WO2013162330A1 - Chimeric mesenchymal stem cell population and preparation method therefor, and method for producing parathyroid hormone using tonsil-derived stem cells - Google Patents

Chimeric mesenchymal stem cell population and preparation method therefor, and method for producing parathyroid hormone using tonsil-derived stem cells Download PDF

Info

Publication number
WO2013162330A1
WO2013162330A1 PCT/KR2013/003644 KR2013003644W WO2013162330A1 WO 2013162330 A1 WO2013162330 A1 WO 2013162330A1 KR 2013003644 W KR2013003644 W KR 2013003644W WO 2013162330 A1 WO2013162330 A1 WO 2013162330A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
mesenchymal stem
chimeric
stem cell
parathyroid
Prior art date
Application number
PCT/KR2013/003644
Other languages
French (fr)
Korean (ko)
Inventor
김한수
우소연
유경하
조경아
조인호
박윤신
Original Assignee
이화여자대학교 산학협력단
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from KR20120044970A external-priority patent/KR101508413B1/en
Priority claimed from KR1020130045033A external-priority patent/KR101520531B1/en
Application filed by 이화여자대학교 산학협력단 filed Critical 이화여자대학교 산학협력단
Publication of WO2013162330A1 publication Critical patent/WO2013162330A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources
    • 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

Definitions

  • the present invention is a method for producing a chimeric mesenchymal stem cell group comprising the step of mixed culture of tonsil-derived mesenchymal stem cells obtained from a separate donor, chimeric mesenchymal stem cell group prepared by the method, parathyroid gland from tonsil stem cells A method for inducing differentiation of tissue cells and a method for producing parathyroid hormone from parathyroid tissue cells obtained by the method.
  • human tonsils are a type of lymphatic epithelial tissue that exists in the oropharyngeal nasopharynx of the human body, and performs its immune function until puberty and then gradually degenerates, causing the tonsils to become abnormally enlarged (adenotonsillar hyperplasia). Infections such as tonsillitis and tonsillitis can be removed by tonsillectomy.
  • Parathyroid hormone is a hormone composed of 84 amino acids secreted from the parathyroid gland, and is a major hormone that controls calcium concentration in vivo, and can be used as a therapeutic agent for parathyroid hypoplasia and osteoporosis. Increasingly, however, no method has yet been developed to effectively produce parathyroid hormone.
  • the present inventors have made diligent efforts to improve the utility of stem cells derived from tonsils, resulting in tonsil tissues obtained from a single donor even if mixed cultures of stem cells derived from different donors are cultured.
  • the stem cell group can be used for stem cell banks, and also that parathyroid tissue cells can be differentiated from tonsil stem cells, thereby confirming that parathyroid hormone is normally produced and secreted, thereby completing the present invention.
  • One object of the present invention is to provide a method for producing a chimeric mesenchymal stem cell population comprising a mixed culture of tonsil-derived mesenchymal stem cells obtained from separate donors.
  • Another object of the present invention is to provide a chimeric mesenchymal stem cell group produced by the above method.
  • Still another object of the present invention is to provide a method for producing a chimeric mesenchymal stem cell group having improved proliferative capacity, including freezing and thawing the chimeric mesenchymal stem cell group.
  • Still another object of the present invention is to provide a chimeric mesenchymal stem cell group having improved proliferative capacity prepared by the above method.
  • Still another object of the present invention is to provide a method for differentiating the chimeric mesenchymal stem cell population.
  • Still another object of the present invention is to provide a cell therapeutic agent comprising stem cells isolated from the chimeric mesenchymal stem cell group or cells differentiated therefrom as an active ingredient.
  • Another object of the present invention is to provide a method for differentiating parathyroid tissue cells from tonsil stem cells.
  • Still another object of the present invention is to provide a method for producing parathyroid hormone using tonsil stem cells.
  • Still another object of the present invention is to provide parathyroid tissue cells differentiated from tonsil stem cells.
  • Still another object of the present invention is to provide a method for treating parathyroid dysfunction or osteoporosis, comprising administering a cell therapy comprising parathyroid tissue cells to a subject suspected of hypothyroidism or osteoporosis.
  • the chimeric mesenchymal stem cell group of the present invention can be produced not only from the tonsil tissue discarded, but also from a separate tonsil tissue, and thus can be produced economically, and since it shows the same stem cell activity even after freezing and thawing, stem cells It can be used as a cell source for banks.
  • the method of differentiating parathyroid tissue cells from tonsill stem cells of the present invention can effectively differentiate parathyroid tissue cells in a short time by using tonsil tissue discarded after tonsillectomy, the parathyroid tissue cells obtained therefrom are present in the body It can produce parathyroid hormone in the same way as parathyroid tissue cells and regulate the secretion of parathyroid hormone according to the calcium concentration outside the cell, so it can be effectively used for the treatment of diseases requiring parathyroid hormone including hypothyroidism and osteoporosis.
  • a large amount of parathyroid hormone is secreted out of the cells so that parathyroid hormone can be easily obtained.
  • T-MSCs express specific surface antigens.
  • T-MSCs can differentiate into mesodermal and endoderm cells.
  • Figure 5 is a diagram illustrating the process of separating the amygdala stem cells from tonsils obtained by tonsillectomy.
  • FIG. 6 is a view showing the change in cell shape according to differentiation period from tonsil derived cells.
  • FIG. 7 is a view showing the results of the PTH concentration irradiation secreted into the cell culture medium during the differentiation period.
  • FIG. 8 is a view showing the expression pattern of PTH protein according to the parathyroid differentiation period of tonsil cells.
  • Figure 9 shows the distribution of PTH in the intracellular matrix during differentiation into parathyroid tissue.
  • FIG. 10 is a diagram showing the expression and distribution of CHGA, a PTH secretion related protein.
  • 11 is a diagram showing the expression distribution of cells of PTH and CHGA.
  • FIG. 12 is a diagram showing the regulation of PTH secretion according to the extracellular calcium concentration change.
  • FIG. 13 is a view showing the change in the cell shape of tonsil stem cells according to the extracellular calcium concentration change.
  • FIG. 14 is a view showing the change in the expression of calcium-sensitive receptors in tonsil stem cells according to the extracellular calcium concentration changes.
  • 15 is a diagram showing the cell microstructure of tonsil stem cells differentiated into parathyroid tissue.
  • 16 is a view showing the bone formation capacity of PTH produced and secreted in tonsil stem cells.
  • the present invention is a method for producing a chimeric mesenchymal stem cell population comprising the step of mixed culture of tonsil-derived mesenchymal stem cells obtained from a separate donor Or it provides a method for mixed culture of tonsil-derived mesenchymal stem cells.
  • the method for producing a chimeric mesenchymal stem cell group of the present invention or a method for mixed culture of mesenchymal stem-derived mesenchymal stem cells comprises the steps of: (i) obtaining mononuclear cells from each tonsil tissue isolated from a separate donor; (ii) culturing the obtained mononuclear cells to select mesenchymal stem cells; And, (iii) mixing and culturing the selected mesenchymal stem cells.
  • the term "tonsil” or “tonsil” is located at the back of the neck and the back of the nose, primarily to protect the body from substances such as bacteria that invade from the outside and act as lymphatic epithelial immune tissue. Means the organization to perform;
  • the amygdala includes a pharyngeal tonsil, ear pharyngeal tonsil, palate tonsil, tongue tonsil, and the like.
  • the amygdala is used as a source tissue for providing tonsil-derived mesenchymal stem cells.
  • mesenchymal stem cells refers to undifferentiated stem cells that can be differentiated into bone, cartilage, fat, myeloid epilepsy, muscle, nerves, and the like. But also in cord blood, peripheral blood, and other tissues.
  • the mesenchymal stem cells are not particularly limited thereto, but may be cells that can be mixed and cultured with each mesenchymal stem cell derived from a person's tonsil and derived from different individuals, and are adipocytes, bone Not only can they differentiate into mesodermal tissue cells such as cells, chondrocytes, but also endoderm tissue cells such as parathyroid tissue cells.
  • stem cell refers to a cell having self-replicating ability and capable of differentiating into two or more new cells, and includes totipotent stem cells and pluripotent stem cells. And multipotent (pluripotent) stem cells.
  • Tototent stem cells are pluripotent cells that can develop into a complete individual. Cells up to 8-cells after fertilization of eggs and sperm have this property and isolate the cells. When transplanted into the uterus means a cell that can develop into a complete individual.
  • Pluripotent stem cells are cells that can develop into various cells and tissues derived from ectoderm, mesoderm, and endodermal layer, and are located inside the blastocyst appearing 4-5 days after fertilization. It is derived from the inner cell mass, which is called embryonic stem cells, and refers to cells that differentiate into various other tissue cells but do not form new life.
  • fat cell refers to a major cell constituting adipose tissue that stores energy in the form of fat.
  • the adipocytes may exist in two forms: white fat cells comprising large fat droplets surrounded by a cytoplasmic layer and polygonal brown fat cells comprising a significant amount of cytoplasm with evenly dispersed fat droplets.
  • chondrocytes refers to the only cells found in the cartilage, and can produce and maintain a cartilage substrate mainly composed of collagen and proteoglycans.
  • the composition of chondrocytes in cartilage depends on the type of cartilage and the location of the tissue.
  • bone cell refers to a cell that is differentiated from osteoblasts in developing bone, which is present inside the bone cavity contained in bone tissue, has a flat ellipsoidal form of the same type as the bone cavity, and is extremely It has many processes and is involved in mineral metabolism of surrounding bone tissue.
  • parathyroid tissue cells refers to cells constituting the parathyroid gland, an endocrine organ attached to the thyroid gland, secretes parathormone and regulates metabolism of calcium and phosphorus in body fluids. In the present invention, as long as the parathyroid hormone is secreted, it may be included in parathyroid tissue cells.
  • a method for obtaining mononuclear cells from tonsil tissue is not particularly limited thereto, and may be obtained by grinding the tonsil tissue and filtering the same. .
  • the method for selecting mesenchymal stem cells by culturing mononuclear cells is not particularly limited thereto, and may be performed by selecting cells attached to the bottom of the culture vessel by culturing using a stem cell culture medium.
  • the stem cell culture medium is not particularly limited to this, but DMEM-HG (Dulbecco's modified Eagle's medium-High Glucose) medium, including FBS (Invtrogen) and antibiotics, Keratinocyte-SFM, RPMI-1640 may be used,
  • the antibiotic is not particularly limited thereto, but may be streptomycin, ampicillin, or the like.
  • mixed culture refers to a method of mixing and culturing mesenchymal stem cells isolated from tonsils obtained from different separate donors. Even in the case of tissue cells of the same character, cells separated from different donors have different characteristics of the cells, and thus do not exhibit uniform characteristics. However, tonsil-derived mesenchymal stem cells of the present invention exhibit the same characteristics as stem cells obtained from a single donor even when cultured by mixing stem cells obtained from different donors.
  • the present invention provides a chimeric mesenchymal stem cell population prepared by the above method.
  • Said chimeric mesenchymal stem cell population comprises (i) mesenchymal stem cells isolated from separate tonsil tissue obtained from separate donors; (ii) exhibit the same cellular activity as tonsil-derived mesenchymal stem cells obtained from a single donor; (iii) the expression of CD14, CD34 and CD45 is reduced; (iv) the expression of CD73, CD95 and CD105 is increased; (v) negative for immunorelevant expressing antibodies; (vi) can differentiate into mesodermal cells; And (vii) differentiate into endoderm cells.
  • chimeric mesenchymal stem cell group refers to mesenchymal stem cells obtained by mixed culture of tonsil-derived mesenchymal stem cells obtained from separate donors, wherein the chimeric mesenchymal stem cell groups are different donors. Each mesenchymal stem cell isolated from is included, but exhibits the same level of cellular characteristics as the mesenchymal stem cells isolated from a single donor.
  • the chimeric mesenchymal stem cell population may exhibit the phenotype of a typical stem cell immunophenotype of T-MSC. That is, the expression of CD14, CD34 and CD45 is weak while the expression of CD73, CD95 and CD105 is increased (FIG.
  • FIG. 2A the immune-related expressing antibodies
  • HLA-DR the immune-related expressing antibodies
  • CD40, CD80, CD86 the immune-related expressing antibodies
  • FIG. 2B the immune-related expressing antibodies
  • they can be differentiated into mesodermal cells, adipocytes, bone cells and chondrocytes (FIG. 3A), and also into endoderm cells, parathyroid tissue cells (FIG. 3B).
  • chimera when mixed cultured tonsil-derived mesenchymal stem cells obtained from separate donors, chimera was formed to form one mesenchymal stem cell (Fig. 4b). ), Mesenchymal stem cells obtained from a mixed culture of tonsil-derived mesenchymal stem cells obtained from a single donor and tonsil-derived mesenchymal stem cells obtained from a separate donor were compared in terms of CFU-F formation ability and proliferative capacity. The results confirmed similar levels (FIG. 1A).
  • the present invention provides a method for producing a chimeric mesenchymal stem cell group having improved proliferative capacity, including freezing and thawing the chimeric mesenchymal stem cell group.
  • the chimeric mesenchymal stem cell group having improved proliferative capacity may exhibit (i) differentiation capacity equivalent to that of the chimeric mesenchymal stem cell group that has not undergone freezing and thawing; And, (ii) an improved level of proliferative capacity than the chimeric mesenchymal stem cell population that did not perform freezing and thawing.
  • freeze of the present invention refers to the action of freezing using a cooling medium such as liquid nitrogen in order to preserve the tonsil-derived mesenchymal stem cells of the present invention.
  • a solution containing a cryoprotectant may be used to prevent cell damage during freezing.
  • freeze protection agent refers to a substance which is added to a liquid medium for the purpose of alleviating the East Sea when biological cells are stored alive in a frozen state, and the freeze protection agent is not particularly limited thereto. However, glycerol, sugar, glucose and the like can be used.
  • thawing in order to utilize the frozen tonsil-derived mesenchymal stem cells of the present invention, means the action to increase the temperature to room temperature so that the cells exhibit normal physiological activity.
  • enhanced proliferative capacity of the present invention refers to a phenomenon in which the rate of proliferation of tonsil-derived mesenchymal stem cells provided by the present invention increases after applying a freezing and thawing process.
  • the mesenchymal stem cells obtained by mixing and cultured tonsil-derived mesenchymal stem cells obtained from a single donor and tonsil-derived mesenchymal stem cells obtained from a separate donor are frozen and thawed.
  • Mixed cultured mesenchymal stem cells were confirmed to exhibit a relatively high level of proliferative capacity (FIG. 1B).
  • the tonsil-derived mesenchymal stem cells of the present invention can be differentiated into mesodermal cells and endoderm cells, and show the same differentiation and proliferative capacity as stem cells derived from a single donor even when mixed with other donor stem cells. In addition, even after freezing and thawing, it is possible to maintain the differentiation capacity and proliferative capacity of the first stem cells as they are, and thus can be utilized as a cell source essential for forming a stem cell bank.
  • the present invention provides a method for differentiating the chimeric mesenchymal stem cell population into various cells.
  • the method of differentiating the chimeric mesenchymal stem cell group into various cells is not particularly limited, but the chimeric mesenchymal stem cell group is differentiated into chondrocytes containing ascorbic acid, L-proline, TGF-3, BMP-6 and ITS.
  • chimeric mesenchymal stem cell group was cultured in culture medium containing ascorbic acid 2-phosphate, dexamethasone and ⁇ -glycerophosphate to differentiate into bone cells, or the chimeric mesenchyme Stem cell populations are cultured in culture medium containing 3-isobutyl-1-methyl-gentine, dexamethasone, indomethacin and insulin to differentiate into adipocytes, or the chimeric mesenchymal stem cell population is activated by activin A (activin A). ) And Shh (sonic hedgehog) can be cultured in a culture medium containing the parathyroid tissue cells.
  • the term "differentiation" of the present invention generally refers to a phenomenon in which a relatively simple system is separated into two or more qualitatively different sub systems, specifically, during cell division and growth. It refers to a phenomenon in which structures and functions are specialized to each other, that is, a phenomenon in which a form or a function of a cell or tissue of an organism changes to perform a task given to each. For example, qualitatively between parts of a biological system that were initially nearly homogeneous, such as head or torso distinctions between eggs that were initially homogenous in population development, or cells such as myocytes or neurons. Phosphorus difference arises, or as a result, the phenomenon divided into sub division or sub-system which can be distinguished qualitatively is mentioned.
  • the method for measuring the degree of differentiation of the chimeric mesenchymal stem cell group differentiated by the above method is not particularly limited to this, using flow cytometry, immunocytochemical method, PCR and gene expression profile which are well known in the art.
  • -PCR Oil red O staining, Safranin O staining, Type II collagen immunohistochemical staining, alkaline phosphate staining and Alizarin S staining can be used.
  • tonsil mesenchymal stem cells (tonsillar mesenchymal stem cells, T-MSC) was confirmed that can be differentiated into adipocytes, bone cells and chondrocytes that are mesodermal cells (Fig. 3a) ), As well as parathyroid tissue cells that are endoderm cells can be differentiated (Fig. 3b).
