KR20150016117A - Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow - Google Patents

Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow Download PDF

Info

Publication number
KR20150016117A
KR20150016117A KR1020140097129A KR20140097129A KR20150016117A KR 20150016117 A KR20150016117 A KR 20150016117A KR 1020140097129 A KR1020140097129 A KR 1020140097129A KR 20140097129 A KR20140097129 A KR 20140097129A KR 20150016117 A KR20150016117 A KR 20150016117A
Authority
KR
South Korea
Prior art keywords
cells
mesenchymal stem
bone marrow
stem cells
derived mesenchymal
Prior art date
Application number
KR1020140097129A
Other languages
Korean (ko)
Inventor
김경숙
배상철
Original Assignee
코아스템(주)
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 코아스템(주) filed Critical 코아스템(주)
Priority to KR1020140097129A priority Critical patent/KR20150016117A/en
Priority to PCT/KR2014/011055 priority patent/WO2016017863A1/en
Publication of KR20150016117A publication Critical patent/KR20150016117A/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells

Abstract

In the present invention, provided is a pharmaceutical composition for treating or preventing autoimmune diseases, including mesenchymal stem cells derived from bone marrow as an active ingredient, wherein the mesenchymal stem cells derived from bone marrow plays roles in inhibiting generation of anti-dsDNA antibodies, proteinuria and total IgG without any toxicity, the proliferation of T cells and B cells and expression of cytokines in T cells, thereby having effects of preventing or treating autoimmune diseases. In the present invention, systemic lupus erythematosus is inflammation in the kidney by autoantibody and immune complex and immunocyte infiltration is increased in systemic lupus erythematosus. When mesenchymal stem cells derived from bone marrow of the present invention are administered, systemic lupus erythematosus is prevented or treated by reducing immunocyte infiltration.

Description

FIELD OF THE INVENTION [0001] The present invention relates to a pharmaceutical composition for preventing or treating autoimmune diseases, which comprises human bone marrow-derived mesenchymal stem cells as an active ingredient.

The present invention relates to a pharmaceutical composition for preventing or treating an autoimmune disease comprising human bone marrow-derived mesenchymal stem cells as an active ingredient.

Autoimmune disease is a disease caused by the immune function of our body attacking itself and it is a common example that it is formed over a long period of time and the symptoms persist chronically and usually lead to permanent damage of organs. There is no reality. In the past 20 to 30 years, knowledge about autoimmune diseases has developed a lot, but the precise generation mechanism, autoantigen stagnation, and regulatory genetic factors are still unclear. Autoimmune diseases can be broadly divided into organ-specific diseases and systemic diseases.

The organ specific autoimmune disease is caused by the immune response to the organ specific antigen and can occur in almost all organs of our body. Systemic autoimmune disease is not caused by an immune response to any particular cell but by an immune response to an antigen expressed throughout the body. These systemic autoimmune diseases can also selectively cause diseases in specific organs.

Examples of autoimmune diseases include: (i) Rheumatoid Arthritis, in which the immune system attacks the tissues of various joints, (ii) autoimmunity of the central nervous system induced by T cells, Multiple sclerosis (MS), which can lead to blindness, premature death, and iii) Insulin-producing cells of the pancreas are produced by the destruction of immune cells and MHC genes are important immune mediators or Type I diabetes (Immune- Inflammatory Bowel Diseases, a disease in which the immune system attacks the intestines, v) Scleroderma, which induces thickening of the skin or blood vessels, and vi) Systemic autoimmunity leads to deep fatigue, rash, arthralgia, etc. In severe cases, the immune system may cause systemic inflammation of the kidneys, brain, And the like sex lupus (Systemic Lupus Erythematosus, SLE).

Numerous papers and patent documents are referenced and cited throughout this specification. The disclosures of the cited papers and patent documents are incorporated herein by reference in their entirety to better understand the state of the art to which the present invention pertains and the content of the present invention.

The present inventors have intensively studied to develop a stem cell capable of effectively treating an autoimmune disease and safely applicable to a human body, and as a result, it has been found that mesenchymal stem cells obtained from human bone marrow are very effective for preventing or treating autoimmune diseases The present invention has been completed.

Accordingly, it is an object of the present invention to provide a pharmaceutical composition for the prevention or treatment of autoimmune diseases.

Other objects and advantages of the present invention will become more apparent from the following detailed description of the invention and claims.

According to one aspect of the present invention, there is provided a pharmaceutical composition for preventing or treating an autoimmune disease comprising human bone marrow-derived mesenchymal stem cells as an active ingredient, wherein said human bone marrow-derived mesenchymal stem cell comprises (i) CD105, CD29 , ≪ / RTI > CD44, CD73 and CD90; And (ii) at least one surface antigen selected from the group consisting of CD34, CD45 and HLA-DR.

The present inventors have intensively studied to develop a stem cell capable of effectively treating an autoimmune disease and safely applicable to a human body, and as a result, it has been found that mesenchymal stem cells obtained from human bone marrow are very effective for preventing or treating autoimmune diseases .

The human bone marrow-derived mesenchymal stem cells of the present invention comprise at least one, preferably at least two, more preferably at least three, selected from the group consisting of CD105, CD29, CD44, CD73 and CD90, , Even more preferably at least four, and most preferably five, surface antigens. According to an embodiment of the present invention, the human bone marrow-derived mesenchymal stem cells express 70-100% of the stem cell surface surface antigens CD105, CD29, CD44, CD73 and CD90, more preferably 80-100 %, And even more preferably 90-99%.

Meanwhile, the human bone marrow-derived mesenchymal stem cells of the present invention exhibit the immunological characteristics of negative for CD34, CD45 and HLA-DR, which are hematopoietic stem cell markers surface antigen. According to one embodiment of the present invention, the human bone marrow-derived mesenchymal stem cells express 0-10% of CD34, CD45 and HLA-DR which are hematopoietic stem cell surface marker antigens, more preferably 0.1-7 %, And most preferably 0.1-5%.

The term "% " used in reference to the expression rate of the surface antigen means the ratio of cells expressing surface antigens in the cells to be analyzed. For example, a 95% expression rate of the CD29 surface antigen means that 95% of the cells express the CD29 surface antigen in the cell sample to be analyzed.

According to an embodiment of the present invention, the autoimmune diseases that can be prevented or treated by the composition of the present invention include lupus (systemic lupus erythematosus), rheumatoid arthritis, systemic sclerosis (Scleroderma) Atopic dermatitis, atopic dermatitis, alopecia areata, type I or immuno-mediated diabetes, psoriasis, pemphigus, asthma, aphthas stomatitis, chronic thyroiditis, inflammatory bowel disease, Behcet's disease, Crohn's disease, dermatomyositis, myasthenia gravis, Grave's disease, polyarteritis nodosa, hypertrophic sclerosis, multiple sclerosis, autoimmune hemolytic anemia, autoimmune hemolytic anemia, autoimmune encephalomyelitis, myasthenia gravis, Ankylosing spondylitis, Fibromyalgia syndrome, or Temporal arteritis.

According to another embodiment of the present invention, the autoimmune disease which can be prevented or treated by the composition of the present invention is lupus, rheumatoid arthritis, multiple sclerosis or Type I or immune-mediated diabetes.

According to a particular embodiment of the invention, the autoimmune disease which can be prevented or treated by the composition of the invention is lupus.

According to one embodiment of the present invention, human bone marrow-derived mesenchymal stem cells express / secrete TGFβ (transforming growth factor beta) -1 in a cell.

TGF-β is known to have three subtypes of TGF-β1, 2, and 3 in humans and is known to be a strong regulatory substance that lowers the intensity of the immune response. The human bone marrow-derived mesenchymal stem cells of the present invention reduce the autoimmune response by expressing / secreting TGF-beta1 which lowers the intensity of the immune response.

The human bone marrow-derived mesenchymal stem cells of the present invention show increased levels of COX-2, HO-1, IFN-y, IL-4 and IL-10.

As used herein, the term " highly expressed " or " increased expression " means that the degree of expression of a nucleotide sequence of interest in a sample to be examined (e.g., human bone marrow-derived mesenchymal stem cells) (Preferably, 1.2 times or more) in comparison with the case where it is compared with the case of the case where the value

According to one embodiment of the present invention, the human bone marrow-derived mesenchymal stem cells of the present invention inhibit the production of anti-dsDNA antibodies.

According to one embodiment of the present invention, the human bone marrow-derived mesenchymal stem cells of the present invention inhibit the production of proteinuria.

According to one embodiment of the present invention, the human bone marrow-derived mesenchymal stem cells of the present invention inhibit the production of IgG.

According to the present invention, the human bone marrow-derived mesenchymal stem cells of the present invention reduce immune cell infiltration. The human bone marrow-derived mesenchymal stem cells of the present invention may infiltrate into the site or organ where the autoimmune disease occurs (for example, in the case of lupus, the immune cell is infiltrated into the kidney) .

According to one embodiment of the present invention, the human bone marrow-derived mesenchymal stem cells of the present invention inhibit the proliferation of T cells or B cells involved in the immune response.

According to the present invention, the inhibition of T-cell proliferation is by a soluble mediator or cell-cell contact.

According to one embodiment of the present invention, the human bone marrow-derived mesenchymal stem cells of the present invention inhibit cytokine expression of T cells. The cytokines of T cells whose expression is inhibited by the human bone marrow-derived mesenchymal stem cells of the present invention are IL-2, IFN-g, IL-4 and IL-5.

