention includes the step of culturing PGE2 expressing cells in a medium to which 15-PGDH inhibitor is added, wherein the cells secrete PGE2 (Prostaglandin E2) or TGF-β1 (Transforming growth factor beta 1). It is intended to provide a method for producing a characteristic PGE2 or TGF-β1.
본 발명은 PGE2를 발현하는 세포 및 15-PGDH 억제제를 포함하는 이식체를 제공하고자 한다. The present invention seeks to provide an implant comprising a cell expressing PGE2 and a 15-PGDH inhibitor.
본 발명은 이식체 상에서 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양하는 단계를 포함하는 이식체 제조 방법을 제공하고자 한다. The present invention is to provide a method for producing an implant comprising the step of culturing by adding a 15-PGDH inhibitor to the cells expressing PGE2 on the implant.
본 발명은 PGE2를 발현하는 세포 및 15-PGDH 억제제를 포함하는 복합체를 제공하고자 한다. The present invention seeks to provide a complex comprising a PGE2 expressing cell and a 15-PGDH inhibitor.
본 발명은 PGE2를 발현하는 세포 및 15-PGDH 억제제 첨가하여 얻은 배양물을 제공하고자 한다. The present invention seeks to provide a culture obtained by adding PGE2 expressing cells and a 15-PGDH inhibitor.
또한, 본 발명은 PGE2(prostaglandin E2)를 발현하는 세포에 15-PGDH(15-hydroxyprostaglandin dehydrogenase) 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 창상의 예방 또는 치료용 약제학적 조성물을 제공하고자 한다. The present invention also provides a pharmaceutical composition for preventing or treating wounds comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). I would like to.
또한, 본 발명은 PGE2(prostaglandin E2)를 발현하는 세포에 15-PGDH(15-hydroxyprostaglandin dehydrogenase) 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 화장품 조성물을 제공하고자 한다. In addition, the present invention is to provide a cosmetic composition comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2).
본 발명에 따른 약제학적 조성물은 DNCB로 유발된 AD 마우스 모델에서 항염증 효과, 혈청 내 IgE 감소 효과, NO 및 MDA 감소 효과, 활성화된 T 세포 증식 억제 효과, 및 피부섬유아세포 및 상피각직세포 증식능 향상 효과를 가질 수 있다. The pharmaceutical composition according to the present invention has anti-inflammatory effect, serum IgE reduction effect, NO and MDA reduction effect, activated T cell proliferation inhibitory effect, and dermal fibroblast and epithelial keratinocyte proliferation ability in DNCB-induced AD mouse model Can have an effect.
도 1은 본 발명의 실시예에 따른 시험 일정을 나타낸다. 1 shows a test schedule according to an embodiment of the present invention.
도 2는 일자별 DNCB로 유도된 아토피 유발 마우스의 육안병변 결과를 나타낸다. Figure 2 shows the results of macroscopic lesions of atopy-induced atopy-induced mice by day.
도 3은 DNCB로 유도된 아토피 유발 마우스의 피부 조직학적 분석 결과를 나타낸다. Figure 3 shows the results of skin histology of DNCB-induced atopic dermatitis mice.
도 4는 DNCB로 유도된 아토피 유발 마우스 혈청에서 IgE 및 TGF-β1의 농도를 나타낸다.4 shows the concentrations of IgE and TGF-β1 in DNCB induced atopy induced mouse serum.
도 5는 DNCB로 유도된 아토피 유발 마우스의 혈청 및 피부조직에서 NO 및 MDA의 농도를 나타낸다. Figure 5 shows the concentrations of NO and MDA in serum and skin tissue of DNCB induced atopic dermal mice.
도 6은 중간엽줄기세포의 활성화 T세포 면역억제작용을 나타낸다. Figure 6 shows the activated T-cell immunosuppression of mesenchymal stem cells.
도 7은 활성화T세포 면역표현형 분석 결과를 나타낸다. Figure 7 shows the results of activated T cell immunophenotype analysis.
도 8은 피부섬유아세포와 상피각질세포의 증식능 분석 결과를 나타낸다. 8 shows the results of analysis of proliferative capacity of dermal fibroblasts and epidermal keratinocytes.
1. 면역질환 또는 염증질환용 약제학적 조성물1. Pharmaceutical composition for immune disease or inflammatory disease
제1구현예에 따르면,According to the first embodiment,
본 발명은 PGE2(prostaglandin E2)를 발현하는 세포에 15-PGDH(15-hydroxyprostaglandin dehydrogenase) 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 면역질환 또는 염증질환의 예방 또는 치료용 약제학적 조성물을 제공하고자 한다. The present invention provides a pharmaceutical composition for the prevention or treatment of immune diseases or inflammatory diseases, including cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). To provide.
본 발명의 면역질환 또는 염증질환의 예방 또는 치료용 약제학적 조성물에 따르면, 상기 세포는 인간의 성체줄기세포, 인간의 만능줄기세포, 유도만능줄기세포(induced pluripotent stem cells), 동물의 배아줄기세포 또는 동물의 성체줄기세포일 수 있다. 상기 성체줄기세포는 중간엽 줄기세포, 인간 조직 유래 중간엽 기질세포(mesenchymal stromal cell), 인간 조직 유래 중간엽 줄기세포, 다분화능 줄기세포 또는 양막상피세포일 수 있다. 또한, 상기 성체줄기세포는 제대 유래 중간엽 줄기세포, 제대혈 유래 중간엽 줄기세포, 골수 유래 중간엽 줄기세포, 지방 유래 중간엽 줄기세포, 근육 유래 중간엽 줄기세포, 신경 유래 중간엽 줄기세포, 피부 유래 중간엽 줄기세포, 양막 유래 중간엽 줄기세포 또는 태반 유래 중간엽 줄기세포일 수 있다. According to the pharmaceutical composition for preventing or treating an immune disease or inflammatory disease of the present invention, the cells are human adult stem cells, human pluripotent stem cells, induced pluripotent stem cells, embryonic stem cells of animals Or adult stem cells of an animal. The adult stem cells may be mesenchymal stem cells, human tissue-derived mesenchymal stromal cells, human tissue-derived mesenchymal stem cells, multipotent stem cells or amniotic epithelial cells. In addition, the adult stem cells are umbilical cord-derived mesenchymal stem cells, umbilical cord blood-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, fat-derived mesenchymal stem cells, muscle-derived mesenchymal stem cells, nerve-derived mesenchymal stem cells, skin It may be derived mesenchymal stem cells, amnion derived mesenchymal stem cells or placental derived mesenchymal stem cells.
본 발명의 면역질환 또는 염증질환의 예방 또는 치료용 약제학적 조성물에 따르면, 상기 면역질환 또는 염증질환은 자가면역질환, 이식거부, 관절염, 이식편대숙주병, 세균감염, 패혈증 또는 염증일 수 있다. 상기 자가면역질환은 크론씨병, 홍반병, 아토피, 류마티스 관절염, 하시모토 갑상선염, 악성빈혈, 에디슨씨 병, 제1형 당뇨, 루프스, 만성피로증후군, 섬유근육통, 갑상선기능저하증과 항진증, 경피증, 베체트병, 염증성 장질환, 다발성 경화증, 중증 근무력증, 메니에르 증후군(Meniere's syndrome), 길리안-바레 증후근(Guilian-Barre syndrome), 쇼그렌 증후군(Sjogren's syndrome), 백반증, 자궁내막증, 건선, 백반증, 전신성 경피증, 천식 또는 궤양성 대장염일 수 있다. According to the pharmaceutical composition for preventing or treating an immune disease or inflammatory disease of the present invention, the immune disease or inflammatory disease may be autoimmune disease, transplant rejection, arthritis, graft-versus-host disease, bacterial infection, sepsis or inflammation. The autoimmune diseases include Crohn's disease, erythema, atopy, rheumatoid arthritis, Hashimoto's thyroiditis, pernicious anemia, Edison's disease,
제2구현예에 따르면,According to the second embodiment,
본 발명은 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 피험체에 투여하는 단계를 포함하는 인간을 제외한 피험체의 면역 반응 또는 염증반응을 억제하는 방법을 제공하고자 한다. The present invention provides a method for inhibiting an immune response or an inflammatory response in a subject other than a human, including administering a cell or a culture thereof to the subject by adding a 15-PGDH inhibitor to a PGE2 expressing cell. To provide.