  • the present invention provides a cell therapeutic agent comprising stem cells isolated from the chimeric mesenchymal stem cell group, or cells differentiated from the chimeric mesenchymal stem cell group as an active ingredient.
  • the differentiated cells are not particularly limited thereto, but may preferably be all cells that can be used for cell therapy, more preferably may be mesodermal cells or endoderm cells, most preferably fat Cells, chondrocytes, osteocytes, parathyroid tissue cells, and the like.
  • cellular therapeutic agent refers to a medicinal product (US FDA regulation) used for the purpose of treatment, diagnosis, and prevention of cells and tissues prepared through isolation, culture, and special manipulation from an individual.
  • drugs used for the purpose of treatment, diagnosis, and prevention through a series of actions, such as proliferating and screening live autologous, allogeneic, or heterologous cells in vitro or by altering the biological characteristics of the cells in order to restore tissue function. Means.
  • the chimeric mesenchymal stem cell population of the present invention is a variety of therapeutic protocols in which the tissues or organs of the body are fortified, treated or replaced by engraftment, transplantation or infusion of a desired cell population, for example, a stem cell or a derived cell population.
  • a desired cell population for example, a stem cell or a derived cell population.
  • the chimeric mesenchymal stem cell population of the present invention can replace or enhance existing tissue, resulting in new or changed tissue, or bind to biological tissue or structure.
  • the chimeric mesenchymal stem cell group of the present invention can be used for strengthening, treating or replacing human cartilage, tendons, ligaments, parathyroid tissues and the like.
  • the invention is a parathyroid tissue cell from tonsils, comprising culturing tonsils in a medium comprising Activin A and Sonic hedgehog (Shh) It provides a way to differentiate.
  • parathyroid tissue when inducing parathyroid tissue from embryonic stem cells or adult stem cells derived from the thymus, it took longer than 6 months to induce parathyroid tissue.
  • the present invention first identified a method capable of inducing differentiation of parathyroid tissue in a short period of time from amygdala stem cells, which have never been considered as candidates for differentiation into parathyroid tissue.
  • the terms "tonsil”, “stem cell”, “parathyroid tissue cells” are as described above, in the present invention, the tonsil is used as a source tissue for providing tonsil-derived mesenchymal stem cells.
  • tonsils means mesenchymal stem cells (T-MSCs) derived from tonsils.
  • Parathyroid tissue cell differentiation method of the present invention comprises culturing the amygdala stem cells in a medium containing activin A and sonic hedgehog (Shh) at a concentration of 50 ng / ml ⁇ 300 ng / ml, respectively,
  • the culture period of the cells is preferably 4 days or more, more preferably 7 days or more.
  • the amygdala stem cells may be collected from tonsil tissue isolated from a patient who has undergone tonsillectomy, but are not limited thereto.
  • the collected tonsil is washed with buffer, and then treated with enzyme mixture to decompose the connective tissue.
  • the suspended tissue is centrifuged and the cell mixture in the cut tissue is ficol Immunohistochemical method using cell surface markers among the cells showing adhesion to the bottom of the culture plate after culturing only the precipitated part of the lower layer by cell fractionation and centrifugation using Tonsillar stem cells can be isolated using
  • activin A which is used as a differentiation inducing agent, is a member of the TGF-beta superfamily, first known as a protein that promotes FSH release, but it is known as mesoderm induction, neuronal differentiation, bone remodeling, hematopoiesis, It is known to have a wide range of biological activities.
  • Sonic hedgehog proteins are one of three proteins in a family of mammalian signaling pathways called hedgehogs, which play a key role in regulating vertebrate organogenesis such as finger growth in the extremities and brain organization.
  • Sonic hedgehog is a type of morphogen that has different effects on developing embryonic cells depending on its concentration, and is known to control cell division of adult stem cells and to induce some cancers. .
  • activin A and sonic hedgehog can be purchased by using any commercially available activin A and sonic hedgehog.
  • the concentrations of activin A and sonic hedgehog are less than 50 ng / ml, the period required for differentiation of parathyroid tissue cells from amygdala stem cells may be extended to several months or the differentiation efficiency may be inferior. If the concentration of the differentiation induction agent exceeds 300 ng / ml, there is no improvement in the efficiency of differentiation induction, it is inefficient to use such a high concentration of chemicals.
  • the concentrations of Activin A and Sonic Hedgehog are each 80 ng / ml to 200 ng / ml, most preferably 100 ng / ml each.
  • the parathyroid tissue from the amygdala stem cells may be shortened only by 4 days of culture. Can differentiate cells.
  • the method of the present invention to induce differentiation of parathyroid tissue cells by culturing tonsil stem cells in a medium containing activin A and sonic hedgehog at a concentration of 100 ng / ml, respectively, for 4 to 25 days. If the culture period exceeds 4 days, the medium can be replaced with fresh medium containing activin A and sonic hedgehog every 4-5 days.
  • the medium used to induce differentiation may be used a conventional medium that can be used for the culture of stem cells, for example, DMEM (Dulbecco's modified Eagle medium), Keratinocyte-SFM (Keratinocyte serum free medium), RPMI-1640, and the like, and preferably, DMEM medium may be used.
  • the stem cell medium may be supplemented with an additive.
  • an additive may contain neutral buffers (such as phosphates and / or high concentrations of bicarbonate) and protein nutrients (such as serum, such as FBS, serum substitutes, albumin, or essential and non-essential amino acids, such as glutamine) in isotonic solutions.
  • neutral buffers such as phosphates and / or high concentrations of bicarbonate
  • protein nutrients such as serum, such as FBS, serum substitutes, albumin, or essential and non-essential amino acids, such as glutamine
  • lipids fatty acids, cholesterol, HDL or LDL extracts of serum
  • other components found in most preservative media of this kind such as insulin or transferrin, nucleosides or nucleotides, pyruvate salts, any ionized form or salt
  • Sugar sources such as glucose, selenium, glucocorticoids such as hydrocortisone and / or reducing agents such as ⁇ -mercaptoethanol.
  • the parathyroid hormone produced by parathyroid tissue cells after 4 days of culture as a result of incubating the amygdala stem cells in a medium containing activin A and sonic hedgehog at a concentration of 100 ng / ml, respectively.
  • the production of was significantly increased, and the intracellular microstructure was confirmed using electron microscopy.
  • the differentiated cells were differentiated into hormone secretion tissues because of the presence of a basic granular microstructure and a granular and granular precursor structure. could be confirmed.
  • the present invention provides a method for producing parathyroid hormone using tonsilocytes.
  • the present invention comprises the steps of i) culturing amygdala stem cells in a medium containing Activin A and Sonic hedgehog (Shh) to differentiate into parathyroid tissue cells, and ii) i) step It provides a method for producing parathyroid hormone using tonsils, comprising the step of separating the parathyroid hormone from the culture solution or cell lysate obtained in the.
  • parathyroid tissue cells differentiated from tonsil stem cells produce and secrete parathyroid hormone like normal parathyroid tissue cells. Therefore, parathyroid hormone can be obtained by separating parathyroid hormone from culture or cell lysate of parathyroid tissue cells differentiated from tonsil stem cells.
  • tonsil stem cells, activin A, and sonic hedgehog are as described above.
  • the step of culturing the amygdala stem cells in a medium containing activin A and sonic hedgehog to differentiate into parathyroid tissue cells is the same as described above.
  • Separation of parathyroid hormone from the culture or cell lysate may be carried out using a general method that can be used for the separation and purification of proteins, for example, affinity chromatography, ion exchange chromatography, coprecipitation, etc.
  • the method may be used, but the present invention is not limited thereto and may be appropriately selected by a person skilled in the art.
  • parathyroid hormone is produced in the parathyroid tissue cells and then present in the cells in the form of secretory granules, depending on the extracellular calcium concentration is determined whether the secretion outside the cell.
  • the method of the present invention in order to facilitate the secretion of parathyroid hormone produced by parathyroid tissue cells derived from tonsil stem cells to facilitate the separation of parathyroid hormone, the method of the present invention is the step i) Thereafter, adjusting the calcium concentration of the culture medium may further include secreting parathyroid hormone out of the cells.
  • the method may further comprise adjusting the calcium concentration of the culture medium to less than 1.5 mM.
  • the calcium concentration of the culture medium for promoting secretion of parathyroid hormone is preferably less than 1.0 mM, more preferably less than 0.5 mM, and most preferably less than 0.1 mM.
  • the parathyroid hormone may be lysed to separate the parathyroid hormone from the lysate without secreting the parathyroid hormone out of the cell.
  • Parathyroid hormone production method of the present invention by using tonsil stem cells that can be obtained from tonsil tissue discarded after tonsillectomy, to facilitate the production of parathyroid hormone that can be useful for the treatment of parathyroid hypoplasia or osteoporosis, etc.
  • tonsil stem cells that can be obtained from tonsil tissue discarded after tonsillectomy, to facilitate the production of parathyroid hormone that can be useful for the treatment of parathyroid hypoplasia or osteoporosis, etc.
  • it is possible to easily obtain a large amount of hormones by promoting the secretion of parathyroid hormone through the regulation of calcium concentration outside the cell.
  • the amygdala stem cells are cultured in a medium containing activin A and sonic hedgehog to induce differentiation of parathyroid tissue and confirm the expression patterns of parathyroid hormone in each period. It was confirmed that the expression of parathyroid hormone was significantly increased from the point of time elapsed, and that the expression level was significantly increased after 7 days after induction of differentiation, thereby making the parathyroid tissue differentiated from tonsil stem cells by the method of the present invention. It was confirmed that parathyroid hormone could be produced from the cells.
  • the cumulative secretion of parathyroid hormone increased as the differentiation-inducing period elapsed, and it was confirmed that a large amount of parathyroid hormone can be obtained by continuously culturing parathyroid tissue differentiated from tonsil stem cells.
  • the external calcium concentration is lower than the normal concentration (1.5 mM)
  • it is confirmed that the parathyroid hormone is secreted to the outside of the cell it is also possible to control the secretion of parathyroid hormone by controlling the calcium concentration of the culture medium when inducing differentiation of parathyroid tissue cells It was.
  • the present invention provides parathyroid tissue cells differentiated from tonsil stem cells.
  • parathyroid tissue cells of the present invention can be obtained by the parathyroid tissue cell differentiation method comprising culturing tonsils in the medium containing the amygdala stem cells as described above, activin A and Sonic hedgehog, preferably
  • the amygdala stem cells can be obtained by culturing at least 4 days in a medium containing activin A and sonic hedgehog at a concentration of 50 ng / ml to 300 ng / ml, respectively, which is most preferred for obtaining parathyroid tissue cells.
  • the concentrations of activin A and sonic hedgehog are 100 ng / ml each.
  • Parathyroid tissue cells obtained by differentiation from amygdala stem cells by the differentiation method of the present invention as described above can produce parathyroid hormone, and can control the secretion of parathyroid hormone in response to changes in calcium concentration outside the cell, If the calcium concentration is lower than the normal concentration (1.5 mM) to secrete parathyroid hormone outside the cell, if the calcium concentration outside the cell is higher than the normal concentration can suppress the secretion of parathyroid hormone.
  • the parathyroid tissue cells of the present invention can function as parathyroid tissue cells, like parathyroid tissue cells present in the body, and are treatable by diseases or parathyroid hormone due to impaired function of parathyroid tissue or lack of parathyroid hormone. It can be usefully used for the treatment of known osteoporosis and the like.
  • the present invention provides a cell therapy agent of hypothyroidism or osteoporosis comprising the parathyroid tissue cells of the present invention.
  • cell therapeutic agent is as described above.
  • the cell therapy agent comprising the parathyroid tissue cells of the present invention can be used include, for example, hypothyroidism or osteoporosis.
  • hypothyroidism is a disease caused by hypocalcemia caused by lowered calcium in the blood due to parathyroid hormone dysfunction.
  • the only way to treat it is to take excess calcium, but to take excess calcium instead of parathyroid hormone.
  • osteoporosis refers to a condition in which bone fractures occur due to a decrease in bone quantity due to a decrease in the amount of bone and qualitative changes
  • an agent that mainly suppresses bone resorption is mainly used.
  • the use of parathyroid hormone has been suggested as a direct stimulator of bone formation.
  • the use of parathyroid hormone can only be used for a short period of time (up to two years, US FDA), expensive, subcutaneous injections such as daily insulin injections, and rapid bone loss after the end of treatment. It has been reported. Therefore, if you use a cell therapy containing parathyroid tissue that can produce parathyroid hormone rather than administration of parathyroid hormone and can regulate hormone secretion according to the extracellular calcium concentration, osteoporosis can be treated without side effects. .
  • treatment in the present invention means any action that improves or benefits the condition of the disease by administration of the cell therapy.
  • the cell therapy agent of the present invention contains 1.0 ⁇ 10 5 to 1.0 ⁇ 10 9 cells, preferably 1.0 ⁇ 10 6 to 1.0 ⁇ 10 8 cells, more preferably 1.0 ⁇ 10 7 cells per ml. can do.
  • the cell therapy of the invention can be used unfrozen or frozen for future use. If frozen, standard cryopreservatives (eg DMSO, glycerol, Epilife® cell freezing medium (Cascade Biologics)) can be added to the cell population prior to freezing.
  • standard cryopreservatives eg DMSO, glycerol, Epilife® cell freezing medium (Cascade Biologics)
  • the cell therapy agent may be administered by formulating a unit dosage form pharmaceutical preparation suitable for administration in the body of a patient according to a conventional method in the pharmaceutical field, wherein the preparation is effectively administered by one or several administrations. Contains the amount.
  • Formulations suitable for this purpose include parenteral injectables such as injectable ampoules, injectables such as infusion bags, sprays such as aerosol preparations and the like.
  • the injection ampoule may be mixed with the injection solution immediately before use, and physiological saline, glucose, mannitol, and Ringer's solution may be used as the injection solution.
  • the infusion bag can also be made of polyvinyl chloride or polyethylene and can be used in Baxter, Becton Dickinson, Medcep, National Hospital Products or Terumo.
  • the injection bag of the yarn can be illustrated.
  • one or more pharmaceutically acceptable conventional inert carriers such as preservatives, analgesic agents, solubilizers or stabilizers in the case of injections, etc. It may further include a base, excipients, lubricants or preservatives.
  • the cell therapy or pharmaceutical preparation of the present invention thus prepared may be administered with other stem cells used for transplantation and other uses, or in the form of mixtures with such stem cells, using administration methods commonly used in the art.
  • administration methods commonly used in the art.
  • the administration can be both non-surgical administration using a catheter and surgical administration methods such as injection or transplantation after dissection of the disease site, but non-surgical administration using a catheter is more preferable.
  • parenteral administration for example, in addition to direct lesions, transplantation by intravascular injection, which is a general method of hematopoietic stem cell transplantation, is also possible according to a conventional method.
  • the daily dose of the stem cells may be administered 1.0 ⁇ 10 4 to 1.0 ⁇ 10 10 cells / kg body weight, preferably 1.0 ⁇ 10 5 to 1.0 ⁇ 10 9 cells / kg body weight divided into one or several times .
  • the actual dosage of the active ingredient should be determined in light of several relevant factors such as the disease to be treated, the severity of the disease, the route of administration, the patient's weight, age and gender, and therefore, the dosage may be It is not intended to limit the scope of the present invention in terms of aspects.
  • the present invention provides a method for treating parathyroid function-related diseases or osteoporosis, comprising administering a cell therapy comprising parathyroid tissue cells to an individual suspected of hypothyroidism or osteoporosis. Provide a method.
  • Administration of the cell therapy of the present invention is applicable to any animal, and animals include humans and primates, as well as domestic animals such as cattle, pigs, sheep, horses, dogs, mice, rats, and cats.
  • the term "administration" means introducing the cell therapy of the invention into a patient in any suitable manner and includes the transplantation of differentiated cells. Routes of administration of the cell therapy products of the invention can be administered via a variety of routes as long as they can reach the desired tissue.
  • Example 1 Obtaining Amygdala Mesenchymal Stem Cells
  • 5 is a method for isolating and culturing stem cells from the obtained tonsil tissue.
  • the collected tonsils were washed with physiological saline, and the enzyme mixture 210 U / mL collagenase type I (Invitrogen) and 10 ⁇ g / mL DNAse (Sigma-Aldrich, St. Louis, MO). ) was mixed in the culture medium RPMI-1640 (Roswell Park Memorial Institute medium 1640, Invitrogen Corporation, Carlsbad, Calif.) For 30 minutes at 37 °C conditions connective tissue in the amygdala. The suspended tissue was centrifuged and filtered to obtain cells. The obtained cells were washed twice with RPMI 1640/20% NHS (normal human serum) (PAA Laboratories, GmbH, Paching, Austria) and once with RPMI 1640/10% NHS.
  • RPMI-1640 normal human serum
  • Washed cells were subjected to Ficoll-Paque (GE Healthcare, Little Chalfont, Buckinghamshire, UK) gradient centrifugation to obtain mononuclear cells.