According to one embodiment of the present invention, the pharmaceutical composition for the prevention or treatment of autoimmune disease of the present invention is a composition for parenteral administration, in particular, for intravascular administration, wherein when the composition is administered into a blood vessel, Leaf stem cells inhibit the production of anti-dsDNA antibodies, proteinuria and total IgG without toxicity, inhibit the proliferation of T cells and B cells, inhibit the expression of cytokines of T cells and prevent or treat autoimmune diseases Indicates therapeutic efficacy.

The composition of the present invention comprises (a) a pharmaceutically effective amount of the aforementioned human bone marrow-derived mesenchymal stem cells; And (b) a pharmaceutically acceptable carrier. As used herein, the term " pharmaceutically effective amount " means an amount sufficient to achieve the therapeutic efficacy or activity of the human bone marrow derived mesenchymal stem cells described above.

When the composition of the present invention is manufactured from a pharmaceutical composition, the pharmaceutical composition of the present invention includes a pharmaceutically acceptable carrier. The pharmaceutically acceptable carriers to be contained in the pharmaceutical composition of the present invention are those conventionally used in the formulation and include lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia rubber, calcium phosphate, alginate, gelatin, But are not limited to, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methylcellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, mineral oil, saline, or a medium, but are not limited thereto.

The human bone marrow-derived mesenchymal stem cells of the present invention are used by suspending cells in a pharmaceutically acceptable carrier and filling the cells into a vial, a vinyl bag, a syringe or the like.

The pharmaceutical composition of the present invention may further contain a lubricant, a wetting agent, a sweetening agent, a flavoring agent, an emulsifying agent, a suspending agent, a preservative, etc. in addition to the above components. Suitable pharmaceutically acceptable carriers and formulations are described in detail in Remington ' s Pharmaceutical Sciences (19th ed., 1995).

The pharmaceutical composition of the present invention can be administered orally, topically (buccal, sublingual, skin or ocular), transdermal or parenteral (subcutaneous, intradermal, intramuscular, intravascular or intraarticular) Parenteral administration, more preferably intravenous administration.

The appropriate dosage of the pharmaceutical composition of the present invention may vary depending on factors such as the formulation method, administration method, age, body weight, sex, pathological condition, food, administration time, administration route, excretion rate, . Typical dosages of the pharmaceutical compositions of this invention are 10 < 2 > -10 < 10 > cells per day on an adult basis.

The pharmaceutical composition of the present invention may be formulated into a unit dose form by formulating it using a pharmaceutically acceptable carrier and / or excipient according to a method which can be easily carried out by a person having ordinary skill in the art to which the present invention belongs. Or by intrusion into a multi-dose container. The formulations may be in the form of solutions, suspensions, syrups or emulsions in oils or aqueous media, or in the form of excipients, powders, powders, granules, tablets or capsules, and may additionally contain dispersing or stabilizing agents.

The features and advantages of the present invention are summarized as follows:

(I) The present invention provides a pharmaceutical composition for preventing or treating an autoimmune disease comprising human bone marrow-derived mesenchymal stem cells as an active ingredient.

(Ii) The human bone marrow-derived mesenchymal stem cells of the present invention inhibit anti-dsDNA antibody, proteinuria and total IgG production without inhibiting toxicity, inhibit the proliferation of T cells and B cells, To prevent or treat autoimmune diseases.

(Iii) Inflammation of the kidney due to autoantibodies and immunoconjugates occurs in lupus, and invasion of immune cells is increased. When human bone marrow-derived mesenchymal stem cells of the present invention are administered, immune cell infiltration into the kidney To prevent or treat lupus.