본 발명의 면역 반응 또는 염증반응을 억제하는 방법에 따르면, 상기 방법은 면역 반응 또는 염증반응을 억제하여, 피험체의 면역질환 또는 염증질환의 예방 또는 치료를 수행할 수 있다. According to the method for inhibiting an immune response or an inflammatory response of the present invention, the method may inhibit the immune response or an inflammatory response, thereby performing the prevention or treatment of an immune disease or an inflammatory disease in a subject.
본 발명의 면역 반응 또는 염증반응을 억제하는 방법에 따르면, 상기 투여는 복강 또는 혈관 내 투여, 병변으로의 직접 투여 또는 관절의 활강(Synovialcavity) 내 투여일 수 있다. According to the method for inhibiting the immune response or inflammatory response of the present invention, the administration may be intraperitoneal or vascular administration, direct administration to the lesion, or administration in the synovial cavity of the joint.
본 발명의 면역 반응 또는 염증반응을 억제하는 방법에 따르면, 상기 피험체는 소, 개, 돼지, 닭, 양, 말 또는 인간을 제외한 모든 동물을 포함할 수 있다. According to the method for suppressing the immune or inflammatory response of the present invention, the subject may include all animals except cattle, dogs, pigs, chickens, sheep, horses or humans.
제3구현예에 따르면,According to the third embodiment,
본 발명은 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양하는 단계를 포함하는 면역억제제 또는 항염증제의 제조 방법을 제공하고자 한다. The present invention is to provide a method for producing an immunosuppressant or anti-inflammatory agent comprising the step of culturing by adding a 15-PGDH inhibitor to PGE2 expressing cells.
본 발명의 면역억제제 또는 항염증제의 제조 방법에 따르면, 상기 15-PGDH 억제제의 농도는 배지에 대하여 0.01 내지 100㎍/ml이며, 첨가하여 배양하는 시간은 0.1 내지 200 시간일 수 있다. According to the method for preparing an immunosuppressant or anti-inflammatory agent of the present invention, the concentration of the 15-PGDH inhibitor is 0.01 to 100 µg / ml with respect to the medium, and the time of addition and incubation may be 0.1 to 200 hours.
제4구현예에 따르면, According to the fourth embodiment,
본 발명은 PGE2를 발현하는 세포를 15-PGDH 억제제를 첨가한 배지에서 배양하는 단계를 포함하며, 배양시 상기 세포에서 PGE2(Prostaglandin E2) 또는 TGF-β1(Transforming growth factor beta 1)이 분비되는 것이 특징인 PGE2 또는 TGF- β1의 제조 방법을 제공하고자 한다. The present invention includes the step of culturing PGE2 expressing cells in a medium to which 15-PGDH inhibitor is added, wherein the cells secrete PGE2 (Prostaglandin E2) or TGF-β1 (Transforming growth factor beta 1). It is intended to provide a method for producing a characteristic PGE2 or TGF-β1.
제5구현예에 따르면,According to the fifth embodiment,
본 발명은 PGE2를 발현하는 세포 및 15-PGDH 억제제를 포함하는 이식체를 제공하고자 한다. The present invention seeks to provide an implant comprising a cell expressing PGE2 and a 15-PGDH inhibitor.
본 발명의 이식체에 따르면, 상기 PGE2를 발현하는 세포는 15-PGDH 억제제와 함께 배양된 후 제거될 수 있다. According to the implant of the present invention, the cells expressing PGE2 can be removed after being incubated with the 15-PGDH inhibitor.
제6구현예에 따르면,According to the sixth embodiment,
본 발명은 이식체 상에서 PGE2를 발현하는 세포에 15-PGDH 억제제를 첨가하여 배양하는 단계를 포함하는 이식체 제조 방법을 제공하고자 한다. The present invention is to provide a method for producing an implant comprising the step of culturing by adding a 15-PGDH inhibitor to the cells expressing PGE2 on the implant.
본 발명의 이식체의 제조 방법에 따르면, 상기 방법은 배양 이후에 세포를 제거하는 단계를 더욱 포함할 수 있다. According to the method for producing an implant of the present invention, the method may further comprise the step of removing the cells after the culture.
제7구현예에 따르면, According to the seventh embodiment,
본 발명은 PGE2를 발현하는 세포 및 15-PGDH 억제제를 포함하는 복합체를 제공하고자 한다. The present invention seeks to provide a complex comprising a PGE2 expressing cell and a 15-PGDH inhibitor.
본 발명의 복합체에 따르면, 상기 세포의 PGE2에 15-PGDH 억제제가 결합되어 있을 수 있다. 상기 세포의 PGE2에 15-PGDH 억제제가 결합되어 PGE2가 활성화될 수 있다. According to the complex of the present invention, 15-PGDH inhibitor may be bound to PGE2 of the cells. A 15-PGDH inhibitor may be bound to PGE2 of the cells to activate PGE2.
제8구현예에 따르면, According to the eighth embodiment,
본 발명은 PGE2를 발현하는 세포 및 15-PGDH 억제제 첨가하여 얻은 배양물을 제공하고자 한다. The present invention seeks to provide a culture obtained by adding PGE2 expressing cells and a 15-PGDH inhibitor.
본 발명에 따른 면역질환 및 염증질환의 예방 또는 치료용 약제학적 조성물은 부작용이 알려진 기존의 면역억제제 및 염증억제제를 대체하는, 부작용이 없고 경제적으로 사용될 수 있는 세포치료제로서 클론스병, 류마티스 관절염, 아토피 등 자가면역질환과 같은 면역질환 및 염증질환의 예방 또는 치료에 유용하게 사용될 수 있다. Pharmaceutical compositions for the prevention or treatment of immunological and inflammatory diseases according to the present invention is a cell therapy that can be used economically without side effects and replace the existing immunosuppressive agents and inflammatory inhibitors known to have side effects, such as Clone's disease, rheumatoid arthritis, atopy It can be usefully used for the prevention or treatment of immune diseases such as autoimmune diseases and inflammatory diseases.
2. 창상 치료용 약제학적 조성물2. Pharmaceutical composition for the treatment of wounds
본 발명은 PGE2(prostaglandin E2)를 발현하는 세포에 15-PGDH(15-hydroxyprostaglandin dehydrogenase) 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 창상의 예방 또는 치료용 약제학적 조성물을 제공하고자 한다. The present invention is to provide a pharmaceutical composition for preventing or treating wounds comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2). .