  • Dulbecco's modified Eagle's medium-High Glucose (DMEM-HG) containing 10 8 fetal mononuclear cells (Fetal bovine serum, FBS, Invitrogen), 100 ⁇ g / ml streptomycin and 100 U / ml ampicillin (Invitrogen) The medium was inoculated and incubated for 48 hours. After incubation, unattached cells are removed, and adherent cells, considered tonsillar mesenchymal stem cells (T-MSCs), are passaged to 3-5 generations over 4 weeks. Used.
  • T-MSCs tonsillar mesenchymal stem cells
  • T-MSCs incubated above were washed with PBS (pH 7.4), and suspended by adding cryodium (50% FBS, 40% DMEM and 10% DMSO) to the washed T-MSCs. Aliquots were included to contain 2 ⁇ 10 6 cells per vessel.
  • the suspension dispensed vessel was frozen at ⁇ 70 ° C. for 24 hours and stored in ⁇ 200 ° C. liquid nitrogen for 10 days (Liu Y, et al., Biotechnol. Prog., 26: 1635-1643, 2010).
  • the stored vessel was thawed in 37 ° C. water for 2 minutes using a known method, and the thawed cell suspension was centrifuged to obtain T-MSC.
  • cryopreserved-and-thawed cells which have undergone freezing and thawing, were cultured for 7 days and applied to the same method as described above.
  • FC cultures obtained from a single donor there was no difference between FC cultures obtained from a single donor and mixed FC cultures obtained from several donors in terms of proliferative capacity (FIG. 1B).
  • mixed CTC cultures obtained from several donors showed relatively high levels of proliferative capacity.
  • T-MSC Cell phenotype of T-MSC was analyzed by using FACSCalibur system (Becton Dickinson, Franklin Lakes, NJ) to confirm the expression of the surface antigens of the FC and CTC.
  • FC and CTC were incubated for 14 days and 7 days, respectively, and stained at 4 ° C. for 30 minutes using respective antibodies (2 ⁇ g / ml) capable of reacting with surface antigen and analyzed using the FACSCaliur system. .
  • the antibodies used are as follows; Hematopoietic cell markers (PE-CD14, PE-CD34 and PerCP-CD45), primitive cell markers (PE-CD73, PE-CD90, and PE-CD105), class II MHCs and auxiliaries Co-stimulatory molecules markers (PE-HLA-DR, FITC-CD40, PE-CD80 and FITC-CD86) and follicular dendritic cell markers (PE-CD11b, PE-CD21, PE -CD23, PE-CD35 and PE-CD54).
  • Hematopoietic cell markers PE-CD14, PE-CD34 and PerCP-CD45
  • primitive cell markers PE-CD73, PE-CD90, and PE-CD105
  • Co-stimulatory molecules markers PE-HLA-DR, FITC-CD40, PE-CD80 and FITC-CD86
  • follicular dendritic cell markers PE-CD11b, PE-CD21, PE -CD23,
  • an IgG1 or IgG2a (Becton Dickinson) isotype antibody, which is an antibody capable of performing nonspecific binding, was used as a control.
  • the immunophenotype of T-MSC showed a typical stem cell phenotype. That is, the expression of CD14, CD34 and CD45 was weak while the expression of CD73, CD95, CD105 was increased, indicating that T-MSC was different from hematopoietic stem cells (FIG. 2A). In addition, immune-related expressing antibodies (HLA-DR, CD40, CD80, CD86) were also negative (FIG. 2B).
  • T-MSC is a different cell from FDC.
  • T-MSCs were used to induce differentiation into mesodermal cells, adipocytes, bone cells and chondrocytes.
  • adipocyte culture Invtorgen
  • T-MSCs were inoculated into a 14 ml conical tube containing chondrocyte culture medium (Invtorgen) containing dexamethasone, ascorbic acid, L-proline, TGF-3, BMP-6 and ITS. Incubate for 3 weeks with replacement every 3-4 days. After the incubation was terminated, the cultured cells were harvested, and the harvested cells were fixed with 4% paraformaldehyde and embedded with paraffin to obtain paraffin blocks. The obtained paraffin block was cut to a thickness of 5 ⁇ m to obtain flakes. The obtained flakes were placed on a slide glass, and then paraffin was removed to fix cell sections on the slide glass.
  • chondrocyte culture medium Invtorgen
  • T-MSCs were induced to differentiate into parathyroid cell-like cells, which are endoderm cells.
  • T-MSC RPMI 1640 medium containing 5% FBS, activin A (activin A, 100 ng / ml, R & D System, Inc. Minneapolis) and sonic hedgehog (Shh) (100 ng / ml, R & D Systems) Inoculated and cultured for 3 weeks while replacing the culture every 4 days. After the incubation was completed, the expression of endoderm-specific protein was confirmed by reverse transcriptase chain reaction (RT-PCR) method.
  • RT-PCR reverse transcriptase chain reaction
  • GCM2 glial cells missing homolog 2
  • CaSR calcium sensing receptor
  • CCL21 specific for parathyroid tissue cells chemokine (CC motif) ligand 21
  • PTH parathyroid hormone
  • RT system Promega, Madison, Wis.
  • the sequence of the primer used is described in Table 1, the overall method was performed according to the user's manual of TRIZOL (Invitrogen).
  • FoxA2 forkhead box A2, SOX17: sex determining region Y (SRY) -box 17, CXCR4: chemokine (CXC motif) receptor 4, Eya1: eyes absent homolog 1, Six1: SIX homeobox 1, Pax1: paired box 1, BMP4: bone morphogenic protein 4, GCM2: glial cells missing homolog 2, CaSR: calcium sensing receptor, CCL21: chemokine (CC motif) ligand 21, PTH: parathyroid hormone, and GAPDH: glyceraldehyde-3-phosphate dehydrogenase
  • T-MSCs can be differentiated into parathyroid tissue cells, which are endoderm cells.
  • Human CD4 + T cells were obtained from the peripheral blood of healthy donors and PMA (Phorbol 12- myristate 13-acetate) (Sigma-Aldrich) was used as a mitosis promoter.
  • CD4 + T cells were labeled with carboxyfluorescein diacetate (succinimidyl ester) (CFSE).
  • CFSE carboxyfluorescein diacetate
  • the specific method is as follows: The cells were suspended in PBS containing 0.1% BSA pre-warmed at a concentration of 10 6 cells / ml, followed by the addition of 10 ⁇ M CFSE. Subsequently, the cells were incubated at 37 ° C. for 10 minutes, and the cells were left in 5 times ice-cold culture to terminate staining, followed by centrifugation to obtain CD4 + T cells labeled with CFSE.
  • the obtained CFSE-labeled CD4 + T cells (3 ⁇ 10 4 cells) were resuspended in culture and washed three times, and the washed cells were wells (10 4 cells) with or without cultured T-MSCs or not. The wells were inoculated. Then, PMA at a final concentration of 1 to 200 ng / ml was added to each well inoculated with the cells, and cultured for 3 days, and then the proliferation of CFSE + CD + T cells was measured by flow cytometry, and ModiFitLT The reduction of CFSE positive cells was analyzed using the software.
  • Each T-MSC from three independent donors was incubated for four weeks, either individually or mixed together. After the incubation was completed, chimerism was confirmed by extracting genomic DNA from each cultured cell and performing STR analysis by a known method (Kristt DM, et al., J. Biomol. Tech., 16: 380-391, 2005; Kim DW, et al., Blood, 103: 1941-1948, 2004). At this time, the extracted genomic DNA was amplified by PCR, STR analysis was performed using capillary electrophoresis (ABI 3130Xl Genetic analyzer, Carlsbad, CA), and the result analysis was performed using GeneMapper software.
  • chimeras (heterozygote) were formed at three sites of D8S1179, D3S1358 and D18S51 among the total 15 sites (FIG. 4B).
  • T-MSCs derived from amygdala of various origins can form chimeras in mixed cultures, and thus, T-MSCs exhibited similar and similar levels of cellular characteristics regardless of origin, resulting in mixed Through culture, it was found that one mesenchymal stem cell group could be formed.
  • T-MSCs For further study of the differentiation of amygdala stem cells into parathyroid tissue, differentiation of T-MSCs from single individuals into parathyroid cell-like cells was induced. Specifically, 5% of T-MSCs were induced. Inoculate DMEM medium containing FBS, activin A (activin A, 100 ng / ml, R & D System, Inc. Minneapolis) and sonic hedgehog (Shh, 100 ng / ml, R & D Systems), and incubate the culture every 4 days Incubate for 21-25 days with replacement.
  • activin A activin A
  • sonic hedgehog Shh, 100 ng / ml, R & D Systems
  • amygdala stem cells formed a multi-layer accumulating into several layers as shown in FIG.
  • PTH Whenever exchanged with cell culture medium containing differentiation inducer by differentiation period (0, 4, 7, 10, 15, 20 days), PTH is collected and secreted into the extracellular cells during differentiation into parathyroid tissue. It was used as a sample for analyzing the concentration of.
  • Cell cultures collected at each time were filtered through a 0.45 ⁇ m syringe filter to remove cell suspensions and cell debris, and the filtered cell cultures were freeze-dried and concentrated using a freeze dryer (Operon).
  • Lyophilized powder samples were resuspended in phosphate buffered saline (pH 7.4) and analyzed for PTH concentration by immunochemistry. That is, the concentration of PTH released by the ELCIA method using the binding force with the antibody to the PTH protein was investigated.
  • the cells treated with the differentiation inducing liquid secreted statistically significant amount of PTH into the cell culture fluid compared with the cells that did not differentiate.
  • the increase was significantly increased from 7 days to 10 days after the differentiation induction solution, and the amount was up to 40 pg / ml, which was 15-60 pg / ml of normal PTH concentration range in vivo. It was found to be included in the range (FIG. 7B).
  • T-MSCs were collected during differentiation, the cell surface was washed twice with PBS (Phosphate buffered saline, pH 7.4), and then RIPA buffer (25 mM Tris-HCl pH 7.6, 150 mM NaCl, 1% NP- 40, 1% sodium deoxycholate, 0.1% SDS) was used to extract the intracellular protein portion.
  • the extracted protein was quantified by BCA (bicinchoninic acid) method using albumin as a standard solution, and used for intracellular PTH protein expression.
  • SDS-PAGE 10-12% SDS (sodium deoxycholate) -polyacrylamide gel (SDS-PAGE) was prepared, and 30 ⁇ g of protein samples quantified at each stage of differentiation period were loaded on pre-prepared SDS-PAGE, followed by electrophoresis. My proteins were separated by their size. After electrophoresis, proteins of each size separated on the gel were transferred to the nitrocellulose membrane using an electric method.
  • the primary antibody-specific secondary antibody mouse-IgG-HRP (horse radish peroxidase) conjugate was further reacted (diluted at 1: 3000 ratio).
  • the response of the PTH antibody bound to the intracellular PTH protein antigen was examined by ECL (enhanced chemiluminescence) method. That is, the expression level of PTH, CaSR, CHGA, and Cyr61 present in the extracted protein was examined by detecting the degree of ECL reaction by HRP enzyme in the secondary antibody bound to the PTH antibody as the primary antibody.
  • the cells were washed with PBS, and the cells were fixed with 10% formalin solution.
  • the fixed cells were inactivated nonspecific proteins with 2% BSA (bovine serum albumin) solution, and then treated with 2% BSA solution containing primary antibody (PTH, CHGA, Cyr61) to each cell for 4 hours at 37 ° C. Reacted for a while.
  • the unreacted antibody was washed twice with PBS, and then the fluorescent secondary antibody (mouse IgG-FITC, rabbit-IgG-Rhodamine, Invitrogen) corresponding to the primary antibody was included.
  • the 2% BSA solution was reacted at 37 ° C. for 1 hour.
  • DAPI 6-Diamidino-2-Phenylindole, Dihydrochloride
  • Molecular Probes® 6-Diamidino-2-Phenylindole, Dihydrochloride
  • the cell surface was washed twice with PBS, and then the cover glass to which the cells were attached was removed, placed on a glass for fluorescence microscopy, and then attached to the glass with a mounting solution. The expression and intracellular distribution of each antibody on the microscope were examined.
  • the expression pattern of PTH dispersed and distributed in the cytoplasm appears in the form of granules, and the hormonal storage and secretion process is performed through the formation of secretory granules and similar mechanisms like other secretory hormones. It could be predicted that it would come true.
  • the intracellular distribution patterns of CHGA were found not to be expressed in undifferentiated tonsil cells as shown in FIG. 10, but as the differentiation period elapsed in the differentiated tonsil cells, The expression of CHGA was increased and evenly distributed in the cytoplasm.
  • PTH is a major hormone that works to maintain homeostasis of calcium in vivo by recognizing changes in extracellular calcium concentrations. In vivo application and use of PTH produced from parathyroid tissue cells differentiated from tonsil cells, it is important to investigate whether PTH normally performs its own functions.
  • the extracellular calcium concentration was variously controlled to investigate the change of PTH secretion capacity according to the external calcium concentration.
  • differentiation inducing agent was treated with T-MSC for 7 days to induce differentiation into parathyroid tissue.
  • the cells differentiated into parathyroid glands were cultured in culture medium in which the calcium concentration in the cell culture medium was varied at normal concentration (1.5 mM), low concentration (0.09 mM) and high concentration (3.0 mM).
  • the cultured T-MSC was collected for each time period, and the amount of PTH secreted into the cell culture fluid according to the external calcium concentration was measured. It was investigated by ELCIA method.
  • the secretion of PTH was significantly lower than the normal calcium concentration.
  • the differentiated parathyroid tissue can also control the level of PTH secretion according to extracellular calcium concentration, and can control PTH secretion to perform the unique function of PTH, which contributes to maintaining homeostasis of calcium concentration by controlling calcium absorption / resorption. The result shows that there is.
  • the cell appearance of tonsils also shows that a large number of particles of secretory granules are formed, thereby showing that a change in cell morphology according to secretory granulation can also be induced (FIG. 13).
  • CaSR calcium sensitive receptor
  • CaSR is a receptor that exists on the surface of parathyroid cells, and it recognizes the extracellular calcium concentration and plays a role in maintaining and controlling the homeostasis of the calcium concentration in the blood.
  • the calcium concentration is low, it recognizes and promotes the secretion of the stored PTH to the outside of the cell, where the secreted PTH moves to bone tissue through blood circulation and is present on the surface of osteoblasts (PTH receptor, PTHR)
  • PTH receptor PTHR
  • CaSR having such a function is regulated by the amount of the receptor itself. Therefore, the expression level of CaSR and the expression level of PTH were examined using tonsil cells exposed to various extracellular calcium concentrations, and the correlation between the generated PTH and CaSR was investigated.
  • the expression level of CaSR is increased in the low calcium state, and the expression level is significantly decreased in the high calcium state, and CaSR may act as a major factor in the normal homeostasis control process of PTH produced in tonsil cells. I could see that.
  • Example 13 Confirmation of intracellular microstructure of tonsil stem cells differentiated into parathyroid tissue
  • differentiated tissues When stem cells are used to induce differentiation into specific organs, differentiated tissues have structural similarities with actual organ tissues or cells in order to perform specific organ specific functions.
  • amygdala stem cells differentiated into parathyroid tissue was examined by electron microscopy to investigate similarity with normal parathyroid tissue.
  • Electron microscopy was used to investigate changes in intracellular microstructure of T-MSCs differentiated into parathyroid glands.
  • the intracellular microstructure changes were investigated using cells at day 7 of differentiation, the point of maximum PTH secretion.
  • a control group cells on day 7 which were not treated with differentiation-inducing activin A and Shh were used.
  • the cells collected in pellet form were stored in 2.5% glutaraldehyde solution and fixed at 4 ° C.
  • the undifferentiated tonsils showed the basic structure of the nucleus, mitochondria and Golgi like other normal cells (Fig. 15a). ).
  • Osteoblast function by cell culture fluid obtained from T-MSCs differentiated into parathyroid tissue releasing PTH using preosteoblast MC3T3-E1 cells was investigated.
  • the culture medium was replaced every 3-4 days, and treated with various concentrations of cell culture medium (conditioned medium, CM) containing PTH secreted out of the cell (10, 50, 100, 1000 pg in PTH content in the cell culture solution).
  • CM cell culture medium
  • the culture solution is removed, and the cells attached to the bottom of each well are washed with PBS, and then fixed with 60% isopropyl alcohol for 5 minutes, washed twice with distilled water, 2 It was stained for 3 minutes with% Alizarin red S aqueous solution (pH 4.2). After staining was completed, the Alizarin red S aqueous solution was removed and immediately washed three times with distilled water, and then compared with a control group (MC3T3-E1 cultured without inducing differentiation) by observing with a phase contrast microscope.
  • a control group M3T3-E1 cultured without inducing differentiation
  • osteocalcin protein which is a representative bone formation marker
  • the expression of osteocalcin also increased as the concentration of CM increased, which was lower than that of the hormone estrogen. It showed a similar degree of bone formation ability as treated (Fig. 16).