1A to 1C are images showing that the shape of a cell is not changed during the second to ninth subculture of human bone marrow-derived mesenchymal stem cells in a cell culture flask. FIG. 1A is a graph showing the bone marrow-derived mesenchymal stem of healthy donor 1 1B is an image of a donor 2, and FIG. 1C is an image of a donor 3.
FIG. 2a-c is a graph showing the growth rate of healthy human bone marrow-derived mesenchymal stem cells according to the culture period. FIG. 2A is a graph showing the proliferation rate of bone marrow-derived mesenchymal stem cells of Donor 1, FIG. 2B, Donor 2, and Donor 3 according to the incubation period.
3-5 are graphs showing surface antigens of mesenchymal stem cells during the third, fifth, seventh, and ninth subculture using flow cytometry analyzers (FACs). FIG. 3 is a graph showing the results of surface antigen analysis during the incubation period of the mesenchymal stem cell of Donor 1. FIG. 3-5 is a graph showing the surface antigen analysis during the third subculture, B is the cell in the fifth subculture, C Is a graph showing the results of surface antigen analysis of the cells in the seventh subculture, and D in the ninth subculture. FIG. 4 shows the results of analysis of the mesenchymal stem cells of Donor 2, and FIG. 5 shows the results of Donor 3 analysis.
6 is an image showing the stability of cell culture through karyotype analysis during the subculture of bone marrow-derived mesenchymal stem cells of a healthy donor. FIG. 6A is an image showing stability of the bone marrow-derived mesenchymal stem cells of Donor 1 without changing the karyotype during the subculture period, FIG. 6B is the image of Donor 2, and FIG. 6C is image of Donor 3. In each figure, A is the third cultured cell, B is the 5th, C is the 7th, and D is the 9th subculture.
FIG. 7 shows the results of RT-PCR measurement of gene expression levels of TGF-β, COX-2 and HO-1 in human bone marrow-derived mesenchymal stem cells.
FIG. 8 shows the results of RT-PCR measurement of gene expression levels of CCL2, IFN-g, IL-10, IL-4 and TGF-β in human bone marrow-derived mesenchymal stem cells.
FIG. 9 is a graph showing the expression level of TGF-β1 in secretory proteins secreted by cells in cell culture medium during the fourth passage of bone marrow-derived mesenchymal stem cells from three donors.
FIG. 10 shows the results of measuring the ability of the bone marrow-derived mesenchymal stem cells to differentiate into adipocytes.
Fig. 11 shows the result of measurement of immunogenicity of bone marrow-derived mesenchymal stem cells.
FIG. 12 shows the results of measuring the effect of suppressing the immune response of bone marrow-derived mesenchymal stem cells.
Fig. 13 shows the result of measurement of cell group change of MRL / lpr mouse organ. Expression of mouse organ cell phenotype is measured by flow cytometry. Tracheal cells were cultured in the presence of B220-APC, CD4-APC, CD11c-APC, CD3-PE, CD8-PE, CD11b-PE, CD138-APC + IgG-PE, CD4-FITC + Foxp3- Lt; RTI ID = 0.0 &gt; monoclonal < / RTI &gt;
14 shows the results of measurement of cytokine expression level in MRL / lpr mouse spleen cells. Total RNA was isolated from immune cells of mouse spleen. Gene expression levels of IL-2, IFN-γ, IL-4, IL-10, TNF-α, IL-1β, IL-12 and IL-6 were analyzed by RT-PCR. The PCR products were electrophoresed on agarose gels and stained with thiamine bromide.
FIG. 15 shows the results of measurement of survival rate (a), body weight (b), anti-dsDNA amount (c), IgG amount (d) and proteinuria amount (e) according to repeated administration of human bone marrow-derived mesenchymal stem cells. The mouse is divided into three groups. 200 μl of vehicle was intravenously injected into each mouse of the control group every 2 weeks from 10 to 20 weeks of age, and the mice were injected intravenously every 2 weeks from 10 to 20 weeks of age with the administration of human bone marrow-derived mesenchymal stem cells Mice were injected intravenously with 1 x 10 6 human bone marrow-derived mesenchymal stem cells and injected intraperitoneally with 50 mg / kg of cyclophosphamide intraperitoneally to each mouse in the CPM-treated group every 2 weeks from 10 to 20 weeks of age. The survival rate was examined at 28 weeks of age (a). The mice were weighed at 28 weeks of age to assess toxicity (b). The anti-dsDNA concentration was measured every two weeks using an ELISA (c). IgG concentration was measured by ELISA every two weeks (d). The proteinuria concentration was measured by ELISA every 2 weeks (e). Significance was determined using Student's t test for controls (* p <0.05, ** p <0.01, *** p <0.001).
FIG. 16 shows the result of measurement of anti-dsDNA amount (a), IgG amount (b) and proteinuria amount (c) according to single administration of human bone marrow-derived mesenchymal stem cells. The mouse is divided into three groups. At 9 weeks of age, 200 [mu] l vehicle was intravenously injected into each mouse, and 1 x 10 < 6 &gt; human bone marrow-derived mesenchymal stem cells were intravenously injected into each mouse of 9-week-old human bone marrow-derived mesenchymal stem cell- , 9-week-old cyclophosphamide-treated group 50 mg / kg of CPM was intravenously injected into each mouse. The anti-dsDNA concentration was measured every two weeks using an ELISA (a). IgG concentration was measured every two weeks using ELISA (b). The proteinuria concentration was measured by ELISA every 2 weeks (c). Significance was determined using Student's t test for controls (* p <0.05, ** p <0.01, *** p <0.001).
FIG. 17 shows the results of measurement of anti-dsDNA amount (a), IgG amount (b) and proteinuria amount (c) according to single administration of human bone marrow-derived mesenchymal stem cells. The mouse is divided into three groups. Control mice at 12 weeks of age were injected intravenously with 200 [mu] l vehicle, and 1 x 10 < 6 &gt; human bone marrow-derived mesenchymal stem cells were intravenously injected into each mouse of the 12-week-old human bone marrow-derived mesenchymal stem cell treatment group , 12-week-old cyclophosphamide-treated group Intravenous injection of 50 mg / kg CPM into each mouse. The anti-dsDNA concentration was measured every two weeks using an ELISA (a). IgG concentration was measured every two weeks using ELISA (b). The proteinuria concentration was measured by ELISA every 2 weeks (c). Significance was determined using Student's t test for controls (* p <0.05, ** p <0.01, *** p <0.001).
18 shows the results of measurement of anti-dsDNA amount (a), IgG amount (b) and proteinuria amount (c) according to repeated administration of human bone marrow-derived mesenchymal stem cells. The mouse is divided into three groups. 200 μl of vehicle was intravenously injected into each mouse of the control group every 3 weeks from 12 weeks to 20 weeks of age, and every 3 weeks from 12 to 20 weeks of age, intraperitoneal injections of human bone marrow-derived MSCs Mice were injected intravenously with 1 x 10 6 human bone marrow derived mesenchymal stem cells and injected intraperitoneally with 50 mg / kg cyclophosphamide intraperitoneally to each mouse of CPM treated group every 2 weeks from 12 to 20 weeks of age. The anti-dsDNA concentration was measured every two weeks using an ELISA (a). IgG concentration was measured every two weeks using ELISA (b). The proteinuria concentration was measured by ELISA every 2 weeks (c). Significance was determined using Student's t test for controls (* p <0.05, ** p <0.01, *** p <0.001).
FIG. 19 shows the results of measurement of cell group changes of MRL / lpr mouse organs after treatment with human bone marrow-derived mesenchymal stem cells. Expression of phenotype in mouse organ cells was measured by flow cytometry. Tracheal cells were cultured in the presence of B220-APC, CD4-APC, CD11c-APC, CD3-PE, CD8-PE, CD11b-PE, CD138-APC + IgG-PE, CD4-FITC + Foxp3- Lt; RTI ID = 0.0 &gt; monoclonal &lt; / RTI &gt; Significance was determined using Student's t test for controls (* p <0.05).
FIG. 20 shows the results of measurement of cytokine expression level in MRL / lpr mouse spleen cells after treatment with human bone marrow-derived mesenchymal stem cells. Total RNA was isolated from immune cells of spleen cells. The gene expression levels of IL-2, IFN-γ, IL-4, IL-10, TNF-α, IL-1β, IL-12 and IL-6 were analyzed by RT-PCR. The PCR products were electrophoresed on agarose gels and stained with thiamine bromide.
FIG. 21 shows the histological changes in MRL / lpr mouse kidney after administration of human bone marrow-derived mesenchymal stem cells. MRL / lpr mouse elongation was assessed with HE-stained sections. (C), CPM-administered MRL / lpr mouse 25 (b), human bone marrow derived mesenchymal stem cell-administered MRL / lpr mouse at 25 weeks (a) Week (d). x 400 times.
22 shows the results of measurement of survival rate (a) and body weight (b) according to repeated administration of human bone marrow-derived mesenchymal stem cells. The mouse is divided into three groups. At 12 weeks of age, each mouse in the control group was intravenously injected with 200 [mu] l vehicle and once at 12 weeks, 4 x 10 4 , 4 x 10 5 , 4 x 10 6 human bone marrow-derived mesenchymal stem cells were intravenously injected, and 50 mg / kg of cyclophosphamide was intravenously injected into each mouse of CPM treatment group once every 12 weeks.
Figure 23 shows the anti-dsDNA concentration measured every two weeks using an ELISA (a). IgG concentration was measured every two weeks using ELISA (b). The proteinuria concentration was measured by ELISA every 2 weeks (c). Significance was determined using Student's t test for controls (* p <0.05, ** p <0.01, *** p <0.001).
FIG. 24 shows the results of histologic changes in the kidney according to administration of human bone marrow-derived mesenchymal stem cells.
25 is a schematic diagram showing immunological control of human bone marrow-derived mesenchymal stem cells.
26 shows the results of measurement of phenotype of human bone marrow-derived mesenchymal stem cells. Mouse marrow cells were stained with mouse monoclonal antibodies (CD34-PE, CD45-PE, CD103-PE, CD73-PE, CD90-PE, CD105-PE, CD44-FITC and Sca-1-FITC) ). BM-MSCs cultured for 20 days were treated with mouse monoclonal antibodies (CD34-PE, CD45-PE, CD103-PE, CD73-PE, CD90-PE, CD105-PE, CD44-FITC and Sca-1-FITC) And then stained (b).
FIG. 27 shows the results of measuring the effect of human bone marrow-derived mesenchymal stem cells on B / T cell proliferation. Splenocytes were activated with LPS and ConA in the presence or absence of radioactively treated human bone marrow-derived mesenchymal stem cells in 96-well plates. Cell proliferation was assessed as [ 3 H] -thymidine hybridization. BM-MSC co-incubated with Balb / c-derived B / T cells (a). Human bone marrow derived mesenchymal stem cells are co-cultured with C57BL / 6-derived B / T cells (b). Human bone marrow derived mesenchymal stem cells are co-cultured with MRL / lpr-derived B / T cells (c). Significance was determined using Student's t test for controls (* p <0.05, ** p <0.01, *** p <0.001).
FIG. 28 shows the results of measuring the effect of human bone marrow-derived mesenchymal stem cells on T cell cytokine production. Total RNA was isolated from T cells. Gene expression levels of IL-2, IFN-y, IL-4, and IL-5 were analyzed using RT-PCR. The PCR products are electrophoresed on agarose gels and then stained with thiamine bromide (a). IFN-γ and IL-2 levels in immune cell supernatants were measured using ELISA (b).
FIG. 29 shows the results of measurement of direct or indirect effects of human bone marrow-derived mesenchymal stem cells on B / T cell proliferation. B / T cells were co-cultured with human bone marrow-derived mesenchymal stem cells using a transwell system. Cell proliferation was assessed using [ 3 H] -thymidine incorporation. Splenocytes from MRL / lpr mice are activated with LPS in the presence or absence of radioactively treated MSCs in 96 well plates (a). Splenocytes from MRL / lpr mice are activated with ConA in the presence or absence of radioactively treated MSCs in 96-well plates (b).
30 shows the results of measurement of the degree of water-soluble factor expression in human bone marrow-derived mesenchymal stem cells. On day 20, human bone marrow-derived mesenchymal stem cells were recovered. Gene expression levels of soluble elements were analyzed using RT-PCR (a). Levels of soluble elements in human bone marrow-derived mesenchymal stem cell supernatants were measured using ELISA (b). Significance was determined using the Student's t test for bone marrow cells (*** p <0.001).
31 shows the results of measurement of the expression of IDO in BM-MSC. IDO levels in human bone marrow-derived mesenchymal stem cell supernatants were measured using an ELISA (a). Human bone marrow derived mesenchymal stem cell-mediated T cell immunosuppression mechanism (b).
FIG. 32 shows the results of measurement of T-cell motility behavior by human bone marrow-derived mesenchymal stem cells. Human bone marrow-derived mesenchymal stem cells are stained with CMTMR dye (red) at 37 ° C for 15 minutes. T cells are stained with CFSE dye (green) at 37 ° C for 15 minutes. Two-dimensional cell tracking is performed using Imaris software. A snapshot image of interactions between human bone marrow derived mesenchymal stem cells and T cells.
33 shows the results of measurement of migration of T cells by human bone marrow-derived mesenchymal stem cells. The migration of T cells according to human bone marrow-derived mesenchymal stem cell concentration was measured by counting the number of cells located in the lower chamber after 1.5 hours of incubation. Significance was determined using the Student's t test for controls (*** p <0.001).
Fig. 34 shows the results of measurement of the expression level of chemokines in human bone marrow-derived mesenchymal stem cells. Human bone marrow-derived mesenchymal stem cells were recovered on day 20. Gene expression levels of chemokines were analyzed using RT-PCR (a). CCL2, CCL5 and CXCL12 levels in human bone marrow-derived mesenchymal stem cell supernatants were measured using an ELISA (b). Significance was determined using the Student's t test for bone marrow cells (*** p <0.001).
FIG. 35 shows the results of measuring the expression of chemokines after knocking down CCL2 and CXCL12 genes using siRNA, respectively.
FIG. 36 shows the results of analysis of T cells involved in migration of human bone marrow-derived mesenchymal stem cells.
FIG. 37 shows the results of analysis of contact patterns and contact time between human bone marrow-derived mesenchymal stem cells and T cells.
FIG. 38 shows the results of analysis of the role of CCR2 expressed by T cells in contact with mouse MSCs and T cells.

Hereinafter, the present invention will be described in more detail with reference to Examples. It is to be understood by those skilled in the art that these embodiments are only for describing the present invention in more detail and that the scope of the present invention is not limited by these embodiments in accordance with the gist of the present invention .