본 발명의 창상의 예방 또는 치료용 약제학적 조성물에 따르면, 상기 세포는 인간의 성체줄기세포, 인간의 만능줄기세포, 유도만능줄기세포(induced pluripotent stem cells), 동물의 배아줄기세포 또는 동물의 성체줄기세포일 수 있다. 상기 성체줄기세포는 중간엽 줄기세포, 인간 조직 유래 중간엽 기질세포(mesenchymal stromal cell), 인간 조직 유래 중간엽 줄기세포, 다분화능 줄기세포 또는 양막상피세포일 수 있다. 또한, 상기 성체줄기세포는 제대 유래 중간엽 줄기세포, 제대혈 유래 중간엽 줄기세포, 골수 유래 중간엽 줄기세포, 지방 유래 중간엽 줄기세포, 근육 유래 중간엽 줄기세포, 신경 유래 중간엽 줄기세포, 피부 유래 중간엽 줄기세포, 양막 유래 중간엽 줄기세포 또는 태반 유래 중간엽 줄기세포일 수 있다. According to the pharmaceutical composition for preventing or treating wounds of the present invention, the cells are human adult stem cells, human pluripotent stem cells, induced pluripotent stem cells, embryonic stem cells of animals or adults of animals Stem cells. The adult stem cells may be mesenchymal stem cells, human tissue-derived mesenchymal stromal cells, human tissue-derived mesenchymal stem cells, multipotent stem cells or amniotic epithelial cells. In addition, the adult stem cells are umbilical cord-derived mesenchymal stem cells, umbilical cord blood-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, fat-derived mesenchymal stem cells, muscle-derived mesenchymal stem cells, nerve-derived mesenchymal stem cells, skin It may be derived mesenchymal stem cells, amnion derived mesenchymal stem cells or placental derived mesenchymal stem cells.
본 발명의 창상의 예방 또는 치료용 약제학적 조성물에 따르면, 상기 창상은 좌상(contusion or bruise), 비-치유 외상성 창상, 방사선조사에 의한 조직의 파괴, 찰과상(abrasion), 골괴저, 열상(laceration), 결출상(avulsion), 관통상(penetrated wound), 총상(gunshot wound), 절상, 화상, 동상, 피부궤양, 피부 건조, 피부각화증, 갈라짐, 터짐, 피부염, 피부사상균증에 의한 통증, 수술상, 혈관질환 창상, 각막창상 등의 창상, 욕창, 와창, 당뇨성피부미란과 같은 당뇨병 및 순환불량에 관련된 상태, 만성궤양, 성형수술 후 봉합부위, 척추상해성 창상, 부인과적 창상, 화학적 창상 및 여드름을 포함할 수 있다. 바람직하기는, 상기 창상은 찰과상, 열상, 자상, 절상, 결출상, 관통상 또는 피부궤양을 포함할 수 있으나, 이에 한정되는 것은 아니다. According to the pharmaceutical composition for the prophylaxis or treatment of the wound of the present invention, the wound is contusion or bruise, non-healing traumatic wound, destruction of tissue by irradiation, abrasion, bone necrosis, laceration ), Avulsion, penetrated wounds, gunshot wounds, cuts, burns, frostbite, skin ulcers, dry skin, keratosis, cracking, rupture, dermatitis, pain caused by dermatophytosis, surgery, Vascular disease wounds, corneal wounds, bedsores, swellings, conditions related to diabetes and circulatory failure such as diabetic skin erosion, chronic ulcers, sutures after plastic surgery, spinal injury wounds, gynecological wounds, chemical wounds and acne It may include. Preferably, the wound may include, but is not limited to, abrasions, lacerations, cuts, cuts, crystallizations, penetrating wounds or skin ulcers.
3. 화장품 조성물3. Cosmetic composition
본 발명은 PGE2(prostaglandin E2)를 발현하는 세포에 15-PGDH(15-hydroxyprostaglandin dehydrogenase) 억제제를 첨가하여 배양한 세포 또는 그것의 배양물을 포함하는 화장품 조성물을 제공하고자 한다. The present invention is to provide a cosmetic composition comprising cells cultured by adding 15-PGDH (15-hydroxyprostaglandin dehydrogenase) inhibitor to cells expressing PGE2 (prostaglandin E2).
본 발명의 화장품 조성물에 따르면, 상기 화장품 조성물은 피부개선, 주름 개선, 발모, 탈모방지, 세치개선 등의 목적을 위해 사용될 수 있다. According to the cosmetic composition of the present invention, the cosmetic composition may be used for the purpose of skin improvement, wrinkle improvement, hair growth, hair loss prevention, rinse improvement.
본 발명의 화장품 조성물에 따르면, 상기 화장품 조성물은 용액, 로션, 오일에 물이 분산된 또는 물에 오일이 분산된 형태의 에멀젼의 형태 또는 젤, 립스틱, 에어로졸의 형태일 수 있다. According to the cosmetic composition of the present invention, the cosmetic composition may be in the form of gel, lipstick, aerosol or emulsion in the form of a solution, lotion, water dispersed in oil or oil dispersed in water.
본 발명의 화장품 조성물에 따르면, 상기 화장품 조성물은 오일, 지방, 유화제, 가습제, 습윤제, 연화제, 보존제, 계면활성제, 농후제, 향료, 색소, 염료 및 알콜, 폴리올, 전해질, 실리콘 유도체 등의 기타 첨가제 등의 통상적으로 이용되는 성분들을 조합하여 제조될 수 있다. According to the cosmetic composition of the present invention, the cosmetic composition is an oil, a fat, an emulsifier, a humidifier, a humectant, a softener, a preservative, a surfactant, a thickening agent, a perfume, a pigment, a dye and an alcohol, a polyol, an electrolyte, a silicone derivative, and the like. It can be prepared by combining commonly used components such as additives.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로서, 본 발명의 범위가 이들 실시예에 의해 제한되는 것으로 해석되지는 않는 것은 당업계에서 통상의 지식을 가진 자에게 있어서 자명할 것이다. Hereinafter, the present invention will be described in more detail with reference to Examples. These examples are only for illustrating the present invention, it will be apparent to those skilled in the art that the scope of the present invention is not to be construed as being limited by these examples.
<< 실시예Example >>
실시예Example 1. In 1. In vivovivo 유효성 시험 Validation test
지방 유래 중간엽 줄기세포(이하 지방줄기세포, ADSC)의 면역질환의 일종인 아토피 피부염에 대한 유효성 평가는 DNCB 아토피 피부염 발현 생쥐를 이용하여 실험을 진행하였다.To evaluate the effectiveness of atopic dermatitis, a type of immune disease of adipose-derived mesenchymal stem cells (hereinafter, adipose stem cells, ADSC), experiments were conducted using DNCB atopic dermatitis-expressing mice.
1-1. 아토피 피부염 질환 동물모델 확립1-1. Established animal model of atopic dermatitis disease
(1) Animals(1) Animals
실험동물은 5주령 된. BALB/c mice(초기체중 18~20 g) 수컷을 실험동물 사육실에서 1주간 적응시킨 뒤 실험에 사용하였다. 실험기간 중 물은 자유로이 섭취시켰으며, 사육실 온도(22±2℃)와 상대습도(60±5%)를 알맞게 유지하고, 명암 (06:00 AM~18:00 PM)은 12시간 주기로 조절하였다. Experimental animals were 5 weeks old. BALB / c mice (18–20 g initial weight) were used in the experiment after adapting the males in the laboratory for one week. During the experiment, water was freely ingested. The room temperature (22 ± 2 ℃) and relative humidity (60 ± 5%) were properly maintained, and the contrast (06:00 AM ~ 18: 00 PM) was adjusted every 12 hours. .
(2) DNCB 조제(2) DNCB preparation
1차, 2차 Sensitization에 사용될 DNCB는 acetone : olive oil 혼합용액 (4:1) 으로 1% (1차 sensitization)와 0.2%(2차 sensitization)의 농도로 용해한 후 사용하였다. DNCB to be used for primary and secondary sensitization was dissolved in acetone: olive oil mixed solution (4: 1) at concentrations of 1% (primary sensitization) and 0.2% (secondary sensitization).