  • PTH produced by the method of the present invention is capable of performing physiological functions unique to PTH when applied in vivo, and thus can be used as a therapeutic agent.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Organic Chemistry (AREA)
  • Biotechnology (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Rheumatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a method for preparing a chimeric mesenchymal stem cell population comprising a step of mixing and culturing tonsil tissue-derived mesenchymal stem cells obtained from different donors, and a chimeric mesenchymal stem cell population prepared by the method. Additionally, the present invention relates to a method for inducing differentiation of parathyroid tissue cells from tonsil-derived stem cells, a method for producing parathyroid hormones from the parathyroid tissue cells obtained from the method, a cell therapeutic agent including the parathyroid tissue cells for treating hypoparathyroidism or osteoporosis, and a method for treating hypoparathyroidism or osteoporosis using the cell therapeutic agent.

Description

키메라 중간엽 줄기세포군, 그의 제조방법 및 편도줄기세포를 이용하여 부갑상선 호르몬을 생산하는 방법 Chimera mesenchymal stem cell population, preparation method thereof and method of producing parathyroid hormone using tonsilocytes
본 발명은 별개의 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포를 혼합배양하는 단계를 포함하는 키메라 중간엽 줄기세포군의 제조방법, 상기 방법에 의하여 제조된 키메라 중간엽 줄기세포군, 편도줄기세포로부터 부갑상선 조직세포의 분화를 유도하는 방법 및 상기 방법에 의해 수득한 부갑상선 조직세포로부터 부갑상선 호르몬을 생산하는 방법에 관한 것이다.The present invention is a method for producing a chimeric mesenchymal stem cell group comprising the step of mixed culture of tonsil-derived mesenchymal stem cells obtained from a separate donor, chimeric mesenchymal stem cell group prepared by the method, parathyroid gland from tonsil stem cells A method for inducing differentiation of tissue cells and a method for producing parathyroid hormone from parathyroid tissue cells obtained by the method.
최근에 들어, 기관 재생과 면역반응 조절 능력을 가지는 성체 줄기세포에 대한 연구가 활발히 진행되고 있는데, 현재 임상 및 연구에서 사용되고 있는 성체 줄기세포의 공급원은 골수, 지방세포, 제대혈등이 있다(Ding DC, et al., Cell Transplant., 20:5-14, 2011; Lee OK, et al., Blood, 103:1669-1675, 2004). 그러나, 종래의 성체 줄기세포는 사용에 몇 가지 제한점이 있는데, 조직을 채취하는 과정이 침습적이고, 충분한 양을 얻기 어려우며, 대부분이 중배엽 기원의 조직이여서 외배엽과 내배엽 기원의 조직으로 분화가 용이하지 않다(Jung YJ, et al., Stem Cells Dev., 15:687-695, 2006). 이러한 제한점을 극복하기 위하여 줄기세포의 공급원을 다양화 하는 연구가 필요하다(Uranio MF, et al., Mol. Reprod. Dev., 78361L:373, 2011; Cho KA, et al., Cell Biol. Int., 33:772-777, 2009; Ivanovski SS, et al., Oral Dis., 12:358-363, 2006).In recent years, studies have been actively conducted on adult stem cells having the ability of organ regeneration and immune response regulation. Sources of adult stem cells used in clinical and research are bone marrow, adipocytes, and cord blood (Ding DC). , et al., Cell Transplant., 20: 5-14, 2011; Lee OK, et al., Blood, 103: 1669-1675, 2004). However, conventional adult stem cells have some limitations in their use, and the process of collecting tissue is invasive, difficult to obtain a sufficient amount, and most of them are tissues of mesodermal origin, and thus are not easy to differentiate into tissues of ectoderm and endoderm origin. (Jung YJ, et al., Stem Cells Dev., 15: 687-695, 2006). To overcome these limitations, research is needed to diversify the source of stem cells (Uranio MF, et al., Mol. Reprod. Dev., 78361L: 373, 2011; Cho KA, et al., Cell Biol. Int , 33: 772-777, 2009; Ivanovski SS, et al., Oral Dis., 12: 358-363, 2006).
한편, 인간의 편도선은 일종의 림프상피 면역조직으로서 인체의 구인두 비인두에 존재하는데, 사춘기까지 그 면역기능을 수행하여 이후에는 서서히 퇴화하므로, 편도선이 비정상적으로 비대해지거나(아데노이드편도 과형성: adenotonsillar hyperplasia), 감염(편도염: tonsillitis)되는 등의 문제가 발생하면 편도선 절제수술(tonsillectomy)을 통해 제거하게 된다. On the other hand, human tonsils are a type of lymphatic epithelial tissue that exists in the oropharyngeal nasopharynx of the human body, and performs its immune function until puberty and then gradually degenerates, causing the tonsils to become abnormally enlarged (adenotonsillar hyperplasia). Infections such as tonsillitis and tonsillitis can be removed by tonsillectomy.
최근, 편도선의 조직에서 중간엽 줄기세포(Mesenchymal stem cells)(MSC)를 추출한 연구결과가 보고되어, 상기 편도조직이 성체 줄기세포의 새로운 공급원으로 주목되고 있다(Janjanin SF, et al., Arthritis Res. Ther., 10:R83, 2008). 상기 편도조직으로부터 유래된 줄기세포는 다른 성체 줄기세포와 동일한 수준의 줄기세포능을 나타내어, 중배엽성 세포(지방세포형성(adipogenesis), 골세포형성(osteogenesis), 연골세포형성(chondrogenesis))로 분화될 수 있다. 이러한 편도조직의 장점은 편도선 절제수술을 통해 버려지는 조직이기 때문에 재료의 수급이 비교적 원활하다는 점이다.Recently, the results of extracting mesenchymal stem cells (MSC) from the tissues of tonsils have been reported, and the tonsils are attracting attention as a new source of adult stem cells (Janjanin SF, et al., Arthritis Res) Ther., 10: R 83, 2008). Stem cells derived from the tonsils exhibit the same level of stem cell capacity as other adult stem cells, and differentiate into mesodermal cells (adipogenesis, osteocytosis, chondrogenesis). Can be. The advantage of such tonsils is that they are discarded through tonsillectomy, so the supply and demand of materials is relatively smooth.
한편, 부갑상선 호르몬(PTH)은 부갑상선에서 분비되는 84개의 아미노산으로 이루어진 호르몬으로서, 생체내에서 칼슘농도를 조절하는 주요 호르몬이며, 부갑상선기능저하증 및 골다공증 등의 치료제로서 사용될 수 있어 부갑상선 호르몬에 대한 수요는 증가하고 있으나, 아직까지 부갑상선 호르몬을 효과적으로 생산할 수 있는 방법은 개발되지 않은 상태이다.Parathyroid hormone (PTH) is a hormone composed of 84 amino acids secreted from the parathyroid gland, and is a major hormone that controls calcium concentration in vivo, and can be used as a therapeutic agent for parathyroid hypoplasia and osteoporosis. Increasingly, however, no method has yet been developed to effectively produce parathyroid hormone.
이러한 배경하에서, 본 발명자들은 편도조직으로부터 유래된 줄기세포의 활용성을 향상시키기 위하여 예의 연구노력한 결과, 서로 다른 기증자로부터 수득한 편도조직 유래 줄기세포를 혼합배양하여도 단일 기증자로부터 수득한 편도조직 유래 줄기세포와 동일한 특성을 나타낼 뿐만 아니라, 상기 혼합배양된 줄기세포를 대상으로 냉동 및 해동과정을 수행할 경우에도 냉동보관 전의 줄기세포와 동일한 세포능력을 가지고 있으며, 줄기세포의 증식율이 증가함을 확인함으로써, 상기 줄기세포군을 줄기세포 은행에 사용가능함을 확인하였으며, 또한 편도줄기세포로부터 부갑상선 조직세포가 분화될 수 있으며, 이로부터 부갑상선 호르몬이 정상적으로 생산되고 분비됨을 확인하여, 본 발명을 완성하였다.Under these backgrounds, the present inventors have made diligent efforts to improve the utility of stem cells derived from tonsils, resulting in tonsil tissues obtained from a single donor even if mixed cultures of stem cells derived from different donors are cultured. In addition to showing the same characteristics as the stem cells, even when performing the freezing and thawing process for the mixed cultured stem cells have the same cellular capacity as the stem cells before cryopreservation, confirmed that the proliferation rate of stem cells increases Thus, it was confirmed that the stem cell group can be used for stem cell banks, and also that parathyroid tissue cells can be differentiated from tonsil stem cells, thereby confirming that parathyroid hormone is normally produced and secreted, thereby completing the present invention.
본 발명의 하나의 목적은 별개의 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포를 혼합배양하는 단계를 포함하는 키메라 중간엽 줄기세포군(Chimeric mesenchymal stem cell population)의 제조방법을 제공하는 것이다.One object of the present invention is to provide a method for producing a chimeric mesenchymal stem cell population comprising a mixed culture of tonsil-derived mesenchymal stem cells obtained from separate donors.
본 발명의 다른 목적은 상기 방법에 의하여 제조된 키메라 중간엽 줄기세포군을 제공하는 것이다.Another object of the present invention is to provide a chimeric mesenchymal stem cell group produced by the above method.
본 발명의 또 다른 목적은 상기 키메라 중간엽 줄기세포군을 냉동 및 해동시키는 단계를 포함하는 증식능이 향상된 키메라 중간엽 줄기세포군의 제조방법을 제공하는 것이다.Still another object of the present invention is to provide a method for producing a chimeric mesenchymal stem cell group having improved proliferative capacity, including freezing and thawing the chimeric mesenchymal stem cell group.
본 발명의 또 다른 목적은 상기 방법으로 제조된 향상된 증식능을 가지는 키메라 중간엽 줄기세포군을 제공하는 것이다.Still another object of the present invention is to provide a chimeric mesenchymal stem cell group having improved proliferative capacity prepared by the above method.
본 발명의 또 다른 목적은 상기 키메라 중간엽 줄기세포군을 분화시키는 방법을 제공하는 것이다.Still another object of the present invention is to provide a method for differentiating the chimeric mesenchymal stem cell population.
본 발명의 또 다른 목적은 상기 키메라 중간엽 줄기세포군으로부터 분리된 줄기세포 또는 이로부터 분화된 세포를 유효성분으로 포함하는 세포 치료제를 제공하는 것이다.Still another object of the present invention is to provide a cell therapeutic agent comprising stem cells isolated from the chimeric mesenchymal stem cell group or cells differentiated therefrom as an active ingredient.
본 발명의 또 다른 목적은 편도줄기세포로부터 부갑상선 조직세포를 분화시키는 방법을 제공하는 것이다. Another object of the present invention is to provide a method for differentiating parathyroid tissue cells from tonsil stem cells.
본 발명의 또 다른 목적은 편도줄기세포를 이용하여 부갑상선 호르몬을 생산하는 방법을 제공하는 것이다.Still another object of the present invention is to provide a method for producing parathyroid hormone using tonsil stem cells.
본 발명의 또 다른 목적은 편도줄기세포로부터 분화된 부갑상선 조직세포를 제공하는 것이다. Still another object of the present invention is to provide parathyroid tissue cells differentiated from tonsil stem cells.
본 발명의 또 다른 목적은 편도줄기세포로부터 분화된 부갑상선 조직세포를 포함하는 부갑상선 기능저하증 또는 골다공증의 치료를 위한 세포 치료제를 제공하는 것이다. It is still another object of the present invention to provide a cell therapeutic agent for the treatment of parathyroid hypoplasia or osteoporosis comprising parathyroid tissue cells differentiated from tonsil stem cells.
본 발명의 또 다른 목적은 본 발명의 부갑상선 조직 세포를 포함하는 세포치료제를 부갑상선 기능 저하증 또는 골다공증이 의심되는 개체에게 투여하는 단계를 포함하는, 부갑상선 기능 저하증 또는 골다공증의 치료 방법을 제공하는 것이다.Still another object of the present invention is to provide a method for treating parathyroid dysfunction or osteoporosis, comprising administering a cell therapy comprising parathyroid tissue cells to a subject suspected of hypothyroidism or osteoporosis.
본 발명의 키메라 중간엽 줄기세포군은 버려지는 편도조직으로부터 유래될 뿐만 아니라 별개의 편도조직으로부터 제조될 수 있으므로, 경제적으로 제조할 수 있고, 냉동 및 해동후에도 동일한 줄기세포로서의 활성을 나타내므로, 줄기세포은행의 세포공급원으로서 활용될 수 있을 것이다.The chimeric mesenchymal stem cell group of the present invention can be produced not only from the tonsil tissue discarded, but also from a separate tonsil tissue, and thus can be produced economically, and since it shows the same stem cell activity even after freezing and thawing, stem cells It can be used as a cell source for banks.
또한, 본 발명의 편도줄기세포로부터 부갑상선 조직세포를 분화시키는 방법은 편도적출술 후에 폐기되는 편도조직을 이용하여 부갑상선 조직세포를 단기간에 효과적으로 분화시킬 수 있으며, 이로부터 수득되는 부갑상선 조직세포는 체내에 존재하는 부갑상선 조직세포와 동일하게 부갑상선 호르몬을 생산하고 세포 외부의 칼슘 농도에 따라 부갑상선 호르몬의 분비를 조절할 수 있어, 부갑상선기능저하증과 골다공증을 비롯한 부갑상선 호르몬의 작용을 필요로 하는 질병의 치료에 효과적으로 이용될 수 있으며, 이와 같은 본 발명의 부갑상선 조직세포의 세포 외부 칼슘 농도를 조절함으로써 다량의 부갑상선 호르몬을 세포 밖으로 분비시켜 부갑상선 호르몬을 용이하게 수득할 수 있도록 한다.In addition, the method of differentiating parathyroid tissue cells from tonsill stem cells of the present invention can effectively differentiate parathyroid tissue cells in a short time by using tonsil tissue discarded after tonsillectomy, the parathyroid tissue cells obtained therefrom are present in the body It can produce parathyroid hormone in the same way as parathyroid tissue cells and regulate the secretion of parathyroid hormone according to the calcium concentration outside the cell, so it can be effectively used for the treatment of diseases requiring parathyroid hormone including hypothyroidism and osteoporosis. In addition, by adjusting the extracellular calcium concentration of the parathyroid tissue cells of the present invention, a large amount of parathyroid hormone is secreted out of the cells so that parathyroid hormone can be easily obtained.
도 1은 T-MSC가 실험실적 조건의 배양에서 증식됨을 보여주는 도면이다. 1 shows that T-MSCs proliferate in culture under laboratory conditions.
도 2는 T-MSC가 특이적인 표면 항원을 발현함을 보여주는 도면이다.2 shows that T-MSCs express specific surface antigens.
도 3은 T-MSC가 중배엽성 세포 및 내배엽성 세포로 분화될 수 있음을 보여주는 도면이다. 이때, FoxA2는 forkhead box A2를 의미하고; Sox17은 SRY(sex determining region Y)-box 17을 의미하며; CXCR4는 chemokine(C-X-C motif) receptor 4를 의미하고; Eya1은 eyes absent homolog 1을 의미하며; Six1은 SIX homeobox를 의미하고; Pax1은 paired box 1을 의미하며; BMP4는 bone morphogenic protein 4를 의미하고; GCM2는 glial cells missing homolog 2를 의미하며; CaSR은 calcium sensing receptor를 의미하고; CCL21은 chemokine(C-C motif) ligand 21을 의미하며; PTH는 parathyroid hormone을 의미하고; GAPDH는 glyceraldehyde-3-phosphate dehydrogenase를 의미한다.3 shows that T-MSCs can differentiate into mesodermal and endoderm cells. In this case, FoxA2 means forkhead box A2; Sox17 means sex determining region Y-box 17; CXCR4 refers to chemokine (C-X-C motif) receptor 4; Eya1 means eyes absent homolog 1; Six1 means SIX homeobox; Pax1 means paired box 1; BMP4 means bone morphogenic protein 4; GCM2 means glial cells missing homolog 2; CaSR means calcium sensing receptor; CCL21 means chemokine (C-C motif) ligand 21; PTH means parathyroid hormone; GAPDH stands for glyceraldehyde-3-phosphate dehydrogenase.
도 4는 T-MSC가 면역 조절 활성을 가짐을 보여주는 도면이다. 4 shows that T-MSCs have immunomodulatory activity.
도 5는 편도선절제술로 획득한 편도조직으로부터의 편도줄기세포 분리과정을 도식화한 그림이다. Figure 5 is a diagram illustrating the process of separating the amygdala stem cells from tonsils obtained by tonsillectomy.
도 6은 편도유래세포로부터 분화기간에 따른 세포모양의 변화를 보여주는 도면이다. 6 is a view showing the change in cell shape according to differentiation period from tonsil derived cells.
도 7은 분화 기간 동안 세포배양액으로 분비되는 PTH 농도 조사 결과를 보여주는 도면이다.7 is a view showing the results of the PTH concentration irradiation secreted into the cell culture medium during the differentiation period.
도 8은 편도세포의 부갑상선분화기간에 따른 PTH 단백질 발현양상을 보여주는 도면이다. 8 is a view showing the expression pattern of PTH protein according to the parathyroid differentiation period of tonsil cells.