Example

Example 1: Isolation and culture of bone marrow-derived mesenchymal stem cells from healthy donor bone marrow

Approximately 30-50 mL of bone marrow was collected from a posterior superior iliac spine (PSIS) using a Jamshidi needle and transferred to a heparin tube. The heparin tube containing the bone marrow was transferred to a clean room and the subsequent procedure was performed. The heparin tube containing the bone marrow was transferred to a sterile tube and diluted 1: 3 with the basal medium (CSBM, CORESTEM Inc., Korea). The diluted solution was mixed with Ficoll (Fiocoll-Paque PREMIUM, concentration 1.077 g / ) Layer, and centrifuged at 400 x g for 30 minutes. Mononuclear cell washing was performed as follows. The mononuclear cells were harvested by centrifugation at 400 xg for 10 minutes by adding a basic medium to the mononuclear cells after mononuclear cell layer separation, and the harvested mononuclear cells were again added to the basic medium After centrifugation at 100 xg for 10 minutes. The washed mononuclear cells were incubated with 1% penicillin-streptomycin (Biochrmone, Germany), L-alanyl-L-glutamine (Biochrome, Germany), 10% (v / v) fetal bovine serum (FBS; Gibco, USA) (Nunc, USA) containing the basic medium, and then cultured at 37 ° C and 7% CO 2 for 11 days. After the incubation, cells were removed from the bottom of the cell culture flask by changing the medium twice at intervals of 3 or 4 days. Adherent cells stuck at 70-80% or more on the bottom of the cell culture flask were harvested by trypsin treatment (0.125% trypsin-EDTA; Gibco, USA).

Example 2: Cryopreservation and thawing of bone marrow-derived mesenchymal stem cells

Cells were inoculated with 1.5 x 10 5 - 2 x 10 5 cells per flask in a T175 cell culture flask (Nunc, USA) and cultured at 37 ° C and 7% CO 2 for 7 days. The culture medium was changed every 3 or 4 days.

Cells grown above 70-80% were harvested by trypsinization (0.125% trypsin-EDTA). The harvested cells were seeded in 800 μl of a cryopreservation medium consisting of 20% fetal bovine serum and 10% DMSO (dimethylsulfoxide, Sigma, USA) supplemented CSBM at a density of 1 x 10 6 cells / vial Were suspended and dispensed into cryotube vials (Nunc, USA). The cell suspension dispensed into the vials was placed in a cryopreservation container (Nalgene, USA) containing isopropyl alcohol for 24 hours at -80 ° C, and the vials containing the cells were transferred to a liquid nitrogen storage tank And

1 x 10 6 cells / vial in the vial was filled with the cell density to the next quickly removing the liquid nitrogen tank 37 ℃ transferred to a room temperature from the liquid nitrogen storage tank. Cell suspensions thawed in vials were inoculated into cell culture flasks containing cell culture medium under sterile conditions. The density of the inoculated cells was 1.5 x 10 5 - 2 x 10 5 cells per T175 flask. Cell culture medium exchange was performed at 3 or 4 days intervals. Cellularity and proliferation were observed under an optical microscope (Nikon, Japan). As shown in Fig. 1, it was confirmed that the cells were uniformly proliferated in the form of a spindle like fibroblasts (Fig. 1a-c).

Example 3: Analysis of proliferation rate of donor bone marrow-derived mesenchymal stem cells

The proliferation rate of stem cells was measured according to an increase in passage number of isolated cells. Population doubling (PD) is an index of cell proliferation. It can be expressed as population doubling (PD) = log (final number of harvested cells / number of cells inoculated per second) / log 2. The number of cells initially inoculated was determined as 1.5 x 10 5 cells in a T175 flask for each passage number, and the number of cells obtained after culturing was determined to determine the proliferation rate. The bone marrow-derived mesenchymal stem cells were obtained by the above-described separation method and then subcultured. As a result of confirming the growth rate of the cells for about 40 days (P2-P11) through the growth curve graph, it was confirmed that the population doubling time (PDT) was about 75.14 hours (FIG.

Example 4: Analysis of surface antigen expression of donor bone marrow-derived mesenchymal stem cells

(FACS) was used to determine the expression characteristics of stem cell surface antigen on cells. The mesenchymal stem cells isolated from the bone marrow were cultured at 2 × 10 6 cells in 2% fetal bovine serum (PBS) at passage 3 (P3), passage 5 (P5), passage 7 (P7) The cells were washed three times with DPBS (Gibco, USA) added thereto, suspended at a concentration of 1 × 10 5 cells / 100 μl, and then were divided into test tubes and analyzed by FACS caliber (Becton Dickinson, NJ, USA). PE-CD90, PE-CD44, PE-CD73 and PE-CD105 and PE-CD34 and PE-CD45, hematopoietic stem cell markers, PE-HLA-DR (MHC class II), a negative control, and PE-immunoglobulin isotype IgG1, a negative control antibody, were used. All antibodies were purchased from Becton Dickinson, USA . The antibody used was reacted with cells on ice for 30 minutes under the dark condition, and the extra antibody was washed with DPBS (Gibco, USA) and analyzed for flow cytometry.

More than 80% of the cells showed positive immunological characteristics for the stem cell surface surface antigens CD29, CD44, CD49C, CD73 and CD105, and all of the hematopoietic stem cell surface antigen CD34 and CD45 were less than 5% Respectively. As a result, it was confirmed that the isolated cells had characteristics of stem cells. In addition, since HLA-DR (MHC class II) which does not express tissue-specific antigen that causes rejection in tissue or organ transplantation is not expressed, it induces immune response such as rejection reaction which is a problem in transplantation of existing cells or tissues Therefore, it could be confirmed that it could be used as a cell derived from Taga (Fig. 3-5).

Example 5: Karyotype analysis of donor bone marrow-derived mesenchymal stem cells

Analysis was performed to determine whether the cells retained normal chromosome morphology and number of chromosomes during isolation and culturing. And hold the bone marrow-derived mesenchymal stem cells isolated classified (P3, P5, P7, P9 ) to 1.75 x 10 5 were inoculated with cells 37 ℃, 7% of cell growth in a CO 2 conditions, up to 75% density in T75 flasks Were harvested by trypsinization (0.125% trypsin-EDTA) and 25 metaphases of harvested cells were GTG-banded. As a result, the numerical and structural abnormality of the chromosome could not be found, and the mesenchymal stem cell Even in a continuous culture It was confirmed that the normal chromosome was maintained without changing the karyotype (Fig. 6A-C).

Example 6: Expression analysis of secretory factors of bone marrow-derived mesenchymal stem cells of a donor

The factors secreted by human MSC were confirmed by RT-PCR. To this end, human MSCs were recovered in culture flasks and reacted with 200 μl of trizol for 5 minutes at room temperature. 200 ml of chloroform was added to the same tube, followed by vortexing at room temperature for 5 minutes, followed by centrifugation at 4 ° C and 12,000 rpm for 15 minutes. 500 ml of the supernatant was transferred to a new tube, equilibrated with isopropanol, allowed to react at room temperature for 10 minutes, and then centrifuged at 4 ° C and 12,000 rpm for 15 minutes. The supernatant was removed and 1 ml of 75% EtOH was added to the RNA pellet. The RNA pellet was obtained by centrifugation at 4 ° C and 12,000 rpm for 15 minutes. 50 ml of RNase free water was added to the obtained RAN, the RNA pellet was dissolved at room temperature for 10 minutes, and the concentration and purity of RNA were measured using a nano-drop. CDNA was synthesized at a concentration of 1 mg of each RNA.

cDNA synthesis was performed according to the Maxime RT premix kit method. The reverse transcription reaction was carried out in a nucleic acid amplification machine (PCR machine) at 45 ° C for 60 minutes and reacted at 95 ° C for 5 minutes to inactivate the reverse transcriptase. The synthesized cDNA was subjected to PCR.

Real-time PCR machine was Rotor-Gene Q5 Plex. 10 μl of a 10-pmole F / R primer, 0.6 μl of a 1 μg template cDNA, and 7.4 μl of Rnase-free water were used for the test. To make a total volume of 20 μl, and PCR is carried out under the following conditions; Denaturation: 95 ° C for 10 minutes, Denaturation: 95 ° C for 15 seconds, Annealing & extension: 60 ° C for 1 minute. Repeat this process 40 times.

The gene expression of TGF-beta, COX-2 and HO-1 of human MSC was confirmed by RT-PCR (Fig. 7A). TGF-β is an immunomodulatory cytokine that is known to induce the differentiation of regulatory T cells, and is an important factor in the control of autoimmune disease. COX-2 is an enzyme that produces prostaglandins. Activation of COX-2 in MSCs is known to result in the production of prostaglandins and inhibition of proliferating cellular responses by this substance. HO-1 is also an important factor involved in the regulation of immune cells by MSCs.

Treatment of IFN-g with human MSCs resulted in increased expression of IDO in addition to TGF-β, COX-2, and HO-1. IDO is a protein that plays an important role in inhibiting the immune response, and is strongly involved in inhibiting the activity of macrophages, B cells, and the like (Fig. 7B). In addition to gene expression, human MSCs express TGF-β and prostaglandin E (PGE 2 ) proteins.

FIG. 8 shows that the genes of CCL2, IFN-γ, IL-10, IL-4 and TGF-β were highly expressed in human MSCs. CCL2 secreted by MSC is a chemokine that plays an important role in contact with immune T cells. When MSCs express CCL2, T cells migrate to where MSCs are located, which will facilitate the suppression of T cells by MSCs. IFN-y stimulates the contact with T cells by increasing the expression of B7-H1 in MSC, thereby inducing an immunosuppressive reaction effectively by MSC. IL-4 is a cytokine that differentiates T cells into Th2 cells, and activates M2 macrophages (anti-inflammatory actions) to inhibit the activity of M1 macrophages (inflammation) and regulate the inflammatory response. IL-10 secreted by MSCs is a typical anti-inflammatory cytokine that modulates an abnormally activated immune response. These results suggest that MSC may effectively suppress the immune response by controlling various chemokine and cytokine expressions.

Example 7: Confirmatory expression of secretory factors of bone marrow-derived mesenchymal stem cells from donors

As an autoimmune disease, lupus is a disease caused by the collapse of normal immunoregulatory system, and it is aimed to confirm the expression level of immunoregulatory factors secreted from mesenchymal stem cell itself.