(3) Sensitization procedure (3) Sensitization procedure
Negative control을 제외한 나머지 실험군들의 털을 제거한 후 mice의 등에 1% DNCB 150 μL를 지름 0.8cm 크기로 5일간 도포하였으며, 4일후 0.2% DNCB 용액 150 μL를 3일간 도포하여 2차 sensitization을 시켰다. After removing the hairs of the experimental groups except for negative control, 150 μL of 1% DNCB was applied to the back of the mice for 5 days with a diameter of 0.8 cm. After 4 days, 150 μL of 0.2% DNCB solution was applied for 3 days to perform secondary sensitization.
1-2. 줄기세포 배양1-2. Stem Cell Culture
지방줄기세포 배양은 공용IRB 승인을 받아 지방조직을 디에뜨병원(서울소재)에서 공급받아 지방에서 지방줄기세포의 분리는 다음과 같은 방법으로 진행하였다. 우선 Collagenase를 이용하여 조직을 분해하고 cell strainer로 single cell만을 분리하여 배양플라스크에 배양배지와 함께 배양하면서, 지속적으로 계대배양을 통하여 세포를 증식시킨다. 본 시험에 사용한 모든 세포는 5계대 세포를 사용하였다. 인체 골수유래 중간엽 줄기세포(이하 골수줄기세포, BMSC)와 인체 제대 유래 중간엽줄기세포(이하 제대줄기세포, UCSC)는 Promocell(독일)에서 구입하여 사용하였다.Adipose stem cell culture was approved by public IRB, and the adipose tissue was supplied from Diete Hospital (Seoul). The separation of adipose stem cells from fat was carried out as follows. First, collagenase is used to decompose tissues, and to isolate only a single cell with a cell strainer and incubate it with a culture medium in a culture flask while continuously growing cells through passage. All cells used in this test were 5 passage cells. Human bone marrow-derived mesenchymal stem cells (hereinafter referred to as bone marrow stem cells, BMSC) and human umbilical cord-derived mesenchymal stem cells (hereinafter called umbilical cord stem cells, UCSC) were purchased from Promocell (Germany).
1-3. 15-PGDH inhibitor 제조 및 처치1-3. Preparation and treatment of 15-PGDH inhibitor
15-PGDH inhibitor는 조선대학교 조훈 교수님 연구실에서 공급을 받아 최종 농도 5uM로 사용하였다. 또한 각 줄기세포에 15-PGDH inhibitor를 배양배지와 함께 4~12시간 처치 후(priming), 3회 생리식염수로 세척한 세포를 수거하여 시험에 사용하였고(15-PGDH inhibitor로 priming한 지방줄기세포; PADSC, 15-PGDH inhibitor로 priming한 제대줄기세포;PUCSC, 15-PGDH inhibitor로 priming한 골수줄기세포;PBMSC) 시험에 사용한 배양액은 각 줄기세포에 15-PGDH inhibitor를 배양배지와 함께 4~12시간 처치 후, 새로운 배양액으로 교체한 다음 24시간동안 배양 후 그 배양액을 사용하였다. The 15-PGDH inhibitor was supplied from Cho Hun's laboratory and used at a final concentration of 5 uM. In addition, each stem cell was treated with a culture medium with 15-PGDH inhibitor for 4 to 12 hours (priming), and the cells washed three times with physiological saline were collected and used for testing (fat stem cells priming with 15-PGDH inhibitor). ; Umbilical cord stem cells priming with PADSC, 15-PGDH inhibitor; Bone marrow stem cells priming with PUCSC, 15-PGDH inhibitor; PBMSC) The culture medium used to test 15-PGDH inhibitor in each stem cell with culture medium After the time treatment, the culture was replaced with fresh culture and then used for 24 hours.
1-4. 지방줄기세포 투여1-4. Adipose Stem Cell Administration
지방줄기세포의 주입은 마우스 꼬리에 정맥주사방식(Intravenous Injection)으로 주입하였고 세포투여 대조군인 fibroblast(섬유아세포) 투여군과 15-PGDH inhibitor 처치 지방줄기세포도 지방줄기세포와 동일하게 시험하였다. 또한 지방줄기세포의 투여일정(도 1)은 단회 투여로 시험일 12일째 투여하였고 투여 후 2주간 육안병변을 확인한다. The injection of adipose stem cells was injected into the tail of the mouse by intravenous injection. The fibroblast (fibroblast) administration group and 15-PGDH inhibitor treated adipose stem cells were tested in the same manner as the adipose stem cells. In addition, the schedule of administration of adipose stem cells (FIG. 1) was administered on the 12th day of the test day as a single dose and confirmed the gross lesion for 2 weeks after the administration.
1-5. 군 분리 1-5. Military separation
실험군은 다음과 같고 통계학적 유의성이 없게 군 분리를 진행하였다(표 1). 2차 sensitization 하루 후에 새롭게 군 분리를 진행한 후, 세포 투여 군들에게 (1X106cells/head)를 미정맥에 투여하고 Negative control군과 DNCB control군에 생리식염수를 동량 투여하였다.The experimental group was as follows and the group was separated without statistical significance (Table 1). After one day of secondary sensitization, the group was newly separated, and the cell group was administered (1X10 6 cells / head) to the vein and the same amount of physiological saline was administered to the Negative control group and the DNCB control group.
1-6. 육안병변 평가1-6. Visual lesion evaluation
육안변변 평가는 군 분리 시점부터 매일 한번 각각의 마우스에 대해 육안적인 구제 효과를 판정하였다. 질병의 정도를 각질생성 (dryness), 찰과(excoriation), 홍반(erythema), 부종(edema)의 점수를 증상에 따라 각각 0점에서 3점까지 (0, none; 1, mild; 2, moderate; 3, severe) 부여한 후 그 수치를 합산하여 평가하였다. 투여 후 매 3일마다 그 수치를 합산하여 각 군의 평균값으로 군간의 유의적 변화를 비교 분석하였다. The macroscopic evaluation assessed the gross relief effect for each mouse once daily from the time of group separation. Severity of disease from dryness, excoriation, erythema and edema, depending on symptoms, from 0 to 3 points (0, none; 1, mild; 2, moderate) 3, severe), and added and evaluated the values. Every three days after administration, the values were summed up and the mean change of each group was compared.
1-7. 부검1-7. Autopsy
지방줄기세포 투여 후 2주 동안 clinical sign 관찰 후 부검을 실시하였다. 채혈로 혈액을 수집하여 4시간 동안 냉장보관 후 원심분리하여 혈청을 얻었으며 mouse dorsal skin은 NO/MDA level 및 분자생물학 분석시험을 측정하기 위해 냉동보관(-80℃) 하였다. An autopsy was performed after clinical sign observation for 2 weeks after the administration of adipose stem cells. Blood was collected by blood collection and refrigerated for 4 hours, and then serum was obtained by centrifugation. Mouse dorsal skin was stored frozen (-80 ° C) to measure NO / MDA levels and molecular biology assays.
1-8. 피부 조직 검사1-8. Skin biopsy
투여 2주후 부검시 실험동물을 안락사 시키고, 피부염 병변 조직 부위를 확보하고 4% paraformaldehyde에 조직을 고정시킨 후 파라핀으로 포매 후 H&E 염색 및 Toluidine blue염색을 실시한 후 H&E 염색된 피부조직을 현미경관찰을 통하여 상피층 두께의 변화를 비교 분석하고 Toluidine blue염색을 통해 비만세포 탈과립 정도를 확인하였다. Two weeks after administration, the animals were euthanized, the dermatitis lesion tissues were secured, the tissues were fixed in 4% paraformaldehyde, embedded with paraffin, H & E stained and toluidine blue stained, and H & E stained skin tissues were examined by microscopic observation. Changes in epithelial thickness were analyzed and the degree of mast cell degranulation was confirmed by toluidine blue staining.