도 9는 부갑상선조직으로의 분화과정 동안의 PTH의 세포 내 기질에서의 분포양상을 보여주는 도면이다. Figure 9 shows the distribution of PTH in the intracellular matrix during differentiation into parathyroid tissue.
도 10은 PTH 분비관련 단백질인 CHGA의 세포 내 발현 및 분포를 보여주는 도면이다. FIG. 10 is a diagram showing the expression and distribution of CHGA, a PTH secretion related protein.
도 11은 PTH와 CHGA의 세포 내 발현 분포를 보여주는 도면이다. 11 is a diagram showing the expression distribution of cells of PTH and CHGA.
도 12는 세포외부 칼슘 농도 변화에 따른 PTH 분비량의 조절을 보여주는 도면이다.12 is a diagram showing the regulation of PTH secretion according to the extracellular calcium concentration change.
도 13은 세포외부 칼슘 농도 변화에 따른 편도줄기세포의 세포모양의 변화를 보여주는 도면이다.13 is a view showing the change in the cell shape of tonsil stem cells according to the extracellular calcium concentration change.
도 14는 세포외부 칼슘 농도 변화에 따른 편도줄기세포에서의 칼슘감응수용체의 발현 변화를 보여주는 도면이다.14 is a view showing the change in the expression of calcium-sensitive receptors in tonsil stem cells according to the extracellular calcium concentration changes.
도 15는 부갑상선조직으로 분화된 편도줄기세포의 세포미세구조를 보여주는 도면이다.15 is a diagram showing the cell microstructure of tonsil stem cells differentiated into parathyroid tissue.
도 16은 편도줄기세포에서 생성, 분비된 PTH의 골 형성 능력을 보여주는 도면이다.16 is a view showing the bone formation capacity of PTH produced and secreted in tonsil stem cells.
상기 목적을 달성하기 위한 일 실시양태로서, 본 발명은 별개의 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포를 혼합배양하는 단계를 포함하는 키메라 중간엽 줄기세포군(Chimeric mesenchymal stem cell population)의 제조방법 또는 편도조직 유래 중간엽 줄기세포를 혼합배양하는 방법을 제공한다.As one embodiment for achieving the above object, the present invention is a method for producing a chimeric mesenchymal stem cell population comprising the step of mixed culture of tonsil-derived mesenchymal stem cells obtained from a separate donor Or it provides a method for mixed culture of tonsil-derived mesenchymal stem cells.
구체적으로, 본 발명의 키메라 중간엽 줄기세포군의 제조방법 또는 편도조직 유래 중간엽 줄기세포를 혼합배양하는 방법은 (i) 별개의 기증자로부터 분리된 각 편도조직으로부터 단핵세포를 각각 수득하는 단계; (ii) 상기 수득한 단핵세포를 배양하여 중간엽 줄기세포를 선별하는 단계; 및, (iii) 상기 선별된 중간엽 줄기세포를 혼합하여 배양하는 단계를 포함한다. Specifically, the method for producing a chimeric mesenchymal stem cell group of the present invention or a method for mixed culture of mesenchymal stem-derived mesenchymal stem cells comprises the steps of: (i) obtaining mononuclear cells from each tonsil tissue isolated from a separate donor; (ii) culturing the obtained mononuclear cells to select mesenchymal stem cells; And, (iii) mixing and culturing the selected mesenchymal stem cells.
본 발명의 용어 "편도" 또는 "편도조직"이란, 목의 안쪽과 코의 뒷부분에 위치하여, 외부에서 침입하는 세균 등의 물질로부터 일차적으로 우리 몸을 방어함과 동시에 림프상피 면역조직으로 작용을 수행하는 조직을 의미한다. 상기 편도조직은 인두편도, 귀인두관편도, 구개편도, 혀편도 등을 포함한다. 본 발명의 목적상 상기 편도조직은 편도유래 중간엽 줄기세포를 제공하는 원천조직으로 사용된다.As used herein, the term "tonsil" or "tonsil" is located at the back of the neck and the back of the nose, primarily to protect the body from substances such as bacteria that invade from the outside and act as lymphatic epithelial immune tissue. Means the organization to perform; The amygdala includes a pharyngeal tonsil, ear pharyngeal tonsil, palate tonsil, tongue tonsil, and the like. For the purpose of the present invention, the amygdala is used as a source tissue for providing tonsil-derived mesenchymal stem cells.
본 발명의 용어 "중간엽 줄기세포(mesenchymal stem cells; MSCs)"란, 골, 연골, 지방, 골수간질, 근육, 신경 등으로 분화될 수 있는 미분화된 줄기세포를 의미하는데, 성인에서는 일반적으로 골수에 머물러 있지만 제대혈, 말초혈액, 기타 조직 등에도 존재한다. 본 발명의 목적상 상기 중간엽 줄기세포는 특별히 이에 제한되지 않으나, 사람의 편도로부터 유래되어 서로 다른 개체에서 유래된 각각의 중간엽 줄기세포와 혼합배양이 가능한 세포가 될 수 있고, 지방세포, 골세포, 연골세포 등의 중배엽성 조직세포로 분화될 수 있을 뿐만 아니라, 부갑상선 조직세포 등의 내배엽성 조직세포로도 분화될 수 있다.As used herein, the term "mesenchymal stem cells (MSCs)" refers to undifferentiated stem cells that can be differentiated into bone, cartilage, fat, myeloid epilepsy, muscle, nerves, and the like. But also in cord blood, peripheral blood, and other tissues. For the purposes of the present invention, the mesenchymal stem cells are not particularly limited thereto, but may be cells that can be mixed and cultured with each mesenchymal stem cell derived from a person's tonsil and derived from different individuals, and are adipocytes, bone Not only can they differentiate into mesodermal tissue cells such as cells, chondrocytes, but also endoderm tissue cells such as parathyroid tissue cells.
본 발명의 용어 "줄기세포"란, 자기 복제 능력을 가지면서 두 개 이상의 새로운 세포로 분화하는 능력을 갖는 세포를 의미하며, 만능 줄기세포 (totipotent stem cell), 전분화능 줄기세포 (pluripotent stem cells), 다분화능 (다능성) 줄기세포(multpotent stem cells)로 분류할 수 있다. "만능 줄기세포 (totipotent stem cells)"란, 하나의 완전한 개체로 발생해 나갈 수 있는 만능의 성질을 가진 세포로 난자와 정자의 수정 이후 8세포기까지의 세포가 이러한 성질을 가지며 이 세포를 분리하여 자궁에 이식하면 하나의 완전한 개체로 발생해 나갈 수 있는 세포를 의미한다. "전분화능 줄기세포 (pluripotent stem cells)"란, 외배엽, 중배엽, 내배엽층 유래의 다양한 세포와 조직으로 발생할 수 있는 세포로서, 수정 4-5일 후 나타나는 배반포 (blastocyst)의 안쪽에 위치한 내세포괴(inner cell mass)에서 유래하며, 이를 배아 줄기 세포라 하며 다양한 다른 조직 세포로 분화되지만 새로운 생명체를 형성하지는 못하는 세포를 의미한다. As used herein, the term “stem cell” refers to a cell having self-replicating ability and capable of differentiating into two or more new cells, and includes totipotent stem cells and pluripotent stem cells. And multipotent (pluripotent) stem cells. "Totipotent stem cells" are pluripotent cells that can develop into a complete individual. Cells up to 8-cells after fertilization of eggs and sperm have this property and isolate the cells. When transplanted into the uterus means a cell that can develop into a complete individual. "Pluripotent stem cells" are cells that can develop into various cells and tissues derived from ectoderm, mesoderm, and endodermal layer, and are located inside the blastocyst appearing 4-5 days after fertilization. It is derived from the inner cell mass, which is called embryonic stem cells, and refers to cells that differentiate into various other tissue cells but do not form new life.
본 발명의 용어 "지방세포"란, 지방의 형태로 에너지를 저장하는 지방 조직을 구성하는 주된 세포를 의미한다. 상기 지방세포는 세포질 층으로 둘러싸인 커다란 지방 방울을 포함하는 백색지방세포와 지방 방울이 고루 흩어져있는 상당량의 세포질을 포함하는 다각형의 갈색지방세포의 두가지 형태로 존재할 수 있다. As used herein, the term "fat cell" refers to a major cell constituting adipose tissue that stores energy in the form of fat. The adipocytes may exist in two forms: white fat cells comprising large fat droplets surrounded by a cytoplasmic layer and polygonal brown fat cells comprising a significant amount of cytoplasm with evenly dispersed fat droplets.
본 발명의 용어 "연골세포"란, 연골에서 발견되는 유일한 세포를 의미하는데, 주로 콜라겐과 프로테오글리칸으로 구성되는 연골기질을 생산 및 유지할 수 있다. 연골 내에서 연골세포의 구성은 연골의 형태 및 조직의 위치에 따라 달라진다.The term "chondrocytes" of the present invention refers to the only cells found in the cartilage, and can produce and maintain a cartilage substrate mainly composed of collagen and proteoglycans. The composition of chondrocytes in cartilage depends on the type of cartilage and the location of the tissue.
본 발명의 용어 "골세포"란, 발육중인 뼈에 있어서 골아세포로부터 분화되는 세포를 의미하는데, 골조직속에 포함된 골소강의 내부에 존재하고, 골소강과 같은 형의 편평한 타원체 형태를 가지며, 극히 많은 돌기를 지니고, 주위골조직의 무기질대사에 관여한다.As used herein, the term "bone cell" refers to a cell that is differentiated from osteoblasts in developing bone, which is present inside the bone cavity contained in bone tissue, has a flat ellipsoidal form of the same type as the bone cavity, and is extremely It has many processes and is involved in mineral metabolism of surrounding bone tissue.
본 발명의 용어 "부갑상선 조직세포"란, 갑상선에 부착하여 있는 내분비기관인 부갑상선을 구성하는 세포를 의미하는데, 파라토르몬(parathormone)을 분비하며, 체액의 칼슘과 인의 대사를 조절한다. 본 발명에서는 부갑상선 호르몬을 분비하는 한, 부갑상선 조직세포에 포함될 수 있다.The term "parathyroid tissue cells" refers to cells constituting the parathyroid gland, an endocrine organ attached to the thyroid gland, secretes parathormone and regulates metabolism of calcium and phosphorus in body fluids. In the present invention, as long as the parathyroid hormone is secreted, it may be included in parathyroid tissue cells.
본 발명의 편도조직 유래 중간엽 줄기세포를 혼합배양하는 방법에 있어서, 편도조직으로부터 단핵세포를 수득하는 방법은 특별히 이에 제한되지 않으나, 편도조직을 분쇄하고, 이를 여과하는 방법에 의하여 수득할 수 있다.In the method for the mixed culture of tonsil-derived mesenchymal stem cells of the present invention, a method for obtaining mononuclear cells from tonsil tissue is not particularly limited thereto, and may be obtained by grinding the tonsil tissue and filtering the same. .
또한, 단핵세포를 배양하여 중간엽 줄기세포를 선별하는 방법은 특별히 이에 제한되지 않으나, 줄기세포 배양용 배지를 사용하여 배양함으로써 배양용기의 바닥에 부착된 세포를 선별하는 단계에 의하여 수행될 수 있다. 이때, 줄기세포 배양용 배지는 특별히 이에 제한되지 않으나, FBS(Invtrogen) 및 항생제를 포함하는 DMEM-HG(Dulbecco's modified Eagle's medium-High Glucose) 배지, Keratinocyte-SFM, RPMI-1640 등을 사용할 수 있고, 상기 항생제는 특별히 이에 제한되지 않으나, 스트렙토마이신, 암피실린 등이 될 수 있다.In addition, the method for selecting mesenchymal stem cells by culturing mononuclear cells is not particularly limited thereto, and may be performed by selecting cells attached to the bottom of the culture vessel by culturing using a stem cell culture medium. . At this time, the stem cell culture medium is not particularly limited to this, but DMEM-HG (Dulbecco's modified Eagle's medium-High Glucose) medium, including FBS (Invtrogen) and antibiotics, Keratinocyte-SFM, RPMI-1640 may be used, The antibiotic is not particularly limited thereto, but may be streptomycin, ampicillin, or the like.
본 발명의 용어 "혼합배양"이란, 서로 다른 별개의 기증자로부터 수득한 편도조직으로부터 분리된 중간엽 줄기세포를 혼합하여 배양하는 방법을 의미한다. 동일한 성격의 조직세포라 하여도 서로 다른 기증자로부터 분리된 세포인 경우에는 세포의 미세한 특성에 차이가 있어서, 통일된 특성을 나타내지 못하게 된다. 그러나, 본 발명의 편도조직 유래 중간엽 줄기세포는 서로 다른 별개의 기증자로부터 수득한 줄기세포를 혼합하여 배양할 경우에도, 단일 기증자로부터 수득한 줄기세포와 동일한 특성을 나타낸다.The term "mixed culture" of the present invention refers to a method of mixing and culturing mesenchymal stem cells isolated from tonsils obtained from different separate donors. Even in the case of tissue cells of the same character, cells separated from different donors have different characteristics of the cells, and thus do not exhibit uniform characteristics. However, tonsil-derived mesenchymal stem cells of the present invention exhibit the same characteristics as stem cells obtained from a single donor even when cultured by mixing stem cells obtained from different donors.
상기 목적을 달성하기 위한 다른 실시양태로서, 본 발명은 상기 방법에 의하여 제조된 키메라 중간엽 줄기세포군을 제공한다.As another embodiment for achieving the above object, the present invention provides a chimeric mesenchymal stem cell population prepared by the above method.
상기 키메라 중간엽 줄기세포군은 (i) 별개의 기증자로부터 수득한 별개의 편도조직으로부터 분리된 중간엽 줄기세포를 포함하고; (ii) 단일 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포와 동일한 세포활성을 나타내며; (iii) CD14, CD34 및 CD45의 발현이 저하되고; (iv) CD73, CD95 및 CD105의 발현이 증가하며; (v) 면역관련 표현 항체에 대하여 음성을 나타내고; (vi) 중배엽성 세포로 분화될 수 있으며; 및 (vii) 내배엽성 세포로 분화될 수 있다.Said chimeric mesenchymal stem cell population comprises (i) mesenchymal stem cells isolated from separate tonsil tissue obtained from separate donors; (ii) exhibit the same cellular activity as tonsil-derived mesenchymal stem cells obtained from a single donor; (iii) the expression of CD14, CD34 and CD45 is reduced; (iv) the expression of CD73, CD95 and CD105 is increased; (v) negative for immunorelevant expressing antibodies; (vi) can differentiate into mesodermal cells; And (vii) differentiate into endoderm cells.
본 발명의 용어 "키메라 중간엽 줄기세포군"이란, 별개의 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포를 혼합배양하여 수득한 중간엽 줄기세포를 의미하는데, 상기 키메라 중간엽 줄기세포군은 서로 다른 기증자로부터 분리된 각각의 중간엽 줄기세포가 포함되어 있으면서도, 단일 기증자로부터 분리된 중간엽 줄기세포와 동일한 수준의 세포특성을 나타낸다. 본 발명의 목적상, 상기 키메라 중간엽 줄기세포군은 T-MSC의 면역표현형은 전형적인 줄기세포의 표현형을 나타낼 수 있다. 즉, CD14, CD34 및 CD45의 발현은 약한 반면 CD73, CD95 및 CD105의 발현이 증가하고(도 2a), 면역관련 표현 항체(HLA-DR, CD40, CD80, CD86)도 음성을 나타낸다(도 2b). 뿐만 아니라, 중배엽성 세포인 지방세포, 골세포 및 연골세포로 분화될 수 있고(도 3a), 내배엽성 세포인 부갑상선 조직세포로도 분화될 수 있다(도 3b). The term “chimeric mesenchymal stem cell group” refers to mesenchymal stem cells obtained by mixed culture of tonsil-derived mesenchymal stem cells obtained from separate donors, wherein the chimeric mesenchymal stem cell groups are different donors. Each mesenchymal stem cell isolated from is included, but exhibits the same level of cellular characteristics as the mesenchymal stem cells isolated from a single donor. For the purposes of the present invention, the chimeric mesenchymal stem cell population may exhibit the phenotype of a typical stem cell immunophenotype of T-MSC. That is, the expression of CD14, CD34 and CD45 is weak while the expression of CD73, CD95 and CD105 is increased (FIG. 2A), and the immune-related expressing antibodies (HLA-DR, CD40, CD80, CD86) are also negative (FIG. 2B). . In addition, they can be differentiated into mesodermal cells, adipocytes, bone cells and chondrocytes (FIG. 3A), and also into endoderm cells, parathyroid tissue cells (FIG. 3B).
본 발명의 일 실시예에 의하면, 별개의 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포를 혼합배양할 경우, 키메라를 형성하여 하나의 중간엽 줄기세포를 형성할 수 있음을 알 수 있었고(도 4b), 단일 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포와 별개의 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포를 혼합배양하여 수득한 중간엽 줄기세포를 CFU-F 형성능 및 증식능의 측면에서 비교한 결과 유사한 수준을 나타냄을 확인하였다(도 1a). According to one embodiment of the present invention, when mixed cultured tonsil-derived mesenchymal stem cells obtained from separate donors, chimera was formed to form one mesenchymal stem cell (Fig. 4b). ), Mesenchymal stem cells obtained from a mixed culture of tonsil-derived mesenchymal stem cells obtained from a single donor and tonsil-derived mesenchymal stem cells obtained from a separate donor were compared in terms of CFU-F formation ability and proliferative capacity. The results confirmed similar levels (FIG. 1A).