TGFβ (transforming growth factor beta) -1 expression, which directly or indirectly influences the inexperienced T cells among the various factors expressed in the bone marrow mesenchymal stem cell itself, induces differentiation into regulatory T cells, (P4) of bone marrow-derived mesenchymal stem cells isolated from this donor can induce an anti-inflammatory effect and an immune regulating effect which is excessively active against self-antigens when the cells are administered to a patient suffering from lupus erythematosus 50 μl of the cell culture was subjected to a manual test of the kit using a TGF-β ELISA kit (Quantikine, R & D Systems). As a result, it was confirmed that cells that secrete about 36 pg of TGFβ-1 per 10,000 cells in passage number 4 (P4) of bone marrow-derived mesenchymal stem cells of three donors, and secreted at least 20 pg or more ).

Example 8: Analysis of the ability of the bone marrow-derived mesenchymal stem cells of donors to differentiate and regulate immunity

The ability of bone marrow-derived mesenchymal stem cells to differentiate

MSC has multiple differentiation potential into mature cells of various mesenchymal tissues such as chondrocytes, muscle cells, adipocytes, and osteoblasts. Therefore, it was confirmed whether human MSC could be differentiated into various kinds of cell lines in vitro.

Cells were cultured in adipocyte differentiation medium for 14 days to analyze adipocyte differentiation potential. Morphological changes of differentiated adipocytes were confirmed by oil red O staining. Differentiated adipocytes accumulated droplets and turned red when stained with oil red O. This resulted in differentiation into adipocytes (Fig. 10B).

Cells were cultured in osteoclast differentiation medium for 21 days in order to analyze bone cell differentiation ability. An alizarin red staining method was used to confirm the differentiation. In the negative control group of Fig. 10C, no staining with alizarin red was observed. On the other hand, the differentiated cells were stained red with alizarin red to confirm that calcium was formed (Fig. 10D).

To analyze the ability of chondrocyte differentiation, pellet cultures were performed for 28 days in chondrocyte differentiation medium. The characteristics of the differentiated chondrocytes were analyzed by immunohistochemistry (Aggrecan staining method). Aggrecan used in the analysis is protein polysaccharide present in cartilage. As shown in Fig. 10, aggrecan protein was expressed in the differentiated chondrocytes and cartilage formation was observed (Fig. 10E-G).

Immune regulation ability of bone marrow-derived mesenchymal stem cells

We evaluated the immunogenicity of human MSCs and their ability to regulate immune responses to allogeneic immune cells. PBMC were isolated from normal human blood of HLA type and immunogenicity was measured by co-culture with three other human MSCs. Human MSCs did not induce the proliferation and IFN-y production of the homologous PBMCs, indicating no immunogenicity (Fig. 11).

PBMCs induced by proliferation of PHA, which is a proliferation inducer of lymphocytes, and human MSCs were co-cultured to confirm the immune response inhibitory effect of human MSCs. PBMC proliferation was induced by PHA, the proliferation reaction was inhibited by human MSC, and IFN-y production was inhibited (FIG. 12).

Example 9: Effect of MSC on lupus treatment Experimental method

Human MSC

The human stem cells were provided with human MSCs for use in animal experiments.

Lupus animal experiment

MRL / lpr mice, widely known as spontaneous lupus animal models, were used. Human MSCs were intravenously injected into mice at a concentration of 1 x 10 cells / mouse. Cyclophosphamide (CPM), an immunosuppressive agent, was used as a positive control, and the mice were intravenously injected at a concentration of 50 mg / kg.

Mouse MSC culture

Balb / c mice were sacrificed by cervical dislocation and single cells were obtained by separating bone marrow from femur and tibia using a 10 ㎖ syringe. The impurities were filtered using a 0.7 μm filter and the cells were centrifuged at 1,200 rpm for 3 minutes. After centrifugation, the supernatant was removed and red blood cells were removed by treatment with ACK dissolution buffer for 1 minute. The final isolated cells were diluted in a-MEM medium at a cell number of 1 x 10 7 cells / ml and cultured in a 37 ° C incubator fed with 5% CO 2 . 1 &lt; / RTI &gt; ml of fresh medium was added daily for 5 days. After 6 days of culture, the α-MEM medium was removed, washed with PBS, and 1 ml of fresh medium was added. Cells that were cultured for 20 days were used for the experiment.

Cell phenotype analysis

Cell surface analysis was performed using flow cytometry. The recovered cells (1 x 106 cells / ml) were washed with 0.5% BSA / PBS. The supernatant was removed and 50 μl of 0.5% BSA / PBS containing monoclonal antibody was added and stained at 4 ° C for 20 minutes. After washing again with 500 [mu] l of PBS / BSA, 500 [mu] l of PBS / BSA was added and suspended. The monoclonal antibodies may be selected from the group consisting of PE-conjugated CD3, CD8, CD34, CD45, CD73, CD90, CD103, CD105, CD11c, CD11b, IgG, Foxp3, FITC-conjugated CD44, Sac-1, CD11c, CD4, And CD4 were used. Data analysis was performed using WinMDI software.

Reverse transcription polymerase chain reaction (RT-PCR)

Total RNA was isolated from tissues and cells using TRIZOL reagent. Total RNA 0.3 was used to synthesize cDNA at 42 ° C for 60 minutes and 94 ° C for 5 minutes. PCR (polymerase chain reaction) was performed using 3 μl of the prepared cDNA and 10 pM of primer. The basic conditions for the PCR were denaturation starting at 94 ° C for 5 minutes, 30 cycles of 94 ° C for 30 seconds, annealing, 72 ° C for 1 minute, and post-elongation at 72 ° C for 5 minutes Respectively. The annealing temperature for each primer was varied between 54-56 ° C. The resulting PCR product was loaded with 8 μl of 1% agarose gel and electrophoresed at 50 V.

HE staining method

Extracted kidneys from MRL / lpr mice were fixed in 10% paraformaldehyde for 18 hours to make 4 ㎛ paraffin sections. Paraffin was removed from tissue slides using xylenes, and the cells were washed with running water for 10 minutes. After staining with hematoxylin solution for 1 minute, it was left in running water for 1 minute to remove residual staining solution. And then stained with eosin solution for 1 minute. The dyed slides were dehydrated using alcohol and xylene, and observed and photographed with an optical microscope.

Cell migration assay

The transfer ability of T cells to MSCs was measured using Transwell. 1 × 10 5 T cells were placed in the upper chamber, and 0.3 × 10 4 , 1 × 10 4 , and 3 × 10 4 MSCs were added to the lower chamber, followed by incubation at 37 ° C. for 1.5 hours. The number of cells transferred to the lower chamber was then measured using a flow cytometer.

Cell proliferation assay

Thymidine uptake is performed using cells isolated from the spleen of MRL / lpr mice. Splenocytes are diluted in RPMI complete medium at a cell number of 1 x 10 6 cells / ml and dispensed. The splenocytes were treated with 1 ㎍ / ㎖ of LPS and ConA, treated with MSC at various cell concentrations, and cultured in a 37 ° C incubator. After 54 hours, thymidine was treated at a concentration of 1 μCi / well and cultured for 18 hours, collected in a glass fiber filter using a cell harvester, and then dried for 2 hours. The dried glass fiber filter was put into a sample bag and the filter was sufficiently wetted with a scintillation cocktail and sealed. Finally, the amount of radioactivity introduced into the DNA of the cells was measured using a glass fiber filter with a beta counter.

Enzyme immunoassay (ELISA)

Mouse serum and urine were obtained and anti-dsDNA ELISA kit, IgG ELISA kit and proteinuria ELISA kit were used. MSC supernatants were obtained, and TGF-beta ELISA kit, PGE2 ELISA kit and IDO ELISA kit were used, and the procedure was carried out according to the procedure described in each kit.

Cell imaging

Cell imaging was performed using a cell culture microscope. MSCs were treated with 5 μM CMTMR dye and stained at 37 ° C for 15 min. T cells were treated with 5 μM CFSE dye and stained at 37 ° C for 15 min. After the level of 5 x 10 4 cells in 35 culture dishes ㎜ put the MSC of T cells and 5 x 10 3 cells was measured for cell imaging. Cellular images were measured every 6 minutes for 6 hours and data analysis was performed using Imaris software.

Histopathology

Mouse kidney tissue was fixed in 10% formaldehyde solution for 3 days and paraffin blocks were cut into 4 mm slides. Hematoxylin-eosin (H & E) staining or periodic acid-Schiff (PAS) staining was performed to observe histologic changes. Immuno-histochemistry was performed by firstly transferring slides into 0.01 M citrate solution and heating to express the antigen. Slides were then placed in a solution of 3% H 2 O 2 in methanol to block the endogenous peroxidase activity. Slides were incubated overnight at 4 ° C with dilution of CD3, B220, F4 / 80, Foxp3, and CD209b antibodies. Vectastain ABC kit was used to ensure 3,3'-diaminobenzidine (DAB) staining. After immunohistochemical staining, hematoxylin was stained as a control.

Example 10: Effect of human MSC on lupus treatment

In order to examine the therapeutic effect of human MSC, MRL / lpr, widely known as an animal model of spontaneous lupus, was adopted. MRL / lpr shows high levels of autoantibodies such as anti-ssDNA antibody, anti-dsDNA antibody and rheumatoid factor, and high immunoglobulin concentration, resulting in an increase in the amount of immune complex. This overexpressed phenotype is due to autosomal recessive mutations, called lymphoproliferation (lpr). The lpr mutation located on chromosome 19 modifies the transcription of the Fas receptor. As a result, deficiency of Fas signaling inhibits apoptosis and causes lupus symptoms. The difference from the classically used NZB / W F1 in the lupus animal model is that the mouse suddenly dies and both females and males display symptoms of lupus.