1-9. 혈청 내 IgE, PGE2, TGFb1 농도 1-9. Serum IgE, PGE2, TGFb1 Concentrations
혈청 IgE, PGE2 및 TGFb1 level은 각각 mouse ELISA kit를 사용하여 Microplate reader에서 측정하였다. Serum IgE, PGE2 and TGFb1 levels were measured in a microplate reader using a mouse ELISA kit, respectively.
1-10. 손상 피부 및 플라즈마 내의 NO / MDA 수준1-10. NO / MDA levels in damaged skin and plasma
면역학적인 관점에서 NO 합성효소의 발현은 NO의 생성을 증가시키며 병리학적인 조직손상을 나타내는 부위에는 NO의 함량이 증가되는 것으로 알려져 있다. 또한 진피세포 내의 NO의 증가는 지질과산화를 유발하는 ROS의 증가를 촉진시킨다. NO는 알레르기 및 천식과 같은 염증질환에 있어서 다량이 발현되어 염증을 유발시키는 인자이며 NO가 발현하는 기능의 다양성은 농도 및 표적세포의 활성화 여부 등에 따라 달라진다고 알려져 있다. NO(nitric oxide) 함량은 안정된 NO 산화물인 NO2-(nitrite)를 Griess 반응을 이용하여 측정하였다. EDTA가 들어있는 50 mM potassium phosphate buffer로 조직을 마쇄한 후에 4℃, 10,000×g에서 15분 동안 원심분리하여 조직의 상층액과 혈청 100 μL 각각을 96 well plate에 넣고, 여기에 Griess 시약 (0.1% N-1-naphthyl-ethylendiamine / H2O : 1% sulfanilamide / 5% H3PO4 = 1 : 1)을 동량 첨가하여 10분간 반응시킨 후, microplate reader로 570 nm에서 흡광도를 측정하였다. Nitrite의 농도는 sodium nitrite를 이용하여 얻은 표준곡선과 비교하여 표현하였다. From the immunological point of view, the expression of NO synthase increases NO production and is known to increase NO content at sites showing pathological tissue damage. Increasing NO in dermal cells also promotes an increase in ROS that induces lipid peroxidation. NO is a factor that causes inflammation by expressing a large amount in inflammatory diseases such as allergy and asthma, and the variety of functions expressed by NO is known to vary depending on the concentration and activation of target cells. The NO (nitric oxide) content was measured using Griess reaction of NO2- (nitrite), a stable NO oxide. After grinding the tissue with 50 mM potassium phosphate buffer containing EDTA, centrifuge for 15 minutes at 10,000 × g at 4 ° C, and
Allergen의 투여로 인하여 NO 발생을 유도하게 되면 NO와 주변의 활성산소에 의하여 세포의 지질과산화를 유도하여 MDA와 같은 산화 반응물질을 생산한다. MDA는 산화 반응에 의한 다가불포화지방산의 산화물로서 조직세포의 산화적인 손상(염증)의 증거로 알려져 있다. MDA(malondialdehyde)의 함량은 thiobarbituric acid (TBA)를 사용하여 측정하였다. 즉. 증류수 1 mL와 acetic acid에 녹인 29 mM TBA 1 mL에 혈청과 조직 시료 50 μL 첨가하여 95℃에서 1시간 반응한 후에 식힌 다음 5mM HCl을 25 μL 첨가하였다. 여기에 3.5 mL의 n-butanol을 첨가하여 추출하고 butanol 층은 1,500×g에서 5분동안 원심분리 하여 모은 후에 532 nm에서 흡광도를 측정하였다. 이때 표준곡선은 1,1,3,3- tetraethoxypropane을 이용하여 작성하였으며, MDA의 농도는 μM/g, wet tissue로 환산하여 나타내었다.Induction of NO by the administration of Allergen induces lipid peroxidation of cells by NO and surrounding free radicals to produce oxidation reactants such as MDA. MDA is known as evidence of oxidative damage (inflammation) of tissue cells as an oxide of polyunsaturated fatty acids by oxidation. The content of malondialdehyde (MDA) was measured using thiobarbituric acid (TBA). In other words. 50 μL of serum and tissue samples were added to 1 mL of distilled water and 1 mL of 29 mM TBA dissolved in acetic acid. The mixture was cooled at 95 ° C. for 1 hour, cooled, and then 25 μL of 5 mM HCl was added thereto. 3.5 mL of n-butanol was added thereto, and the butanol layer was collected by centrifugation at 1,500 × g for 5 minutes and absorbance was measured at 532 nm. The standard curve was prepared using 1,1,3,3-tetraethoxypropane and the concentration of MDA was expressed in μM / g, wet tissue.
1-11. 자료의 통계처리1-11. Statistical processing of data
실험에서 나온 자료의 통계학적 분석을 위하여 one-way ANOVA를 실시하여 p=0.05 수준에서 군간 유의성을 검정하고 유의성이 인정되면, Dunnett's t-test를 실행하여 대조군과 시험군 간의 통계학적 유의성을 검정했다 (p<0.05).For statistical analysis of the data from the experiment, one-way ANOVA was performed to test the significance between the groups at p = 0.05, and if the significance was recognized, Dunnett's t-test was used to test the statistical significance between the control group and the test group. (p <0.05).
실시예Example 2. In vitro 유효성 시험 2. In vitro validation test
2-1. 중간엽 줄기세포의 활성화 T세포 억제 기능2-1. Inhibitory Function of Activated T Cells in Mesenchymal Stem Cells
인간말초혈액에서 분리한 T세포를 여러 mitogen(anti-CD3, PHA, IL-2)를 처리하여 활성화 시킨후, ADSC, UCSC, BMSC, PADSC, PUCSC, PBMSC와 각각 5일간 공배양(co-culture)하고 배양 종료시 ADAM cell counter를 이용하여 T세포의 증식률을 측정하였다. Co-cuture시 각 중간엽줄기세포 농도를 T세포 대비 1×104(10:1), 5×104(5:1), 1×105(1:1)으로 하여 중간엽줄기세포 농도 의존적 억제가 일어나는지 관찰하며, 각 중간엽 줄기세포들이 cell-contact에 의해 T세포를 억제하는 것인지 아닌지를 확인하기 위하여 transwell plate를 이용하여 T세포와 중간엽줄기세포의 cell contact를 방지한 후 co-culture도 실시하였다. Cell counter로 계수한 세포수는 co-culture를 실시하지 않은 활성화 T세포를 기준(세포수 100%)으로 각 세포수를 %로 환산하여 도표화하였다. T cells isolated from human peripheral blood were activated by treatment with several mitogens (anti-CD3, PHA, IL-2) and co-cultured with ADSC, UCSC, BMSC, PADSC, PUCSC, and PBMSC for 5 days each. ) And the proliferation rate of T cells was measured using the ADAM cell counter at the end of the culture. Mesenchymal stem cell concentration in co-cuture was 1 × 10 4 (10: 1), 5 × 10 4 (5: 1), 1 × 10 5 (1: 1) compared to T cells. In order to observe whether the dependent inhibition occurs and to determine whether each mesenchymal stem cell inhibits T cells by cell-contact or not, use a transwell plate to prevent cell contact between T cells and mesenchymal stem cells. Culture was also conducted. The number of cells counted by the cell counter was tabulated by converting each cell number to% based on the activated T cells without co-culture (100% cell number).