상기 목적을 달성하기 위한 또 다른 실시양태로서, 본 발명은 상기 키메라 중간엽 줄기세포군을 냉동 및 해동시키는 단계를 포함하는 증식능이 향상된 키메라 중간엽 줄기세포군의 제조방법을 제공한다. As another embodiment for achieving the above object, the present invention provides a method for producing a chimeric mesenchymal stem cell group having improved proliferative capacity, including freezing and thawing the chimeric mesenchymal stem cell group.
상기 증식능이 향상된 키메라 중간엽 줄기세포군은 (i) 냉동 및 해동을 수행하지 않은 키메라 중간엽 줄기세포군와 동등한 수준의 분화능을 나타낼 수 있고; 및, (ii) 냉동 및 해동을 수행하지 않은 키메라 중간엽 줄기세포군보다 향상된 수준의 증식능을 나타낼 수 있다.The chimeric mesenchymal stem cell group having improved proliferative capacity may exhibit (i) differentiation capacity equivalent to that of the chimeric mesenchymal stem cell group that has not undergone freezing and thawing; And, (ii) an improved level of proliferative capacity than the chimeric mesenchymal stem cell population that did not perform freezing and thawing.
본 발명의 용어 "냉동"이란, 본 발명의 편도조직 유래 중간엽 줄기세포를 보존하기 위하여 액체 질소등의 냉각매체를 이용하여 냉동시키는 행위를 의미한다. 이때, 냉동시 세포의 손상을 방지하기 위하여 동결보호제를 포함하는 용액을 사용할 수 있다.The term "frozen" of the present invention refers to the action of freezing using a cooling medium such as liquid nitrogen in order to preserve the tonsil-derived mesenchymal stem cells of the present invention. In this case, a solution containing a cryoprotectant may be used to prevent cell damage during freezing.
본 발명의 용어 "동결보호제"란, 생물세포를 동결상태에서 산채로 보존할 경우, 동해를 경감할 목적으로 매액(媒液)에 첨가시키는 물질을 의미하는데, 상기 동결보호제로는 특별히 이에 제한되지 않으나, 글라이세롤, 설탕, 글루코스 등을 사용할 수 있다.The term "freeze protection agent" of the present invention refers to a substance which is added to a liquid medium for the purpose of alleviating the East Sea when biological cells are stored alive in a frozen state, and the freeze protection agent is not particularly limited thereto. However, glycerol, sugar, glucose and the like can be used.
본 발명의 용어 "해동"이란, 상기 냉동된 본 발명의 편도조직 유래 중간엽 줄기세포를 활용하기 위하여, 상온으로 온도를 증가시켜서 세포가 정상적인 생리활성을 나타내도록 하는 행위를 의미한다.The term "thawing" of the present invention, in order to utilize the frozen tonsil-derived mesenchymal stem cells of the present invention, means the action to increase the temperature to room temperature so that the cells exhibit normal physiological activity.
본 발명에서 제공하는 편도조직 유래 중간엽 줄기세포를 혼합하여 배양한 다음, 냉동 및 해동과정을 수행하면, 놀랍게도 냉동 및 해동과정을 수행하지 않은 중간엽 줄기세포 보다도 증식능이 향상됨을 확인할 수 있었다(도 1b).After mixing and incubating the mesenchymal stem cells derived from tonsil tissue provided by the present invention, and performing the freezing and thawing process, it was surprisingly found that the proliferative capacity was improved compared to the mesenchymal stem cells that did not perform the freezing and thawing process. 1b).
상기 목적을 달성하기 위한 또 다른 실시양태로서, 상기 방법으로 제조된 향상된 증식능을 가지는 키메라 중간엽 줄기세포군을 제공한다.As another embodiment for achieving the above object, it provides a chimeric mesenchymal stem cell population having improved proliferative capacity prepared by the above method.
본 발명의 용어 "향상된 증식능"이란, 본 발명에서 제공하는 편도조직 유래 중간엽 줄기세포의 증식속도가 냉동 및 해동과정을 적용한 후에 증가하는 현상을 의미한다.The term "enhanced proliferative capacity" of the present invention refers to a phenomenon in which the rate of proliferation of tonsil-derived mesenchymal stem cells provided by the present invention increases after applying a freezing and thawing process.
본 발명의 일 실시예에 의하면, 단일 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포와 별개의 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포를 혼합배양하여 수득한 중간엽 줄기세포를 냉동 및 해동하면, 혼합배양된 중간엽 줄기세포가 오히려 높은 수준의 증식능을 나타냄을 확인하였다(도 1b).According to an embodiment of the present invention, when the mesenchymal stem cells obtained by mixing and cultured tonsil-derived mesenchymal stem cells obtained from a single donor and tonsil-derived mesenchymal stem cells obtained from a separate donor are frozen and thawed. , Mixed cultured mesenchymal stem cells were confirmed to exhibit a relatively high level of proliferative capacity (FIG. 1B).
본 발명의 편도조직 유래 중간엽 줄기세포는 중배엽성 세포 및 내배엽성 세포로 분화될 수 있고, 다른 기증자 유래의 줄기세포와 혼합하여 배양하여도 단일 기증자 유래의 줄기세포와 동일한 분화능 및 증식능을 나타낼 뿐만 아니라, 냉동 및 해동을 수행하여도, 최초의 줄기세포로서 나타내는 분화능 및 증식능을 그대로 유지할 수 있으므로, 줄기세포은행을 구성하는데 필수적인 세포공급원으로 활용될 수 있다.The tonsil-derived mesenchymal stem cells of the present invention can be differentiated into mesodermal cells and endoderm cells, and show the same differentiation and proliferative capacity as stem cells derived from a single donor even when mixed with other donor stem cells. In addition, even after freezing and thawing, it is possible to maintain the differentiation capacity and proliferative capacity of the first stem cells as they are, and thus can be utilized as a cell source essential for forming a stem cell bank.
상기 목적을 달성하기 위한 또 다른 실시양태로서, 본 발명은 상기 키메라 중간엽 줄기세포군을 다양한 세포로 분화시키는 방법을 제공한다. As another embodiment for achieving the above object, the present invention provides a method for differentiating the chimeric mesenchymal stem cell population into various cells.
상기 키메라 중간엽 줄기세포군을 다양한 세포로 분화시키는 방법은 특별히 이에 제한되지 않으나, 상기 키메라 중간엽 줄기세포군을 아스코빅 산, L-프롤린, TGF-3, BMP-6 및 ITS를 함유한 연골세포 분화배지에 배양하여 연골세포로 분화시키거나, 상기 키메라 중간엽 줄기세포군을 아스코빅산 2-포스페이트, 덱사메타손 및 β-글리세로포스페이트를 함유한 배양배지에 배양하여 골세포로 분화시키거나, 상기 키메라 중간엽 줄기세포군을 3-이소부틸-1-메틸-젠틴, 덱사메타손, 인도메타신 및 인슐린을 함유한 배양배지에 배양하여 지방세포로 분화시키거나, 또는 상기 키메라 중간엽 줄기세포군을 액티빈 A(activin A) 및 Shh(sonic hedgehog)를 함유한 배양배지에 배양하여 부갑상선 조직세포로 분화시킬 수 있다.The method of differentiating the chimeric mesenchymal stem cell group into various cells is not particularly limited, but the chimeric mesenchymal stem cell group is differentiated into chondrocytes containing ascorbic acid, L-proline, TGF-3, BMP-6 and ITS. Cultured in medium to differentiate into chondrocytes, or the chimeric mesenchymal stem cell group was cultured in culture medium containing ascorbic acid 2-phosphate, dexamethasone and β-glycerophosphate to differentiate into bone cells, or the chimeric mesenchyme Stem cell populations are cultured in culture medium containing 3-isobutyl-1-methyl-gentine, dexamethasone, indomethacin and insulin to differentiate into adipocytes, or the chimeric mesenchymal stem cell population is activated by activin A (activin A). ) And Shh (sonic hedgehog) can be cultured in a culture medium containing the parathyroid tissue cells.
본 발명의 용어 "분화(differentiation)"란, 일반적으로 비교적 단순한 계(系)가 둘 이상의 질적으로 다른 부분계(部分系)로 분리되는 현상을 의미하는데, 구체적으로 세포가 분열 증식하여 성장하는 동안에 서로 구조나 기능이 특수화하는 현상, 즉 생물의 세포, 조직 등이 각각에게 주어진 일을 수행하기 위하여 형태나 기능이 변해가는 현상을 의미한다. 예를 들면, 개체발생에서 처음에 동질적이었던 알 부분 사이에 머리나 몸통 등의 구별이 생기거나 세포에도 근세포 또는 신경세포 등의 구별이 생기는 것과 같이 처음에 거의 동질이었던 어떤 생물계의 부분 사이에 질적인 차이가 생기는 것, 또는 그 결과로서 질적으로 구별할 수 있는 부역 또는 부분계로 나누어지는 현상을 들 수 있다. The term "differentiation" of the present invention generally refers to a phenomenon in which a relatively simple system is separated into two or more qualitatively different sub systems, specifically, during cell division and growth. It refers to a phenomenon in which structures and functions are specialized to each other, that is, a phenomenon in which a form or a function of a cell or tissue of an organism changes to perform a task given to each. For example, qualitatively between parts of a biological system that were initially nearly homogeneous, such as head or torso distinctions between eggs that were initially homogenous in population development, or cells such as myocytes or neurons. Phosphorus difference arises, or as a result, the phenomenon divided into sub division or sub-system which can be distinguished qualitatively is mentioned.
한편, 상기 방법으로 분화된 키메라 중간엽 줄기세포군의 분화정도를 측정하는 방법은 특별히 이에 제한되지 않으나, 당해 분야에 익히 공지된 기법인 유세포 분석방법, 면역세포화학적 방법, PCR 및 유전자 발현 프로파일을 사용하여 세포 표면 표지 또는 형태의 변화를 측정하는 방법, 광학 현미경 또는 공초점 현미경을 사용하여 세포의 형태변화를 조사하는 방법, 유전자 발현 프로파일의 변화를 측정하는 방법 등을 사용할 수 있고, 바람직하게는 RT-PCR, Oil red O 염색법, Safranin O 염색법, Type II collagen 면역조직화학 염색법, alkaline phosphate 염색법 및 Alizarin S 염색법 등을 이용할 수 있다.On the other hand, the method for measuring the degree of differentiation of the chimeric mesenchymal stem cell group differentiated by the above method is not particularly limited to this, using flow cytometry, immunocytochemical method, PCR and gene expression profile which are well known in the art. A method of measuring the change in cell surface labeling or morphology, a method of investigating morphological changes of cells using an optical microscope or a confocal microscope, a method of measuring a change in gene expression profile, and the like. -PCR, Oil red O staining, Safranin O staining, Type II collagen immunohistochemical staining, alkaline phosphate staining and Alizarin S staining can be used.
본 발명의 일 실시예에 의하면, 편도조직 유래 중간엽 줄기세포(tonsillar mesenchymal stem cells, T-MSC)는 중배엽성 세포인 지방세포, 골세포 및 연골세포로 분화될 수 있음을 확인하였고(도 3a), 뿐만 아니라 내배엽성 세포인 부갑상선 조직세포로도 분화될 수 있음을 확인하였다(도 3b). According to one embodiment of the invention, tonsil mesenchymal stem cells (tonsillar mesenchymal stem cells, T-MSC) was confirmed that can be differentiated into adipocytes, bone cells and chondrocytes that are mesodermal cells (Fig. 3a) ), As well as parathyroid tissue cells that are endoderm cells can be differentiated (Fig. 3b).
상기 목적을 달성하기 위한 또 다른 실시양태로서, 본 발명은 상기 키메라 중간엽 줄기세포군으로부터 분리된 줄기세포, 또는 상기 키메라 중간엽 줄기세포군으로부터 분화된 세포를 유효성분으로 포함하는 세포 치료제를 제공한다. 이때, 상기 분화된 세포는 특별히 이에 제한되지 않으나, 바람직하게는 세포치료에 사용될 수 있는 모든 세포가 될 수 있고, 보다 바람직하게는 중배엽성 세포 또는 내배엽성 세포가 될 수 있으며, 가장 바람직하게는 지방세포, 연골세포, 골세포, 부갑상선 조직세포 등이 될 수 있다.As another embodiment for achieving the above object, the present invention provides a cell therapeutic agent comprising stem cells isolated from the chimeric mesenchymal stem cell group, or cells differentiated from the chimeric mesenchymal stem cell group as an active ingredient. At this time, the differentiated cells are not particularly limited thereto, but may preferably be all cells that can be used for cell therapy, more preferably may be mesodermal cells or endoderm cells, most preferably fat Cells, chondrocytes, osteocytes, parathyroid tissue cells, and the like.
본 발명의 용어 "세포치료제(cellular therapeutic agent)"란, 개체로부터 분리, 배양 및 특수한 조작을 통해 제조된 세포 및 조직으로 치료, 진단 및 예방의 목적으로 사용되는 의약품(미국 FDA규정)으로서, 세포 혹은 조직의 기능을 복원시키기 위하여 살아있는 자가, 동종, 또는 이종세포를 체외에서 증식 선별하거나 다른 방법으로 세포의 생물학적 특성을 변화시키는 등의 일련의 행위를 통하여 치료, 진단 및 예방의 목적으로 사용되는 의약품을 의미한다. As used herein, the term "cellular therapeutic agent" refers to a medicinal product (US FDA regulation) used for the purpose of treatment, diagnosis, and prevention of cells and tissues prepared through isolation, culture, and special manipulation from an individual. Or drugs used for the purpose of treatment, diagnosis, and prevention through a series of actions, such as proliferating and screening live autologous, allogeneic, or heterologous cells in vitro or by altering the biological characteristics of the cells in order to restore tissue function. Means.
상기 본 발명의 키메라 중간엽 줄기세포군은, 신체의 조직 또는 기관이 목적하는 세포 군집, 예를 들어 줄기세포 또는 유래세포군집의 생착, 이식 또는 주입에 의해 강화, 치료 또는 대체되는 다양한 종류의 치료 프로토콜에 사용될 수 있다. 상기 본 발명의 키메라 중간엽 줄기세포군은 존재하는 조직을 대체 또는 강화시켜, 새롭거나 변화된 조직이 되게 하거나 생물학적 조직 또는 구조와 결합시킬 수 있다. 본 발명의 키메라 중간엽 줄기세포군은 그의 특성상 사람의 연골, 건, 인대, 부갑상선 조직 등의 강화, 치료 또는 대체에 사용될 수 있다.The chimeric mesenchymal stem cell population of the present invention is a variety of therapeutic protocols in which the tissues or organs of the body are fortified, treated or replaced by engraftment, transplantation or infusion of a desired cell population, for example, a stem cell or a derived cell population. Can be used for The chimeric mesenchymal stem cell population of the present invention can replace or enhance existing tissue, resulting in new or changed tissue, or bind to biological tissue or structure. The chimeric mesenchymal stem cell group of the present invention can be used for strengthening, treating or replacing human cartilage, tendons, ligaments, parathyroid tissues and the like.
본 발명의 다른 양태로서, 본 발명은 액티빈(Activin) A 및 소닉 헤지호그(Sonic hedgehog)(Shh)를 포함하는 배지에서 편도줄기세포를 배양하는 단계를 포함하는, 편도줄기세포로부터 부갑상선 조직세포를 분화시키는 방법을 제공한다. In another aspect of the invention, the invention is a parathyroid tissue cell from tonsils, comprising culturing tonsils in a medium comprising Activin A and Sonic hedgehog (Shh) It provides a way to differentiate.
본 발명 이전에 일부 미분화된 인간배아줄기세포 또는 소아의 흉선조직에서 유래한 성체줄기세포로부터 부갑상선 조직을 분화시키기 위한 연구가 이루어진 적은 있었으나, 편도줄기세포로부터 부갑상선 조직을 분화시키는 방법에 대한 연구는 이루어진 적이 없었다.Prior to the present invention, studies have been conducted to differentiate parathyroid tissue from some undifferentiated human embryonic stem cells or adult stem cells derived from thymus tissues of children. Never had.
또한, 배아줄기세포 또는 흉선으로부터 유래한 성체줄기세포로부터 부갑상선 조직을 유도하는 경우, 부갑상선 조직을 유도하기 위하여 6개월 이상의 오랜 시간이 걸렸다.In addition, when inducing parathyroid tissue from embryonic stem cells or adult stem cells derived from the thymus, it took longer than 6 months to induce parathyroid tissue.
하지만, 본 발명에서는 지금까지 부갑상선 조직으로 분화될 수 있는 후보로서 고려된 적이 없었던 편도줄기세포로부터 단기간 내에 부갑상선 조직의 분화를 유도할 수 있는 방법을 최초로 확인하였다.However, the present invention first identified a method capable of inducing differentiation of parathyroid tissue in a short period of time from amygdala stem cells, which have never been considered as candidates for differentiation into parathyroid tissue.