The MRL / MpJ-Fas lpr / J mouse was purchased as one of the MRL / lpr mouse species and the characteristics before and after the onset were analyzed. As described earlier, mice mutate the Fas receptor, resulting in the lymphocyte proliferation and the generation of immune complexes. At about 12 weeks of age, females begin to die on average at about 17 weeks, and males at about 22 weeks. First, 6-week-old mice before the onset of lupus and 25-week old mouse organs with the highest onset were excised and compared. The weight and number of mouse organs increased after the onset (Table 1). To observe the phenotypic changes of the cells obtained from each organ, we analyzed using a flow cytometer. The most prominent change in MRL / lpr mice after onset was the increased proportion of B220 + CD3 + , the lymphoma phenotype in almost all tissues. In addition, the ratio of CD4 + Foxp3 + , the phenotype of Treg cells responsible for immunosuppression, decreased after onset (FIG. 13). In order to analyze cytokine expression as MRL / lpr mice progressed, RNA was isolated from mouse spleen cells and RT-PCR was performed. As a result, it was confirmed that the expression of the inflammatory cytokine of the mice in which the disease progressed was increased before the onset (Fig. 14).

Figure pat00001

MSCs have been reported to be less susceptible to transplantation because of less expression of HLA-DR, which is a major factor in graft-versus host response, and less expression of CD40, CD80 and CD86 phenotypes (Ryan JM, Barry FP, Murphy JM, Mahon BP. , J Inflamm (Lond) Jul 26; 2: 8 (2005)).

MRL / lpr mice were used to measure the lupus improvement effect of human MSCs. MSCs were intravenously injected into mice at a concentration of 1x10 6 cells / mouse. The doses were injected 6 times at intervals of 2 weeks starting from 10 weeks of onset of the onset of the mice. Survival rates and body weights of the mice were measured weekly as an indicator of animal test, and anti-dsDNA, proteinuria and IgG were measured at 2-week intervals. Cyclophosphamide was used as a positive control.

MRL / lpr mice began to die from 13 weeks of age, and around 22 weeks, 90% of mice died. Human MSCs increased the survival rate of mice and 90% of mice survived to 28 weeks of age (Fig. 15A). Human MSC did not affect the body weight of MRL / lpr mice, indicating that human MSCs did not show toxicity to mice (Fig. 15B). The concentration of anti-dsDNA Ab in the blood of MRL / lpr mice increased from 10 weeks of age and peaked at 20 weeks of age. Human MSCs were effective in reducing the concentration of serum anti-dsDNA Ab (Figure 15c). The total IgG concentration in the MRL / lpr mouse increased from 8 weeks, peaked at 20 weeks, and the human MSC inhibited the increase in the total IgG concentration in blood (FIG. 15d). The proteinuria of MRL / lpr mice steadily increased from 20 weeks of age, and human MSCs decreased the proteinuria concentration (Fig. 15e).

In summary, human MSC inhibited the production of anti-dsDNA Ab, proteinuria, and total IgG of MRL / lpr and increased the survival rate of mice. It is similar to the treatment effect of cyclophosphamide currently in clinical use. However, weight-reducing cyclophosphamide showed toxicity, while human MSC did not show toxicity (FIG. 15).

In the following animal experiments, the effect of improving human lupus was confirmed by intravenously injecting human MSC of 1 x 10 6 cells / mouse into MRL / lpr mice only once at 9 weeks of age before the onset of lupus. Human MSC inhibited the production of anti-dsDNA and IgG in MRL / lpr mouse blood and inhibited the production of proteinuria. However, the effect was strongly observed for 3 weeks after administration, but the treatment effect tended to weaken after 4 weeks (Fig. 16).

MRL / lpr mice were injected with 1 x 10 6 cells / mouse of human MSC intravenously once at 12 weeks of the onset of lupus erythematosus, and the improvement of lupus was confirmed. Human MSC inhibited the production of anti-dsDNA and IgG in MRL / lpr mouse blood and inhibited the production of proteinuria. However, the inhibitory effect was observed for 3 weeks after administration, but the therapeutic effect tended to weaken after 4 weeks (Fig. 17). Through the above animal experiments, it was found that when the human MSCs were administered to MRL / lpr mice, the effects varied depending on the number of administrations. The effect continued for 3 weeks after the single dose, but the effect was weaker after 4-5 weeks.

In the following animal experiments, human MSCs were intravenously injected into MRL / lpr mice at a dose of 1 x 10 6 cells / mouse for a total of 3 times at intervals of 3 weeks from the 12th week. Human MSC inhibited the production of anti-dsDNA and IgG in MRL / lpr mouse blood, and also suppressed the production of proteinuria (Fig. 18).

The mice were autopsied and analyzed for organ weight, immune cell phenotype, cytokine expression level, and kidney cell infiltration. As a result of measurement of long term weights of mice, there was no significant difference in long term weight among the groups administered with human MSC compared to the control group. However, the number of cells in all the organs except the thymus cells (Table 2).

Figure pat00002

The ratio of CD138 + IgG and B220 + CD3 + , a plasma cell phenotype, and a lymphocyte phenotype, were decreased in the human MSC-treated group compared to the control group, and the ratio of the Treg cell phenotype The ratio of CD4 + Foxp3 + was increased (Fig. 19). Splenocyte RNA isolation and analysis of cytokine expression level by RT-PCR showed that the amount of inflammatory cytokine expression in the MSC-treated group was significantly decreased compared to the control (Fig. 20).

Lupus disease is caused by inflammation of the kidneys by autoantibodies and immune complexes, and the infiltration of immune cells is increased. Mouse kidneys were isolated and the extent of cell invasion was determined by HE staining. It was confirmed that human MSC reduced immune cell infiltration into the kidney (FIG. 21).

Human MSCs were administered at 4 x 10 4 , 4 x 10 5 , and 4 x 10 6 cells / head at 12 weeks of age in a single intravenous dose to determine the minimum concentration, number of doses, and route of administration for which the human MSCs were effective in MRL / lpr mice. Survival rates and weights were measured weekly. Serum was separated every two weeks to measure the anti-dsDNA antibody and IgG concentration, and protein concentration was measured by separating urine. At 21 weeks of age, observation was terminated and kidneys were extracted and tissue analysis was performed.

The human MSC increased the survival rate of MRL / lpr mice and the lowest effective concentration was 4 x 10 4 cells / 40 g, and treatment effect was obtained by single intravenous administration (Fig. 22). Human MSC significantly inhibited the production of anti-dsDNA antibodies, IgG, and proteinuria in a concentration-dependent manner, but the administration effect was higher as the administration concentration was higher (FIG. 23). Histopathological examination showed that the infiltration of T cells, B cells, neutrophils, macrophages and dendritic cells by inflammatory reaction was increased in the kidney of the control group and decreased by the administration of human MSC. While the proportion of regulatory T cells was increased by administration of human MSC (Figure 24).

Example 11: Study on the mechanism of immunosuppressive action of MSC

MSC inhibits innate immunity and adoptive immunity and is known to inhibit direct contact and solubility factors between cells and cells. The solubility factors secreted by MSC include NO, IDO, TGF-β, IL-10 and PGE 2 . Such solubility factors have been reported to inhibit the activity and function of T cells, B cells, NK cells and dendritic cells (Fig. 25).

Mouse bone marrow derived MSCs were used for the mechanism studies. A flow cytometer was used to identify phenotypes of MSCs produced in mouse bone marrow. The cell phenotype of MSC derived from mouse bone marrow decreased the expression of hematopoietic stem cell phenotypes such as CD34, CD45, and CD103, and the expression of MSC phenotypes CD73, CD90, CD105, CD44, and Sca-1 (Fig. 26).

Suppression of proliferation of mouse T and B cells by MSC

MSC was confirmed by mitogen assays to inhibit the proliferation of mouse T and B cells. MSCs were prepared by using bone marrow cells from Balb / c mice and isolated from splenocytes of Balb / c (syngenic), C57BL6 (allogenic) and MRL / lpr (allogenic) mice of T cells. MSCs and T cells were mixed at a ratio of 0.001-0.1: 1 and treated with ConA to induce proliferation of T cells. Experimental results confirmed that MSC inhibited ConA-induced T cell proliferation (FIG. 27). MSC and B cells were mixed at a ratio of 0.001-0.1: 1 and LPS was treated to induce proliferation of B cells. As a result, it was confirmed that MSC inhibited LPS-induced B cell proliferation (FIG. 27).

Suppression of cytokine expression of mouse T cells by MSC

MSC inhibited cytokine expression in mouse T cells using RT-PCR and ELISA. MSCs were prepared using bone marrow cells from Balb / c mice and T cells were isolated from splenocytes from MRL / lpr (allogenic) mice. MSC and T cells were mixed at a ratio of 0.1: 1 and treated with ConA to induce T cell cytokine expression. As a result, it was confirmed that MSC inhibits the expression of IL-2, IFN-g, IL-4 and IL-5 in T cells (FIG. 28).

MSC immunosuppressive effect on cell contact

Transwell analysis was performed to determine whether the MSC immunosuppressive effect was due to cell contact or by an aqueous mediator. Balb / C derived MSCs and MRL / lpr mouse derived B / T cells were tested at a ratio of 1:10.