2-2. 줄기세포와 공배양된 활성화 T세포의 면역표지 발현 분석2-2. Immunomarker Expression Analysis of Activated T Cells Co-Cultured with Stem Cells
중간엽줄기세포, 15-PGDH inhibitor를 처리한 중간엽줄기세포 각각 인간말초혈액에서 분리한 T세포와 여러 mitogen을 처리한 배양액과 같이 5일간 Transwell에서 공배양하고 배양 종료시 T세포만을 수거하여 유세포분석기(FACS)를 이용해 T세포의 표면항체를 분석하였다.Mesenchymal stem cells and mesenchymal stem cells treated with 15-PGDH inhibitor, respectively, were co-cultured in Transwell for 5 days, such as T cells isolated from human peripheral blood and culture medium treated with various mitogens. Surface antibodies of T cells were analyzed using (FACS).
1-3. 피부섬유아세포와 상피각질세포의 증식능 시험1-3. Proliferative test of dermal fibroblasts and epidermal keratinocytes
지방줄기세포, 제대줄기세포, 골수줄기세포에 15-PGDH inhibitor를 4~12시간 처치하거나 처치하지 않은 후, 15-PGDH inhibitor가 첨가되지 않은 새로운 배양액으로 교체한 다음 24시간 동안 배양 후 그 배양액을 수거하였다. 시험 1일전 피부섬유아세포(HDF)와 상피각질세포(HEK)를 6well에 각 20000개의 세포를 접종하고 시험일에 상기에서 수거한 배양약으로 교체해 주었다. 그런 다음 5% CO2 incubator에 배양하고 3일후에 cell counter기로 세포수를 계수하여 생존 세포수를 확인하였다. 모든 시험은 Triplicate 하여 표준편차 값을 계산하였다. Adipose stem cells, umbilical cord stem cells, and bone marrow stem cells were treated with or without 15-PGDH inhibitor for 4 to 12 hours, and then replaced with fresh culture medium without 15-PGDH inhibitor added and cultured for 24 hours. Collected. Dermal fibroblasts (HDF) and epidermal keratinocytes (HEK) were inoculated with 20,000 cells in 6 wells one day before the test and replaced with the cultures collected above on the test day. Then, the cells were incubated in a 5% CO2 incubator, and after 3 days, cell numbers were counted with a cell counter to confirm viable cell numbers. All tests were triplicated to calculate standard deviation values.
<< 실험예Experimental Example >>
실험예Experimental Example 1. In 1. In vivovivo 유효성 시험 Validation test
1-1. 육안병변 평가 결과1-1. Visual lesion evaluation result
DNCB로 유발된 allergic dermatitis(AD)의 마우스 모델에서 지방줄기세포의 항염증 효과를 관찰하였다(도 2A). MSC 투여 후 매 3일마다 각질생성 (dryness), 찰과 (excoriation), 홍반 (erythema), 부종 (edema)등을 관찰하고 그 중증도를 0~3 점까지 부여하여 그룹간의 유의성을 비교하였다(도 2B).Anti-inflammatory effects of adipose stem cells were observed in a mouse model of DNCB-induced allergic dermatitis (AD) (FIG. 2A). Every 3 days after MSC administration, dryness, abrasion, erythema, and edema were observed and the severity was assigned to 0-3 points to compare the significance between the groups (Fig. 2B).
그 결과, ADSC 및 PADSC를 투여한 경우 10일 후(Day 21)부터 DNCB군에 비해 질환의 중증도가 감소한다는 것이 확인되었다. As a result, it was confirmed that the administration of ADSC and PADSC reduced the severity of the disease compared to the DNCB group from 10 days later (Day 21).
1-2. 피부조직검사 결과1-2. Skin biopsy result
지방줄기세포에 의해 항염증 효과가 나타난 마우스 등에서 조직 구조의 변화와 비만세포 침윤 및 탈과립 현상을 관찰하였다. 실험 종료 후 마우스 등 조직을 적출하여 파라핀 블록을 제작한 후 6 μm 두께로 조직을 절편하여 H&E 염색과 Toluidin blue 염색을 수행함. 각각이 조직들에 대한 H&E 염색을 수행하였다(도 3).Changes in tissue structure, mast cell infiltration and degranulation were observed in mice with anti-inflammatory effects caused by adipose stem cells. After the experiment, tissues such as mice were extracted to prepare paraffin blocks, and the tissues were sliced to a thickness of 6 μm to perform H & E staining and toluidin blue staining. Each performed H & E staining on tissues (FIG. 3).
그 결과, 정상 마우스(N.C. negative control)에 비해 DNCB를 처리한 군 (대조군)에서 눈에 띄게 표피와 진피층의 swelling이 관찰되었으며, Fibroblast를 투여한 군에 비해 ADSC 및 PADSC를 투여한 경우 표피와 진피층의 swelling이 대조군에 비해 상당히 줄어든다는 것이 확인되었다(도 3A). 이로써 DNCB에 의해 유도되는 급성염증반응이 ADSC 및 PADSC에 의해 감소될 수 있음이 입증되었다. As a result, swelling of the epidermis and dermis was noticeably observed in the DNCB-treated group (control group) compared to the normal mouse (NC negative control), and the epidermal and dermal layers were treated with ADSC and PADSC compared with the fibroblast group. It was confirmed that the swelling of was significantly reduced compared to the control (FIG. 3A). This demonstrated that the acute inflammatory response induced by DNCB can be reduced by ADSC and PADSC.
또한, Toluidin blue 염색 후 비만세포 침윤 및 탈과립을 관찰한 결과 ADSC 및 PADSC 투여군에서는 대조군에 비해 매우 적은 수의 비만세포 침윤 및 탈과립이 나타남이 확인되었다(도 3B).In addition, as a result of observing mast cell infiltration and degranulation after Toluidin blue staining, it was confirmed that very few mast cell infiltration and degranulation were observed in the ADSC and PADSC-administered groups (FIG. 3B).
1-3. IgE 및 주요 염증성 cytokine 분석 결과1-3. IgE and Major Inflammatory Cytokine Assays
염증반응 지표 인자 및 염증성 cytokine의 변화를 관찰하기 위해 IgE, PGE2 및 TGF-β1 수준을 Elisa kit을 이용하여 수행하였다(도 4). IgE, PGE2 and TGF-β1 levels were performed using an Elisa kit to observe changes in inflammatory markers and inflammatory cytokine (FIG. 4).
그 결과, 대조군에 비해 ADSC 및 PADSC 투여군의 경우 혈청 내 IgE level이 낮은 상태로 유지되고 있음이 확인되었다. As a result, it was confirmed that serum IgE levels were maintained in the ADSC and PADSC-administered groups as compared to the control group.
1-4. NO 및 MDA 분석 결과1-4. NO and MDA analysis results
알러지 및 천식과 같은 염증성 질환의 경우 nitric oxide(NO) 발생하며 이는 주변의 활성산소에 의한 지질과산화를 유도하여 malondialdehyde(MDA) 산화물이 된다. 따라서 혈청 및 병변 조직에서 NO 및 MDA의 함량을 측정하였다(도 5) Inflammatory diseases such as allergies and asthma develop nitric oxide (NO), which induces lipid peroxidation by the surrounding free radicals, resulting in malondialdehyde (MDA) oxide. Therefore, the content of NO and MDA in serum and lesion tissue was measured (Fig. 5).