본 양태에서 용어 "편도", "줄기세포", "부갑상선 조직세포"는 상기에서 설명한 바와 같으며, 본 발명에 있어서 상기 편도는 편도유래 중간엽 줄기세포를 제공하는 원천조직으로 사용된다. In this embodiment, the terms "tonsil", "stem cell", "parathyroid tissue cells" are as described above, in the present invention, the tonsil is used as a source tissue for providing tonsil-derived mesenchymal stem cells.
본 발명의 용어 "편도줄기세포"는 편도에서 유래한 중간엽 줄기세포(T-MSCs)를 의미한다. The term "tonsilocytes" of the present invention means mesenchymal stem cells (T-MSCs) derived from tonsils.
본 발명의 부갑상선 조직세포 분화 방법은 액티빈 A와 소닉 헤지호그(Shh)를 각각 50 ng/ml ~ 300 ng/ml의 농도로 포함하는 배지에서 편도줄기세포를 배양하는 단계를 포함하며, 편도줄기세포의 배양 기간은 바람직하게는 4일 이상이며, 보다 바람직하게는 7일 이상이다. Parathyroid tissue cell differentiation method of the present invention comprises culturing the amygdala stem cells in a medium containing activin A and sonic hedgehog (Shh) at a concentration of 50 ng / ml ~ 300 ng / ml, respectively, The culture period of the cells is preferably 4 days or more, more preferably 7 days or more.
본 발명의 방법에 있어서, 편도줄기세포는 편도선 절제수술을 받은 환자에서 분리한 편도선 조직으로부터 채취하여 사용할 수 있으나, 이에 제한되지 않는다.In the method of the present invention, the amygdala stem cells may be collected from tonsil tissue isolated from a patient who has undergone tonsillectomy, but are not limited thereto.
환자에서 분리한 편도선 조직으로부터 채취할 경우, 채취한 편도조직을 완충액으로 세척한 후, 효소혼합액을 처리하여 결합조직을 분해하고, 이때 부유된 조직을 원심분리하고, 잘라진 조직 내의 세포 혼합액을 Ficol을 이용한 세포분획법과 원심분리법으로 하층의 침전된 부분만을 배양하고, 세포바닥에의 부착능을 보이는 세포부분만을 분리한 후, 배양접시 바닥에의 부착능을 보이는 세포 중 세포표면 표지자를 이용한 면역화학방법을 이용하여 편도줄기세포를 분리할 수 있다.When collected from tonsil tissue isolated from the patient, the collected tonsil is washed with buffer, and then treated with enzyme mixture to decompose the connective tissue. At this time, the suspended tissue is centrifuged and the cell mixture in the cut tissue is ficol Immunohistochemical method using cell surface markers among the cells showing adhesion to the bottom of the culture plate after culturing only the precipitated part of the lower layer by cell fractionation and centrifugation using Tonsillar stem cells can be isolated using
본 발명의 방법에 있어서, 분화유도제로서 사용되는 액티빈 A는 TGF-베타 수퍼패밀리의 구성원으로서, FSH 방출을 촉진하는 단백질로서 처음 알려졌으나, 중배엽 유도, 신경세포 분화, 골 리모델링, 조혈작용 등의 광범위한 생물학적 활성을 갖는 것으로 알려져 있다. In the method of the present invention, activin A, which is used as a differentiation inducing agent, is a member of the TGF-beta superfamily, first known as a protein that promotes FSH release, but it is known as mesoderm induction, neuronal differentiation, bone remodeling, hematopoiesis, It is known to have a wide range of biological activities.
한편, 소닉 헤지호그 단백질은 헤지호그로 불리는 포유류 시그널링 경로 패밀리 내의 3가지 단백질 중 하나로서, 사지에서 손가락의 성장 및 뇌의 조직화 등과 같은 척추동물 기관형성 조절에서 핵심 역할을 한다. 소닉 헤지호그는 그 농도에 따라 발생중인 배(embryo)의 세포에 상이한 영향을 미치는 몰포젠(morphogen)의 일종이며, 또한 성체 줄기세포의 세포 분열을 제어하며 일부 암의 발생에도 관여하는 것으로 알려져 있다.Sonic hedgehog proteins, on the other hand, are one of three proteins in a family of mammalian signaling pathways called hedgehogs, which play a key role in regulating vertebrate organogenesis such as finger growth in the extremities and brain organization. Sonic hedgehog is a type of morphogen that has different effects on developing embryonic cells depending on its concentration, and is known to control cell division of adult stem cells and to induce some cancers. .
본 발명의 방법에 있어서, 액티빈 A 및 소닉 헤지호그는 시판되는 임의의 액티빈 A 및 소닉 헤지호그를 구입하여 이용할 수 있다.In the method of the present invention, activin A and sonic hedgehog can be purchased by using any commercially available activin A and sonic hedgehog.
본 발명의 방법에서 액티빈 A와 소닉 헤지호그의 농도가 50 ng/ml 미만일 경우, 편도줄기세포로부터 부갑상선 조직세포가 분화되기 위하여 필요한 기간이 수 개월로 길어지거나 분화 효율이 떨어지는 단점이 있으며, 이들 분화유도제의 농도가 300 ng/ml을 초과할 경우, 분화유도 효율에 있어서 개선이 없어 이와 같이 고농도의 화학물질을 사용하는 것은 비효율적이다.In the method of the present invention, when the concentrations of activin A and sonic hedgehog are less than 50 ng / ml, the period required for differentiation of parathyroid tissue cells from amygdala stem cells may be extended to several months or the differentiation efficiency may be inferior. If the concentration of the differentiation induction agent exceeds 300 ng / ml, there is no improvement in the efficiency of differentiation induction, it is inefficient to use such a high concentration of chemicals.
바람직하게는, 액티빈 A와 소닉 헤지호그의 농도는 각각 80 ng/ml 내지 200 ng/ml이며, 가장 바람직하게는 각각 100 ng/ml이다. Preferably, the concentrations of Activin A and Sonic Hedgehog are each 80 ng / ml to 200 ng / ml, most preferably 100 ng / ml each.
본 발명의 방법에 따라, 액티빈 A와 소닉 헤지호그를 각각 50 ~ 300 ng/ml의 농도로 포함하는 배지에서 편도줄기세포를 배양할 경우, 짧게는 4일의 배양만으로도 편도줄기세포로부터 부갑상선 조직세포를 분화시킬 수 있다.According to the method of the present invention, when the amygdala stem cells are cultured in a medium containing activin A and sonic hedgehog at a concentration of 50 to 300 ng / ml, respectively, the parathyroid tissue from the amygdala stem cells may be shortened only by 4 days of culture. Can differentiate cells.
바람직하게는, 본 발명의 방법에 따라 4일 내지 25일 동안 편도줄기세포를 액티빈 A와 소닉 헤지호그를 각각 100 ng/ml의 농도로 포함하는 배지에서 배양함으로써 부갑상선 조직세포의 분화를 유도할 수 있으며, 배양 기간이 4일을 초과할 경우, 4-5일 마다 액티빈 A와 소닉 헤지호그를 포함하는 신선한 배지로 배지를 교체하여 줄 수 있다.Preferably, according to the method of the present invention to induce differentiation of parathyroid tissue cells by culturing tonsil stem cells in a medium containing activin A and sonic hedgehog at a concentration of 100 ng / ml, respectively, for 4 to 25 days. If the culture period exceeds 4 days, the medium can be replaced with fresh medium containing activin A and sonic hedgehog every 4-5 days.
본 발명의 방법에 있어서, 분화 유도시 사용되는 배지는 줄기세포의 배양을 위해 이용될 수 있는 통상적인 배지를 사용할 수 있는데, 예를 들면, DMEM(Dulbecco's modified Eagle medium), Keratinocyte-SFM (Keratinocyte serum free medium), RPMI-1640 등을 사용할 수 있으며, 바람직하게는 DMEM 배지를 사용할 수 있다.In the method of the present invention, the medium used to induce differentiation may be used a conventional medium that can be used for the culture of stem cells, for example, DMEM (Dulbecco's modified Eagle medium), Keratinocyte-SFM (Keratinocyte serum free medium), RPMI-1640, and the like, and preferably, DMEM medium may be used.
상기 줄기세포 배지는 첨가제로 보충될 수 있다. 일반적으로, 등장액 중의 중성 완충제(예컨대 인산염 및/또는 고농도 중탄산염) 및 단백질 영양분(예를 들면 혈청, 예컨대 FBS, 혈청 대체물, 알부민, 또는 필수 아미노산 및 비필수 아미노산, 예컨대 글루타민)을 함유할 수 있다. 나아가, 지질(지방산, 콜레스테롤, 혈청의 HDL 또는 LDL 추출물) 및 이 종류의 대부분의 보존액 배지에서 발견되는 기타 성분(예컨대 인슐린 또는 트랜스페린, 뉴클레오시드 또는 뉴클레오티드, 피루빈산염, 임의의 이온화 형태 또는 염인 당원, 예컨대 글루코스, 셀레늄, 글루코코르티코이드, 예컨대 히드로코르티존 및/또는 환원제, 예컨대 β-메르캅토에탄올)을 함유할 수 있다.The stem cell medium may be supplemented with an additive. Generally, it may contain neutral buffers (such as phosphates and / or high concentrations of bicarbonate) and protein nutrients (such as serum, such as FBS, serum substitutes, albumin, or essential and non-essential amino acids, such as glutamine) in isotonic solutions. Furthermore, lipids (fatty acids, cholesterol, HDL or LDL extracts of serum) and other components found in most preservative media of this kind (such as insulin or transferrin, nucleosides or nucleotides, pyruvate salts, any ionized form or salt) Sugar sources such as glucose, selenium, glucocorticoids such as hydrocortisone and / or reducing agents such as β-mercaptoethanol.
본 발명의 구체적인 일 실시양태에서는, 편도줄기세포를 액티빈 A와 소닉 헤지호그를 각각 100 ng/ml의 농도로 포함하는 배지에서 배양한 결과, 배양 4일 후, 부갑상선 조직세포에서 생성되는 부갑상선 호르몬의 생산이 유의적으로 증가함을 확인하였으며, 전자현미경을 이용하여 세포내 미세구조를 확인한 결과, 분화된 세포는 기본 세포 미세구조 외에 세포과립 구조 및 과립전구체 구조가 존재하여 호르몬 분비조직으로의 분화를 확인할 수 있었다.In a specific embodiment of the present invention, the parathyroid hormone produced by parathyroid tissue cells after 4 days of culture as a result of incubating the amygdala stem cells in a medium containing activin A and sonic hedgehog at a concentration of 100 ng / ml, respectively. The production of was significantly increased, and the intracellular microstructure was confirmed using electron microscopy. As a result, the differentiated cells were differentiated into hormone secretion tissues because of the presence of a basic granular microstructure and a granular and granular precursor structure. Could be confirmed.
또한, 배양배지를 4일 간격으로 신선한 배지로 교체하면서 3주간 배양한 결과, 7일 이후부터 부갑상선 호르몬의 생산이 현저히 증가함을 확인하였으며, 또한 부갑상선 조직에 특이적인 GCM2(glial cells missing homolog 2), CaSR(calcium sensing receptor), CCL21(chemokine(C-C motif) ligand 21) 및 PTH(parathyroid hormone) 유전자의 발현을 확인할 수 있었다.In addition, as a result of culturing for three weeks while replacing the culture medium with fresh medium every four days, it was confirmed that the production of parathyroid hormone was significantly increased after 7 days, and GCM2 (glial cells missing homolog 2) specific to parathyroid tissue. , CaSR (calcium sensing receptor), CCL21 (chemokine (CC motif) ligand 21) and the expression of the parathyroid hormone (PTH) genes were confirmed.
본 발명의 다른 양태로서, 본 발명은 편도줄기세포를 이용하여 부갑상선 호르몬을 생산하는 방법을 제공한다. 구체적으로, 본 발명은 i) 액티빈(Activin) A 및 소닉 헤지호그(Sonic hedgehog)(Shh)를 포함하는 배지에서 편도줄기세포를 배양하여 부갑상선 조직세포로 분화시키는 단계, 및 ii) i) 단계에서 수득한 배양액 또는 세포용해액으로부터 부갑상선 호르몬을 분리하는 단계를 포함하는, 편도줄기세포를 이용하여 부갑상선 호르몬을 생산하는 방법을 제공한다.In another aspect of the present invention, the present invention provides a method for producing parathyroid hormone using tonsilocytes. Specifically, the present invention comprises the steps of i) culturing amygdala stem cells in a medium containing Activin A and Sonic hedgehog (Shh) to differentiate into parathyroid tissue cells, and ii) i) step It provides a method for producing parathyroid hormone using tonsils, comprising the step of separating the parathyroid hormone from the culture solution or cell lysate obtained in the.
편도줄기세포로부터 부갑상선 조직세포를 분화시키는 방법과 관련하여 상기한 바와 같이, 본 발명의 방법에 따라 편도줄기세포로부터 분화된 부갑상선 조직세포는 정상적인 부갑상선 조직세포와 같이 부갑상선 호르몬을 생산하고 분비한다. 따라서, 편도줄기세포로부터 분화된 부갑상선 조직세포의 배양액 또는 세포용해액으로부터 부갑상선 호르몬을 분리함으로써 부갑상선 호르몬을 수득할 수 있다.As described above with regard to the method of differentiating parathyroid tissue cells from tonsil stem cells, parathyroid tissue cells differentiated from tonsil stem cells according to the method of the present invention produce and secrete parathyroid hormone like normal parathyroid tissue cells. Therefore, parathyroid hormone can be obtained by separating parathyroid hormone from culture or cell lysate of parathyroid tissue cells differentiated from tonsil stem cells.
본 발명의 방법에 있어서, 편도줄기세포, 액티빈 A, 소닉 헤지호그에 대해서는 상기에서 설명한 바와 같다.In the method of the present invention, tonsil stem cells, activin A, and sonic hedgehog are as described above.
본 발명의 부갑상선 호르몬 생산 방법에 있어서, 편도줄기세포를 액티빈 A와 소닉 헤지호그를 포함하는 배지에서 배양하여 부갑상선 조직세포로 분화시키는 단계는 앞서 기술한 바와 동일하다.In the parathyroid hormone production method of the present invention, the step of culturing the amygdala stem cells in a medium containing activin A and sonic hedgehog to differentiate into parathyroid tissue cells is the same as described above.
배양액 또는 세포용해액으로부터 부갑상선 호르몬을 분리하는 단계는 단백질의 분리 및 정제에 이용될 수 있는 일반적인 방법을 이용하여 실시될 수 있으며, 예를 들어, 친화성 크로마토그래피, 이온교환 크로마토그래피, 공침전 등의 방법을 이용할 수 있으나, 이에 제한되지 않고 통상의 기술자가 적절히 선택하여 실시할 수 있다.Separation of parathyroid hormone from the culture or cell lysate may be carried out using a general method that can be used for the separation and purification of proteins, for example, affinity chromatography, ion exchange chromatography, coprecipitation, etc. The method may be used, but the present invention is not limited thereto and may be appropriately selected by a person skilled in the art.
한편, 부갑상선 호르몬은 부갑상선 조직 세포내에서 생산된 후 분비과립 형태로 세포내에서 존재하게 되며, 세포 외부 칼슘 농도에 따라 세포 밖으로의 분비 여부가 결정된다.On the other hand, parathyroid hormone is produced in the parathyroid tissue cells and then present in the cells in the form of secretory granules, depending on the extracellular calcium concentration is determined whether the secretion outside the cell.
따라서, 본 발명의 방법에 있어서, 편도줄기세포로부터 유도된 부갑상선 조직세포에서 생산된 부갑상선 호르몬의 세포 밖으로의 분비를 촉진하여 부갑상선 호르몬의 분리를 용이하게 하기 위하여, 본 발명의 방법은 상기 i) 단계 후에, 배양액의 칼슘 농도를 조절하여 부갑상선 호르몬을 세포 밖으로 분비시키는 단계를 추가로 포함할 수 있다.Therefore, in the method of the present invention, in order to facilitate the secretion of parathyroid hormone produced by parathyroid tissue cells derived from tonsil stem cells to facilitate the separation of parathyroid hormone, the method of the present invention is the step i) Thereafter, adjusting the calcium concentration of the culture medium may further include secreting parathyroid hormone out of the cells.
바람직하게는, 부갑상선 조직세포로부터 부갑상선 호르몬의 분비를 촉진하기 위하여, 배양액의 칼슘 농도를 1.5 mM 미만으로 조절하는 단계를 추가로 포함할 수 있다. Preferably, in order to promote secretion of parathyroid hormone from parathyroid tissue cells, the method may further comprise adjusting the calcium concentration of the culture medium to less than 1.5 mM.