When MSCs and B cells were mixed in the lower layer wells, MSCs could inhibit proliferation of B cells by secreting a contact or aqueous mediator, MSCs were added to upper wells, and B cells to MSC Can inhibit the proliferation of B cells only through a water soluble mediator.

As can be seen in Figure 29A, the addition of MSC to the upper and lower wells inhibited the proliferation of B cells but decreased the inhibitory effect when MSC was added to the upper wells. This implies that MSC inhibits the function of B cells not only by a water-soluble mediator, but also by cell-cell interactions. 29b, it was found that MSC inhibited the proliferation of T cells by water-soluble mediator and cell-cell contact.

Solubility Factors Secreted by MSC

RT-PCR and ELISA were performed to analyze the soluble factors secreted by MSC. MSC showed a greater expression of NO, TGF-β and PGE2 than the control group (FIG. 30). MSC itself did not express the inhibitory substance IDO (FIG. 30), but it was confirmed that the IDO expression level of MSC was increased when co-cultured with T cells (FIG. 31).

Observation of interactions between MSC and T cells

To observe the interaction between MSC and T cells directly, cell movement was measured using a real time microscope. Image analysis showed that T cell movement was classified into three types. The first type of T cells migrated freely without any contact with the MSC and moved at a rate of about 5 [mu] m / min (Fig. 32a). The second type of T cells was in continuous contact with the MSC and the rate was reduced to 1.5 占 퐉 / min (Fig. 32b). The third type of T cell is a cell that contacts the MSC in the middle of migration, and at the initial stage of migration it showed a speed of about 5 ㎛ / min, but after contact with MSC it decreased at a rate of about 1 ㎛ / min ).

When the two cells were co-cultured, it was confirmed that T cells gathered around MSC to form flock (Fig. 32d). Next, we investigated the mechanism by which T cells gathered around MSC to form flock. MSC was added to the lower well of the transwell, and T cells were added to the upper well. After 90 minutes, the number of T cells migrated to the lower layer well was measured and the migration of T cells toward the MSC was measured. It was confirmed that as the concentration of MSC in the lower layer well increases, the T cell migration increases (FIG. 33). Next, we investigated the substances that induce T cell migration. Expression of chemokine, a representative substance that causes migration, was analyzed by RT-PCR and ELISA. As shown in FIG. 34, it was confirmed that MSCs expressed CCL2 and CXCL12 at a higher level than the control group.

Chemokine analysis involved when mouse MSC and T cells contacted

To identify the chemokines involved in the contact between mouse MSCs and T cells, we treated each gene with CCL2 and CXCL12 siRNAs that were highly expressed by MSCs. MSCs whose expression is decreased by each gene siRNA are named CCL2-KD MSC and CXCL12-KD MSC. When mouse MSCs were treated with CCL2 siRNA (Fig. 35A) and CXCL12 siRNA (Fig. 35B), the expression of each chemokine gene and the amount of protein produced were inhibited.

MSC was added to the lower well of Transwell and T cells were added to the upper well. At this time, the MSC used a control MSC, a CCL2-KD MSC and a CXCL12-KD MSC that did not process anything. The number of T cells migrated to the lower layer well was measured and the migration of T cells towards the MSC was measured. As a result, migration of T cells to CCL2-KD MSCs was reduced (FIG. 35C). The CXCL12-KD MSCs migrated to the lower layer wells of the upper-layer wells in a concentration-dependent manner with MSC as a result of the same control MSCs. Observations on the image also showed that T cells did not move in the direction of CCL2-KD MSC (Fig. 35D).

Analysis of movement of MSC

The contact between MSC and T cells was confirmed and their movement was analyzed. Based on the contact with the MSC, the T cell that does not contact the MSC is called the wandering T cell and the T cell that tries to contact with the MSC is called the searching T cell and the T cell that contacted the MSC is called the contacting T cell 36A).

CCL2-KD MSCs and CXCL12-KD MSCs were analyzed for changes in contact with T cells. CXCL12-KD MSC showed no significant difference in the contact patterns of control MSCs and T cells. However, CCL2-KD MSCs showed an increase in the proportion of wandering T cells and a reduction in the proportion of contacting T cells compared to the control MSC (Fig. 36B).

Wandering T-cells and searching T-cells were free to move without specific direction, and contacting T-cells in contact with MSC stagnated in one place with little movement (Fig. 36C). These movements were the same regardless of the conditions of the MSC. There was no difference in contact T cell velocities between Wandering T cells and searching T cells compared to control MSC, CCL2-KD MSC, and CXCL12-KD MSC (Fig. 36D).

Contact time analysis of MSC and T cells

MSC and T cells were co-cultured and analyzed for 6 hours. The contact timing of contact between MSC and T cells is indicated by a red arrow. 37A, the contact time between MSC and T cells is not constant but variously observed. However, the contact time of CCL2-KD MSC was shorter than that of control MSC and CXCL12-KD MSC.

The number of contacts of control MSC, CXCL12-KD MSC, and CCL2-KD MSC with T cells observed at 6 hours was 11.4, 10.6, and 10.2, respectively (Fig. 37B). However, contact time showed that the contact time between CCL2-KD MSC and T cells was remarkably low. The contact times of control MSC, CXCL12-KD MSC, and CCL2-KD MSC with T cells were measured at 107, 97 and 34 minutes, respectively (Fig. 37C). As a result, when MSC and T cells contacted, it was found that the chemokine CCL2 expressed by MSC plays an important role.

Role of TCR-expressing CCR2 in the contact between mouse MSC and T cells

The test was performed with a CCR2 antagonist to compensate for the conclusion that CCL2 is important when mouse MSCs and T cells are in contact. CCR2 is a receptor for CCL2, a chemokine receptor expressed in T cells. CCR2 antagonist was treated with T cells at a concentration of 100 μg / ml or more, indicating that the cytotoxicity was more than 100 μg / ml (FIG. 38A).

T cells treated with CCR2 antagonist at a concentration of 3, 10, and 30 μg / ml were inoculated into upper wells of Transwell. MSC was inoculated in the lower well and T cell migration was measured. As the concentration of CCR2 antagonist increases, the number of T cell migrating to MSC decreases (FIG. 38B).

T cells were treated with CCR2 antagonist 30 쨉 g / ml and inoculated into the upper well of the transwell. The MSCs were treated by concentration in the lower layer wells and the migration of T cells was observed. T cells that were not treated with the CCR2 antagonist showed an increase in MSC concentration-dependent migration to MSC. However, T cells treated with CCR2 antagonist inhibited migration to MSC (Fig. 38C). Thus, it has been demonstrated that cell-cell contact occurs through interaction between CCL2 of MSC and CCR2 of T cell.

While the present invention has been particularly shown and described with reference to exemplary embodiments thereof, it is to be understood that the same is by way of illustration and example only and is not to be construed as limiting the scope of the present invention. Accordingly, the actual scope of the present invention will be defined by the appended claims and their equivalents.

Claims (12)

The present invention relates to a pharmaceutical composition for preventing or treating an autoimmune disease comprising human bone marrow-derived mesenchymal stem cells as an active ingredient, wherein the human bone marrow-derived mesenchymal stem cell comprises (i) a cell line consisting of CD105, CD29, CD44, CD73 and CD90 Positive immunological characteristics for at least one surface antigen selected from the group consisting of; And (ii) at least one surface antigen selected from the group consisting of CD34, CD45 and HLA-DR.
The method of claim 1, wherein the autoimmune disease is selected from the group consisting of lupus (systemic lupus erythematosus), rheumatoid arthritis, progressive systemic sclerosis, scleroderma, atopic dermatitis, alopecia areata, psoriasis, Inflammatory bowel disease, Behcet's disease, Crohn's disease, dermatomyositis, polymyositis, multiple sclerosis, autoimmune hemolytic anemia, autoimmune hemolytic anemia, asthma, aphthous stomatitis, chronic thyroiditis, inflammatory bowel disease, Behcet's disease, Which is composed of autoimmune encephalomyelitis, Myasthenia gravis, Grave's disease, Polyarteritis nodosa, Ankylosing spondylitis, Fibromyalgia syndrome and Temporal arteritis. &Lt; / RTI &gt; or a pharmaceutically acceptable salt thereof.
The method according to claim 1, wherein the human bone marrow-derived mesenchymal stem cells express TGF-beta (transforming growth factor beta) -1, COX-2, HO-1, IFN- ?, IL- &Lt; / RTI &gt;
The pharmaceutical composition according to claim 1, wherein said human bone marrow-derived mesenchymal stem cells inhibit the production of anti-dsDNA antibody.
The pharmaceutical composition according to claim 1, wherein the human bone marrow-derived mesenchymal stem cells inhibit the production of proteinuria.
The pharmaceutical composition according to claim 1, wherein the human bone marrow-derived mesenchymal stem cells inhibit the production of IgG.
2. The pharmaceutical composition according to claim 1, wherein said human bone marrow-derived mesenchymal stem cells reduce immune cell infiltration into the kidney.
The pharmaceutical composition according to claim 1, wherein the human bone marrow-derived mesenchymal stem cells inhibit proliferation of T cells or B cells.
9. The pharmaceutical composition according to claim 8, wherein the T cell proliferation inhibition is by a soluble mediator or cell-cell contact.
2. The pharmaceutical composition according to claim 1, wherein said human bone marrow-derived mesenchymal stem cells inhibit cytokine expression of T cells.
2. The pharmaceutical composition according to claim 1, wherein the composition is for parenteral administration.
2. The pharmaceutical composition according to claim 1, wherein the parenteral administration is intravascular administration.
KR1020140097129A 2013-07-30 2014-07-30 Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow KR20150016117A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
KR1020140097129A KR20150016117A (en) 2013-07-30 2014-07-30 Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow
PCT/KR2014/011055 WO2016017863A1 (en) 2014-07-30 2014-11-18 Pharmaceutical composition for preventing or treating autoimmune diseases, containing human bone marrow-derived mesenchymal stem cells as active ingredient