그 결과, ADSC 및 PADSC 투여군의 경우 대조군에 비해 혈청 내 낮은 NO level을 보였으며 조직의 경우 ADSC가 PADSC에 비해 NO 생성 억제효과가 더욱 크게 나타났다(도 5A). 혈청 내 MDA level은 PADSC 투여군에서 정상 마우스와 비슷한 수준으로 나타났으나 ADSC군에서는 음성대조군(N.C)과 별다른 차이를 나타내지 않았다. 조직의 경우 유의적 변화를 보이지 않는 것으로 확인되었다(도 5B). As a result, the ADSC and PADSC-administered groups showed lower NO levels in the serum than the control group, and the tissues showed a greater NO inhibitory effect than the PADSC (FIG. 5A). Serum MDA levels were similar to those of normal mice in the PADSC-administered group, but not significantly different from the negative control group (N.C) in the ADSC group. In the case of tissue, no significant change was found (FIG. 5B).
실험예Experimental Example 2. In vitro 유효성 시험 2. In vitro validation test
2-1. 중간엽 줄기세포의 활성화 T세포 억제 기능 분석 결과2-1. Analysis of Activated T Cell Inhibitory Function in Mesenchymal Stem Cells
인간말초혈액에서 분리한 T세포를 mitogen으로 활성화 시킨 후, 중간엽 줄기세포와 15-PGDH를 처리한 중간엽 줄기세포와 공배양 후, 활성화된 T세포의 수를 측정하였다(도 6). After activating T cells isolated from human peripheral blood with mitogen, after co-culture with mesenchymal stem cells and 15-PGDH treated mesenchymal stem cells, the number of activated T cells was measured (FIG. 6).
그 결과, 공배양시 중간엽줄기세포와 활성화 T세포를 직접 붙여 공배양(Contact co-culture)한 경우, T세포와 중간엽줄기세포의 비율이 1:1인 환경에서 유의적으로 중간엽줄기세포들이 활성화 T세포의 증식을 억제하였고 지방줄기세포, 제대줄기세포의 경우 15-PGDH를 처리한 중간엽 줄기세포가 활성화된 T세포의 증식을 더 많이 억제함이 확인되었다(도 6A). 또한, 공배양시 중간엽줄기세포와 활성화 T세포를 직접 닿지 않게 insert로 분리시켜 공배양(Transwell co-culture)한 경우, T세포와 중간엽줄기세포의 비율이 1:1인 환경에서 유의적으로 중간엽줄기세포들이 활성화 T세포의 증식을 억제하였고 15-PGDH를 처리한 모든 중간엽줄기세포가 활성화된 T세포의 증식을 더 많이 억제한다는 것이 확인되었다(도 6B).As a result, in the case of co-culture by directly attaching mesenchymal stem cells and activated T cells in co-culture, mesenchymal stems were significantly increased in an environment where the ratio of T cells and mesenchymal stem cells was 1: 1. The cells suppressed the proliferation of activated T cells, and in the case of adipose stem cells and umbilical cord stem cells, mesenchymal stem cells treated with 15-PGDH were found to inhibit the proliferation of activated T cells more (FIG. 6A). In addition, when co-culture was carried out by inserting the mesenchymal stem cells and the activated T cells so as not to directly touch the cells, the ratio of T cells and mesenchymal stem cells was significant in a 1: 1 environment. As a result, it was confirmed that mesenchymal stem cells inhibited the proliferation of activated T cells and all mesenchymal stem cells treated with 15-PGDH inhibited the proliferation of activated T cells more (FIG. 6B).
2-2. 활성화 T세포 면역표지 발현 분석 결과2-2. Activation T Cell Immunomarker Expression Assay
인간말초혈액에서 분리한 T세포를 mitogen으로 활성화 시킨 후, 중간엽 줄기세포와 15-PGDH inhibitor를 처리한 중간엽 줄기세포와 각각 공배양된 T세포를 수거하여 유세포분석을 실시하였다. 이 때, regulatory T세포의 표현형인 CD4, CD25, FoxP3의 항원에 대하여 분석하였다(도 7).After activating T cells isolated from human peripheral blood with mitogen, flow cytometry was performed by collecting mesenchymal stem cells and mesenchymal stem cells treated with 15-PGDH inhibitor and cocultured T cells, respectively. At this time, the antigens of CD4, CD25 and FoxP3 which are phenotypes of regulatory T cells were analyzed (FIG. 7).
그 결과, 15-PGDH inhibitor를 처리한 제대줄기세포(PUCSC)와 공배양된 T세포는 regulatory T세포가 15-PGDH inhibitor를 처리하지 않은 제대줄기세포(UCSC)(44%)와 공배양된 T세포에 비해 48%정도 증가하여 92%의 Treg population을 나타내었으며 골수줄기세포(BMSC)와 공배양된 T세포는 39%의 Treg population을 보인 반면, 15-PGDH inhibitor를 처리한 골수줄기세포(PBMSC)와 공배양된 T세포는 98%까지 Treg population이 증가하였다. 이로써, 중간엽줄기세포들은 활성화된 T세포는 억제를 하지만 면역조절을 할 수 있는 regulatory T세포는 증가시키고 15-PGDH inhibitor는 이러한 면역조절을 하는 줄기세포의 능력을 훨씬 강화시킬 수 있음이 입증되었다. As a result, T cells co-cultured with umbilical stem cells treated with 15-PGDH inhibitor (PUCSC) and T cells co-cultured with umbilical stem cells (UCSC) untreated with 15-PGDH inhibitor (44%) The Treg population of 92% was increased by 48% compared to the cells, and the T cells co-cultured with BMSC showed 39% of Treg population, while bone marrow stem cells treated with 15-PGDH inhibitor (PBMSC). ) And T cells co-cultured increased the Treg population by 98%. This demonstrates that mesenchymal stem cells inhibit activated T cells but increase regulatory T cells that are capable of immunomodulation, and that 15-PGDH inhibitors can significantly enhance the ability of stem cells to regulate this immune regulation. .
2-3. 피부섬유아세포와 상피각질세포의 증식능 시험 결과2-3. Proliferative test results of dermal fibroblasts and epidermal keratinocytes
ADSC, UCSC, BMSC와 15-PGDH inhibitor를 priming한 PADSC, PUCSC, PBMSC는 24시간 동안 배양한 후 각각 배양액을 수거하여 인체피부섬유아세포(HDF), 인체상피각질세포(HEK)의 성장률을 비교하였다(도 8). PADSC, PUCSC, and PBMSC priming ADSC, UCSC, BMSC, and 15-PGDH inhibitors were cultured for 24 hours, and then the cultures were harvested to compare the growth rate of human skin fibroblasts (HDF) and human epidermal keratinocytes (HEK). (FIG. 8).
그 결과, HDF의 경우 15-PGDH inhibitor를 priming한 PADSC, PUCSC 배양액으로 배양한 경우, 배양액 대조군에 비해 유의적으로 성장률이 증가하였다. HEK의 경우도 마찬가지로 15-PGDH inhibitor를 priming한 PADSC, PUCSC 배양액으로 배양한 경우 배양액 대조군에 비해 증식률이 증가한다는 것이 확인되었다. As a result, the growth rate of HDF was significantly increased when cultured with PADSC and PUCSC cultures priming 15-PGDH inhibitor. In the case of HEK, it was confirmed that the growth rate was increased when the 15-PGDH inhibitor was incubated with PADSC and PUCSC cultures.
본 발명이 속하는 기술 분야에서 통상의 지식을 가진 자는 본 발명이 본 발명의 본질적인 특성에서 벗어나지 않는 범위에서 변형된 형태로 구현될 수 있음을 이해할 수 있을 것이다. 그러므로 개시된 실시예들은 한정적인 관점이 아니라 설명적인 관점에서 고려되어야 한다. 본 발명의 범위는 전술한 설명이 아니라 특허청구범위에 나타나 있으며, 그와 동등한 범위 내에 있는 모든 차이점은 본 발명에 포함된 것으로 해석되어야 할 것이다.Those skilled in the art will appreciate that the present invention can be implemented in a modified form without departing from the essential features of the present invention. Therefore, the disclosed embodiments should be considered in descriptive sense only and not for purposes of limitation. The scope of the present invention is shown in the claims rather than the foregoing description, and all differences within the scope will be construed as being included in the present invention.
본 발명에 따른 약제학적 조성물은 DNCB로 유발된 AD 마우스 모델에서 항염증 효과, 혈청 내 IgE 감소 효과, NO 및 MDA 감소 효과, 활성화된 T 세포 증식 억제 효과, 및 피부섬유아세포 및 상피각직세포 증식능 향상 효과를 가질 수 있다. 따라서, 본 발명에 따른 약제학적 조성물은 항역질환치료제, 염증질환치료제 또는 창상치료제로서 유효하게 사용될 수 있을 것으로 기대된다. The pharmaceutical composition according to the present invention has anti-inflammatory effect, serum IgE reduction effect, NO and MDA reduction effect, activated T cell proliferation inhibitory effect, and dermal fibroblast and epithelial keratinocyte proliferation ability in DNCB-induced AD mouse model Can have an effect. Therefore, the pharmaceutical composition according to the present invention is expected to be effectively used as an anti-inflammatory disease treatment agent, an inflammatory disease treatment agent or a wound treatment agent.
Claims (24)
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US15/771,853 US20190030076A1 (en) | 2015-10-27 | 2016-10-27 | Composition comprising cells treated with 15-pgdh inhibitor or culture thereof and use thereof |
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR20150149142 | 2015-10-27 | ||
| KR10-2015-0149142 | 2015-10-27 | ||
| KR1020160139967A KR101843927B1 (en) | 2015-10-27 | 2016-10-26 | Composition comprising cells treated with 15-PGDH inhibitor or culture thereof and use thereof |
| KR10-2016-0139967 | 2016-10-26 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2017074043A1 true WO2017074043A1 (en) | 2017-05-04 |
Family
ID=58630664
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2016/012125 Ceased WO2017074043A1 (en) | 2015-10-27 | 2016-10-27 | Composition containing cells treated with 15-pgdh inhibitor or culture thereof and use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2017074043A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2010077101A2 (en) * | 2008-12-30 | 2010-07-08 | 조선대학교산학협력단 | Novel thiazolidinedione derivative and use thereof |
| KR20120085209A (en) * | 2011-01-21 | 2012-07-31 | 인제대학교 산학협력단 | Composition for inhibiting immune response comprising stem cell expressing tryptophan metabolic enzyme gene |
| KR20140010576A (en) * | 2012-07-13 | 2014-01-27 | 충북대학교 산학협력단 | Human neural stem cell expressing therapeutic gene and pharmaceutical composition comprising the same for treating metastatic cancer |
-
2016
- 2016-10-27 WO PCT/KR2016/012125 patent/WO2017074043A1/en not_active Ceased
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2010077101A2 (en) * | 2008-12-30 | 2010-07-08 | 조선대학교산학협력단 | Novel thiazolidinedione derivative and use thereof |
| KR20120085209A (en) * | 2011-01-21 | 2012-07-31 | 인제대학교 산학협력단 | Composition for inhibiting immune response comprising stem cell expressing tryptophan metabolic enzyme gene |
| KR20140010576A (en) * | 2012-07-13 | 2014-01-27 | 충북대학교 산학협력단 | Human neural stem cell expressing therapeutic gene and pharmaceutical composition comprising the same for treating metastatic cancer |
Non-Patent Citations (2)
| Title |
|---|
| SHEIBANE, A. F. ET AL.: "Prostaglandin E2 Exacerbates Collagen-Induced Arthritis in Mice through the Inflammatory Interleukin-23/interleukin-17 Axis", ARTHRITIS & RHEUMATISM, vol. 56, no. 8, 2007, pages 2608 - 2619, XP055382251 * |
| ZHANG, Y. ET AL.: "Inhibition of the Prostaglandin-Degrading Enzyme 15-PGDH Potentiates Tissue Regeneration", SCIENCE, vol. 348, no. 6240, June 2015 (2015-06-01), pages 1223, aaa2340, XP055348197 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2019135644A1 (en) | Composition for improving, preventing or treating skin diseases comprising induced pluripotent stem cell-derived mesenchymal stem cell and exosome derived therefrom | |
| WO2020106099A1 (en) | Exosome and various uses thereof | |
| WO2019135645A1 (en) | Composition for improving, preventing or treating skin disease comprising induced pluripotency stem cell-derived mesenchymal stem cells pretreated with interferon gamma and exosomes derived therefrom | |
| KR101512171B1 (en) | A composition comprising stem cell for preventing or treating of immune or inflammatory disease | |
| WO2012026712A2 (en) | Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing stem cells treated with nod2 agonist or cultured product thereof | |
| WO2016048107A1 (en) | Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, comprising interferon-gamma or interleukin-1 beta treated stem cell or culture thereof | |
| WO2019004738A2 (en) | Use of composition comprising adipose stem cell-derived exosome as effective ingredient in alleviating dermatitis | |
| WO2022119417A1 (en) | Method for preparing high-concentration stem cell exosomes with enhanced anti-inflammatory and regenerative functions using lipopolysaccharide and lipoteichoic acid | |
| WO2020218781A1 (en) | Functional composition containing immortalized stem cell-derived exosome-rich culture medium and rosebud extract as active ingredients | |
| WO2019198995A1 (en) | Exosome-based conversion method for immune cells | |
| WO2015026215A9 (en) | Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing biguanide derivative compound as active ingredient | |
| Guttinger et al. | Allogeneic mesoangioblasts give rise to alpha-sarcoglycan expressing fibers when transplanted into dystrophic mice | |
| WO2015167243A1 (en) | Novel compound having immune disease treatment effect and use thereof | |
| WO2018056676A1 (en) | Pharmaceutical composition comprising purple corn extract for prevention or treatment of skin disease | |
| WO2012008733A2 (en) | Stem cells derived from primary placenta tissue and cellular therapeutic agent containing same | |
| WO2022119418A1 (en) | Method for isolating and culturing cord blood stem cells expressing gdf-3 at high level, and use of gdf-3 | |
| WO2022255759A1 (en) | Combined administration composition for preventing or treating atopic dermatitis, comprising function-enhanced stem cells and regulatory t cells | |
| WO2015023147A1 (en) | Mtor/stat3 signal inhibitor-treated mesenchymal stem cell having immunomodulatory activity, and cell therapy composition comprising same, for preventing or treating immune disorders | |
| WO2017074043A1 (en) | Composition containing cells treated with 15-pgdh inhibitor or culture thereof and use thereof | |
| KR101843927B1 (en) | Composition comprising cells treated with 15-PGDH inhibitor or culture thereof and use thereof | |
| WO2020071665A2 (en) | Composition for extending telomere of cell and preparation method therefor | |
| KR101912142B1 (en) | Composition comprising cells treated with 15-PGDH inhibitor or culture thereof and use thereof | |
| WO2015023165A1 (en) | Inflammation-controlling composite and stabilized mesenchymal stem cells having optimized immunity control function by blocking stat3 signal molecule | |
| WO2022108165A1 (en) | Method for producing exosomes isolated from induced pluripotent stem cell-derived mesenchymal stem cells, and use thereof | |
| WO2015194710A1 (en) | Composition comprising mesenchymal stem cells treated with stat3 inhibitor as active ingredient for preventing or treating immune diseases |
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: 16860226 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 32PN | Ep: public notification in the ep bulletin as address of the adressee cannot be established |
Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205N DATED 03.07.2018) |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 16860226 Country of ref document: EP Kind code of ref document: A1 |