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
KR1020130090477 2013-07-30
KR20130090477 2013-07-30
KR1020140097129A KR20150016117A (en) 2013-07-30 2014-07-30 Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow

Related Child Applications (1)

Application Number Title Priority Date Filing Date
KR1020160023254A Division KR102084974B1 (en) 2013-07-30 2016-02-26 Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow

Publications (1)

Publication Number Publication Date
KR20150016117A true KR20150016117A (en) 2015-02-11

Family

ID=55221152

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020140097129A KR20150016117A (en) 2013-07-30 2014-07-30 Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow

Country Status (2)

Country Link
KR (1) KR20150016117A (en)
WO (1) WO2016017863A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160125759A (en) 2015-04-22 2016-11-01 강원대학교산학협력단 Adult stem cell derived skin tissue of human and cell therapeutic agent
WO2017142165A1 (en) * 2016-02-19 2017-08-24 서울대학교산학협력단 Stem cells overepxressing heme oxygenase-1, and pharmaceutical composition containing same
KR20180023865A (en) * 2016-08-26 2018-03-07 연세대학교 산학협력단 Pharmaceutical composition using secretomes from mesenchymal stem cells for treating rheumatoid arthritis and a method using the same
JP2018517900A (en) * 2015-05-05 2018-07-05 メゾブラスト・インターナショナル・エスアーエールエル Efficacy test
WO2019039922A1 (en) * 2017-08-25 2019-02-28 연세대학교 산학협력단 Pharmacological composition for prevention or treatment of lupus, comprising mesenchymal stem cell-derived secretome
WO2019190246A1 (en) 2018-03-29 2019-10-03 아키소스템바이오스트래티지스(주) Method for isolating stem cells from human umbilical cord
WO2019240507A1 (en) * 2018-06-15 2019-12-19 코아스템(주) Biomarker for predicting effectiveness for autoimmune disease treatment, diagnostic kit, and use for treatment
CN110753751A (en) * 2017-05-04 2020-02-04 迈索布拉斯特国际有限公司 Mesenchymal lineage precursor or stem cells with enhanced immunosuppression
CN117159589A (en) * 2023-11-01 2023-12-05 北京国卫生物科技有限公司 Stem cell treatment method for treating systemic lupus erythematosus

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102180111B1 (en) * 2018-11-09 2020-11-17 코아스템(주) Pharmaceutical composition comprising stem cells treated with protein kinase c activator or culture thereof for prevention and treatment of autoimmune diseases

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100908481B1 (en) * 2006-04-24 2009-07-21 코아스템(주) Mesenchymal stem cell culture medium and culture method of mesenchymal stem cells using the same
KR101301262B1 (en) * 2009-10-23 2013-08-27 가톨릭대학교 산학협력단 Mesenchymal stem cell introduced with a nucleotide sequence encoding TGFβ and use thereof
WO2013085303A1 (en) * 2011-12-06 2013-06-13 주식회사 강스템홀딩스 Canine amniotic membrane-derived multipotent stem cells
KR20130111448A (en) * 2012-03-29 2013-10-10 가톨릭대학교 산학협력단 Composition for treating immune disorders comprising mesenchymal stem cells

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160125759A (en) 2015-04-22 2016-11-01 강원대학교산학협력단 Adult stem cell derived skin tissue of human and cell therapeutic agent
JP2018517900A (en) * 2015-05-05 2018-07-05 メゾブラスト・インターナショナル・エスアーエールエル Efficacy test
US11795435B2 (en) 2015-05-05 2023-10-24 Mesoblast International Sárl; Potency assay
JP2021036892A (en) * 2015-05-05 2021-03-11 メゾブラスト・インターナショナル・エスアーエールエル Potency assay
WO2017142165A1 (en) * 2016-02-19 2017-08-24 서울대학교산학협력단 Stem cells overepxressing heme oxygenase-1, and pharmaceutical composition containing same
KR20170098056A (en) * 2016-02-19 2017-08-29 서울대학교산학협력단 Heme Oxygenase-1 Overexpressing Stem Cell and Pharmaceutical Composition Comprising The Same
KR20180023865A (en) * 2016-08-26 2018-03-07 연세대학교 산학협력단 Pharmaceutical composition using secretomes from mesenchymal stem cells for treating rheumatoid arthritis and a method using the same
KR20180103800A (en) * 2016-08-26 2018-09-19 연세대학교 산학협력단 Pharmaceutical composition using secretomes from mesenchymal stem cells for treating rheumatoid arthritis and a method using the same
JP2020518260A (en) * 2017-05-04 2020-06-25 メゾブラスト・インターナショナル・エスアーエールエル Mesenchymal progenitor or stem cells with improved immunosuppressive activity
CN110753751A (en) * 2017-05-04 2020-02-04 迈索布拉斯特国际有限公司 Mesenchymal lineage precursor or stem cells with enhanced immunosuppression
KR20190023024A (en) * 2017-08-25 2019-03-07 연세대학교 산학협력단 Pharmaceutical composition using secretomes from mesenchymal stem cells for preventing or treating lupus
WO2019039922A1 (en) * 2017-08-25 2019-02-28 연세대학교 산학협력단 Pharmacological composition for prevention or treatment of lupus, comprising mesenchymal stem cell-derived secretome
WO2019190246A1 (en) 2018-03-29 2019-10-03 아키소스템바이오스트래티지스(주) Method for isolating stem cells from human umbilical cord
WO2019240507A1 (en) * 2018-06-15 2019-12-19 코아스템(주) Biomarker for predicting effectiveness for autoimmune disease treatment, diagnostic kit, and use for treatment
KR20190142129A (en) * 2018-06-15 2019-12-26 코아스템(주) Biomarker for treatment of autoimmune diseases, diagnostic kit and the therapeutic use thereof
JP2021527835A (en) * 2018-06-15 2021-10-14 コアステム カンパニー リミテッドCorestem Co.,Ltd. Biomarkers, diagnostic kits and therapeutic uses for predicting efficacy in the treatment of autoimmune diseases
CN117159589A (en) * 2023-11-01 2023-12-05 北京国卫生物科技有限公司 Stem cell treatment method for treating systemic lupus erythematosus
CN117159589B (en) * 2023-11-01 2024-02-13 北京国卫生物科技有限公司 Stem cell treatment method for treating systemic lupus erythematosus

Also Published As

Publication number Publication date
WO2016017863A1 (en) 2016-02-04

Similar Documents

Publication Publication Date Title
KR20150016117A (en) Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow
Wu et al. Donor age negatively affects the immunoregulatory properties of both adipose and bone marrow derived mesenchymal stem cells
Machado et al. Immunological characteristics of mesenchymal stem cells
KR102084974B1 (en) Pharmaceutical Compositions for Preventing or Treating Autoimmune Diseases Comprising Mesenchymal Stem Cells Derived from Human Bone-Marrow
Jones et al. Immunosuppression by mesenchymal stromal cells: from culture to clinic
AU2008287063B2 (en) Method of treating autoimmune disease with mesenchymal stem cells
Gazdic et al. Crosstalk between mesenchymal stem cells and T regulatory cells is crucially important for the attenuation of acute liver injury
JP6764912B2 (en) Cell population with immunomodulatory activity, its preparation method and use
US20080206208A1 (en) Extramedullary adipose tissue cells and use thereof for regenerating hematopoietic and muscular tissues
KR101507174B1 (en) Immune privileged and modulatory progenitor cells
JP6592551B2 (en) Cell population having immunomodulating activity, preparation method thereof, and use thereof
Ji et al. Mesenchymal stem cell transplantation inhibits abnormal activation of Akt/GSK3β signaling pathway in T cells from systemic lupus erythematosus mice
Van Vlasselaer et al. Interleukin-10 inhibits the osteogenic activity of mouse bone marrow
Ghobadinezhad et al. The emerging role of regulatory cell-based therapy in autoimmune disease
KR102246067B1 (en) Composition for promoting cell engraftment comprising stem cell having enhanced efficacy
Chen et al. CXCR4-transduced mesenchymal stem cells protect mice against graft-versus-host disease
Tecchio et al. Uncovering the multifaceted roles played by neutrophils in allogeneic hematopoietic stem cell transplantation
CN108699524B (en) Pharmaceutical composition for preventing or treating T cell mediated diseases
Mittal et al. Mesenchymal stem cells augment regulatory T cell function via CD80-mediated interactions and promote allograft survival
Siepe et al. Human neonatal thymus–derived mesenchymal stromal cells: characterization, differentiation, and immunomodulatory properties
CN113423411A (en) Tolerance vaccine treatment with carbodiimide induces graft tolerance
KR20150042755A (en) Composition for preventing or treating autoimmune diseases or transplant rejection comprising human fetal cartilage-derived cells
Foyle Characterisation of human dental pulp stem cells in an animal model of multiple sclerosis
CN117835994A (en) Methods and compositions for treating liver disease
JP2024517954A (en) Methods and compositions for treating liver disease

Legal Events

Date Code Title Description
A201 Request for examination
E902 Notification of reason for refusal
A107 Divisional application of patent
E601 Decision to refuse application