부갑상선 호르몬의 분비를 촉진하기 위한 배양액의 칼슘 농도는 바람직하게는 1.0 mM 미만이며, 보다 바람직하게는 0.5 mM 미만이며, 가장 바람직하게는 0.1 mM 미만이다.The calcium concentration of the culture medium for promoting secretion of parathyroid hormone is preferably less than 1.0 mM, more preferably less than 0.5 mM, and most preferably less than 0.1 mM.
다르게는, 부갑상선 호르몬을 세포 밖으로 분비시키지 않고, 세포를 용해하여 용해액으로부터 부갑상선 호르몬을 분리할 수도 있다.Alternatively, the parathyroid hormone may be lysed to separate the parathyroid hormone from the lysate without secreting the parathyroid hormone out of the cell.
본 발명의 부갑상선 호르몬 생산 방법은 편도절제술 후에 폐기되는 편도조직으로부터 얻어질 수 있는 편도줄기세포를 이용하여, 부갑상선 기능저하증 또는 골다공증 등의 치료에 유용하게 이용될 수 있는 부갑상선 호르몬을 용이하게 생산할 수 있도록 하며, 또한 세포 외부의 칼슘 농도 조절을 통해 부갑상선 호르몬의 분비를 촉진함으로써 다량의 호르몬을 용이하게 수득할 수 있도록 한다.Parathyroid hormone production method of the present invention by using tonsil stem cells that can be obtained from tonsil tissue discarded after tonsillectomy, to facilitate the production of parathyroid hormone that can be useful for the treatment of parathyroid hypoplasia or osteoporosis, etc. In addition, it is possible to easily obtain a large amount of hormones by promoting the secretion of parathyroid hormone through the regulation of calcium concentration outside the cell.
본 발명의 구체적인 일 실시양태에서는, 편도줄기세포를 액티빈 A와 소닉 헤지호그를 포함하는 배지에서 배양하여 부갑상선 조직의 분화를 유도하면서, 기간별로 부갑상선 호르몬의 발현 양상을 확인한 결과, 분화 유도 후 4일이 경과한 시점부터 부갑상선 호르몬의 발현이 유의적으로 증가하는 것을 확인하였으며, 분화 유도 후 7일 이후부터 발현량이 현저히 증가함을 확인함으로써, 본 발명의 방법에 의해 편도줄기세포로부터 분화된 부갑상선 조직세포로부터 부갑상선 호르몬을 생산할 수 있음을 확인하였다. In one specific embodiment of the present invention, the amygdala stem cells are cultured in a medium containing activin A and sonic hedgehog to induce differentiation of parathyroid tissue and confirm the expression patterns of parathyroid hormone in each period. It was confirmed that the expression of parathyroid hormone was significantly increased from the point of time elapsed, and that the expression level was significantly increased after 7 days after induction of differentiation, thereby making the parathyroid tissue differentiated from tonsil stem cells by the method of the present invention. It was confirmed that parathyroid hormone could be produced from the cells.
또한, 분화유도기간이 경과할수록 부갑상선 호르몬의 누적 분비량 또한 증가함을 확인하여, 편도줄기세포로부터 분화된 부갑상선 조직을 지속적으로 배양할 경우, 많은 양의 부갑상선 호르몬을 수득할 수 있음을 확인하였으며, 세포 외부의 칼슘 농도가 정상 농도(1.5 mM)보다 낮아질 경우, 부갑상선 호르몬이 세포 외부로 분비됨을 확인하여, 부갑상선 조직세포의 분화 유도시 배양액의 칼슘 농도를 조절함으로써 부갑상선 호르몬의 분비를 조절할 수 있음도 확인하였다.In addition, the cumulative secretion of parathyroid hormone increased as the differentiation-inducing period elapsed, and it was confirmed that a large amount of parathyroid hormone can be obtained by continuously culturing parathyroid tissue differentiated from tonsil stem cells. When the external calcium concentration is lower than the normal concentration (1.5 mM), it is confirmed that the parathyroid hormone is secreted to the outside of the cell, it is also possible to control the secretion of parathyroid hormone by controlling the calcium concentration of the culture medium when inducing differentiation of parathyroid tissue cells It was.
본 발명의 또 다른 실시 양태로서, 본 발명은 편도줄기세포로부터 분화된 부갑상선 조직세포를 제공한다. As another embodiment of the present invention, the present invention provides parathyroid tissue cells differentiated from tonsil stem cells.
구체적으로, 본 발명의 부갑상선 조직세포는 상기한 바와 같은, 편도줄기세포를 액티빈 A와 소닉 헤지호그를 포함하는 배지에서 배양하는 단계를 포함하는 부갑상선 조직세포 분화 방법에 의해 얻어질 수 있으며, 바람직하게는 편도줄기세포를 액티빈 A와 소닉 헤지호그를 각각 50 ng/ml 내지 300 ng/ml의 농도로 포함하는 배지에서 적어도 4일 동안 배양함으로써 얻어질 수 있으며, 부갑상선 조직 세포를 얻기 위한 가장 바람직한 액티빈 A와 소닉 헤지호그의 농도는 각각 100 ng/ml이다.Specifically, parathyroid tissue cells of the present invention can be obtained by the parathyroid tissue cell differentiation method comprising culturing tonsils in the medium containing the amygdala stem cells as described above, activin A and Sonic hedgehog, preferably Preferably, the amygdala stem cells can be obtained by culturing at least 4 days in a medium containing activin A and sonic hedgehog at a concentration of 50 ng / ml to 300 ng / ml, respectively, which is most preferred for obtaining parathyroid tissue cells. The concentrations of activin A and sonic hedgehog are 100 ng / ml each.
상기와 같은 본 발명의 분화 방법에 의해 편도줄기세포로부터 분화되어 수득된 부갑상선 조직세포는 부갑상선 호르몬을 생산할 수 있으며, 세포 외부의 칼슘 농도의 변화에 따라 부갑상선 호르몬의 분비를 조절할 수 있어서, 세포 외부의 칼슘 농도가 정상 농도(1.5 mM) 보다 낮으면 부갑상선 호르몬을 세포 밖으로 분비하고, 세포 외부의 칼슘 농도가 정상 농도보다 높으면 부갑상선 호르몬의 분비를 억제할 수 있다.Parathyroid tissue cells obtained by differentiation from amygdala stem cells by the differentiation method of the present invention as described above can produce parathyroid hormone, and can control the secretion of parathyroid hormone in response to changes in calcium concentration outside the cell, If the calcium concentration is lower than the normal concentration (1.5 mM) to secrete parathyroid hormone outside the cell, if the calcium concentration outside the cell is higher than the normal concentration can suppress the secretion of parathyroid hormone.
따라서, 본 발명의 부갑상선 조직 세포는 체내에 존재하는 부갑상선 조직 세포와 마찬가지로 부갑상선 조직 세포로서의 기능을 수행할 수 있으며, 부갑상선 조직의 기능 손상 또는 부갑상선 호르몬의 부족으로 인한 질병 또는 부갑상선 호르몬에 의해 치료가능한 것으로 알려진 골다공증 등의 치료에 유용하게 이용될 수 있다.Thus, the parathyroid tissue cells of the present invention can function as parathyroid tissue cells, like parathyroid tissue cells present in the body, and are treatable by diseases or parathyroid hormone due to impaired function of parathyroid tissue or lack of parathyroid hormone. It can be usefully used for the treatment of known osteoporosis and the like.
따라서, 본 발명의 또 다른 실시 양태로서, 본 발명은 본 발명의 부갑상선 조직 세포를 포함하는 부갑상선 기능 저하증 또는 골다공증의 세포치료제를 제공한다.Accordingly, as another embodiment of the present invention, the present invention provides a cell therapy agent of hypothyroidism or osteoporosis comprising the parathyroid tissue cells of the present invention.
본 발명에 있어서, 용어 "세포치료제"는 상기에서 설명한 바와 같다. In the present invention, the term "cell therapeutic agent" is as described above.
본 발명의 부갑상선 조직 세포를 포함하는 세포치료제가 이용될 수 있는 질병으로서는, 예를 들어, 부갑상선기능저하증, 또는 골다공증이 있다.Diseases in which the cell therapy agent comprising the parathyroid tissue cells of the present invention can be used include, for example, hypothyroidism or osteoporosis.
부갑상선 기능저하증은 부갑상선 호르몬 기능 장애로 인해 혈중 칼슘이 낮아져 생기는 저칼슘혈증에 따른 여러 증상이 발생하는 질환으로서, 그 치료법으로는 과량의 칼슘 복용이 유일한 방법이나, 부갑상선호르몬 대신 칼슘을 과량으로 복용하는 간접적 방법으로는 적정한 혈중 칼슘 농도를 지속적으로 유지하기 어렵고, 저칼슘혈증에 따른 응급상황이 흔히 발생하는 등 치료 효과의 적정성을 확보하기 어려우며, 칼슘제제 장기복용으로 인한 위염과 같은 소화기관계 합병증의 발생이 증가하는 등 여러 문제점이 있다. 따라서, 이와 같은 칼슘 복용을 대신하여, 부갑상선 호르몬을 생산할 수 있는 부갑상선 조직 세포를 포함하는 세포치료제를 이용할 경우, 부작용없이 부갑상선 기능저하증을 치료할 수 있을 것이다.Hypothyroidism is a disease caused by hypocalcemia caused by lowered calcium in the blood due to parathyroid hormone dysfunction.The only way to treat it is to take excess calcium, but to take excess calcium instead of parathyroid hormone. It is difficult to maintain proper blood calcium level continuously by indirect method, and it is difficult to secure adequacy of treatment effect such as emergency frequently due to hypocalcemia, and to develop gastrointestinal complications such as gastritis due to long-term use of calcium preparations. There are several problems, including this increase. Therefore, instead of taking calcium, when using a cell therapy comprising parathyroid tissue cells capable of producing parathyroid hormone, parathyroid hypoplasia can be treated without side effects.
한편, 골다공증은 뼈의 양이 감소하고 질적인 변화로 인해 뼈의 강도가 약해져서 골절이 일어날 가능성이 높은 상태를 말하며, 골다공증 치료제로는 주로 골흡수를 억제하는 제제가 주종이었으나, 최근 골성분 형성을 직접 촉진하는 골형성유도제로 부갑상선 호르몬의 사용이 제시되어 오고 있다. 그러나, 부갑상선 호르몬을 사용할 경우, 현재 단기간 (최대 2년 허용, 미국 FDA) 밖에 사용할 수 없고, 고가이며, 매일 인슐린주사와 같이 피하주사해야 하며, 치료 종료 후 빠른 골소실을 보이는 등의 여러 문제점들이 보고된 바 있다. 따라서, 이처럼 부갑상선 호르몬을 투여하기 보다는 부갑상선 호르몬을 생산할 수 있으며 세포 외부 칼슘의 농도 변화에 따라 호르몬의 분비를 조절할 수 있는 부갑상선 조직 세포를 포함하는 세포치료제를 이용할 경우, 부작용없이 골다공증을 치료할 수 있을 것이다.On the other hand, osteoporosis refers to a condition in which bone fractures occur due to a decrease in bone quantity due to a decrease in the amount of bone and qualitative changes, and as a treatment for osteoporosis, an agent that mainly suppresses bone resorption is mainly used. The use of parathyroid hormone has been suggested as a direct stimulator of bone formation. However, the use of parathyroid hormone can only be used for a short period of time (up to two years, US FDA), expensive, subcutaneous injections such as daily insulin injections, and rapid bone loss after the end of treatment. It has been reported. Therefore, if you use a cell therapy containing parathyroid tissue that can produce parathyroid hormone rather than administration of parathyroid hormone and can regulate hormone secretion according to the extracellular calcium concentration, osteoporosis can be treated without side effects. .
본 발명에서 용어 "치료"는 상기 세포치료제의 투여로 질환의 증세가 호전되거나 이롭게 되는 모든 행위를 의미한다.The term "treatment" in the present invention means any action that improves or benefits the condition of the disease by administration of the cell therapy.
또한, 본 발명의 세포치료제는 1㎖ 당 1.0×105개 내지 1.0×109개, 바람직하게는 1.0×106개 내지 1.0×108개, 보다 바람직하게는 1.0×107개의 세포를 포함할 수 있다.In addition, the cell therapy agent of the present invention contains 1.0 × 10 5 to 1.0 × 10 9 cells, preferably 1.0 × 10 6 to 1.0 × 10 8 cells, more preferably 1.0 × 10 7 cells per ml. can do.
본 발명의 세포치료제는 동결되지 않은 채 사용되거나 차후 사용을 위해 동결될 수 있다. 동결되어야 할 경우, 표준 냉동보존제 (예를 들어 DMSO, 글리세롤, 에피라이프 (Epilife®) 세포 동결 배지 (Cascade Biologics))가 동결 전 세포 집단에 첨가될 수 있다.The cell therapy of the invention can be used unfrozen or frozen for future use. If frozen, standard cryopreservatives (eg DMSO, glycerol, Epilife® cell freezing medium (Cascade Biologics)) can be added to the cell population prior to freezing.
또한, 상기 세포치료제는 약학적 분야의 통상의 방법에 따라 환자의 신체 내 투여에 적합한 단위투여형의 약학적 제제로 제형화시켜 투여할 수 있으며, 상기 제제는 1회 또는 수회 투여에 의해 효과적인 투여량을 포함한다. 이러한 목적에 적합한 제형으로는 비경구투여 제제로서 주사용 앰플과 같은 주사제, 주입 백과 같은 주입제, 및 에어로졸 제제와 같은 분무제 등이 바람직하다. 상기 주사용 앰플은 사용 직전에 주사액과 혼합 조제할 수 있으며, 주사액으로는 생리 식염수, 포도당, 만니톨, 링거액 등을 사용할 수 있다. 또한, 주입 백은 염화폴리비닐 또는 폴리에틸렌 재질의 것을 사용할 수 있으며, 박스터 (Baxter), 벡톤 디킨슨 (Becton Dickinson), 메드셉 (Medcep), 내셔날 호스피탈 프로덕츠 (National Hospital Products) 또는 테루모 (Terumo) 사의 주입 백을 예시할 수 있다.In addition, the cell therapy agent may be administered by formulating a unit dosage form pharmaceutical preparation suitable for administration in the body of a patient according to a conventional method in the pharmaceutical field, wherein the preparation is effectively administered by one or several administrations. Contains the amount. Formulations suitable for this purpose include parenteral injectables such as injectable ampoules, injectables such as infusion bags, sprays such as aerosol preparations and the like. The injection ampoule may be mixed with the injection solution immediately before use, and physiological saline, glucose, mannitol, and Ringer's solution may be used as the injection solution. The infusion bag can also be made of polyvinyl chloride or polyethylene and can be used in Baxter, Becton Dickinson, Medcep, National Hospital Products or Terumo. The injection bag of the yarn can be illustrated.
상기 약학적 제제에는 상기 유효성분 외에 하나 또는 그 이상의 약학적으로 허용가능한 통상의 불활성 담체, 예를 들어, 주사제의 경우에는 보존제, 무통화제, 가용화제 또는 안정화제 등을, 국소 투여용 제제의 경우에는 기제 (base), 부형제, 윤활제 또는 보존제 등을 추가로 포함할 수 있다.In the pharmaceutical preparations, in addition to the active ingredient, one or more pharmaceutically acceptable conventional inert carriers such as preservatives, analgesic agents, solubilizers or stabilizers in the case of injections, etc. It may further include a base, excipients, lubricants or preservatives.
이렇게 제조된 본 발명의 세포치료제 또는 약학적 제제는 당업계에서 통상적으로 사용하는 투여방법을 이용하여 이식 및 기타 용도에 사용되는 다른 줄기세포와 함께 또는 그러한 줄기세포와의 혼합물의 형태로 투여될 수 있으며, 바람직하게는 치료가 필요한 환자의 질환 부위에 직접 생착 또는 이식하거나 복강에 직접 이식 또는 주입하는 것이 가능하나 이에 한정되지는 않는다. 또한, 상기 투여는 카테터를 이용한 비외과적 투여 및 질환부위 절개 후 주입 또는 이식 등 외과적 투여방법 모두 가능하나 카테터를 이용한 비외과적 투여방법이 보다 바람직하다. 또한 통상의 방법에 따라 비경구적으로, 예를 들면 직접 병변에 투여하는 것 외에 조혈계 줄기세포 이식의 일반적 방법인 혈관 내 주입에 의한 이식도 가능하다.The cell therapy or pharmaceutical preparation of the present invention thus prepared may be administered with other stem cells used for transplantation and other uses, or in the form of mixtures with such stem cells, using administration methods commonly used in the art. Preferably, but not limited to engraftment or implantation directly into the disease site of the patient in need of treatment or implantation or injection directly into the abdominal cavity. In addition, the administration can be both non-surgical administration using a catheter and surgical administration methods such as injection or transplantation after dissection of the disease site, but non-surgical administration using a catheter is more preferable. In addition to parenteral administration, for example, in addition to direct lesions, transplantation by intravascular injection, which is a general method of hematopoietic stem cell transplantation, is also possible according to a conventional method.
상기 줄기세포의 1일 투여량은 1.0×104 내지 1.0×1010 세포/kg 체중, 바람직하게는 1.0×105 내지 1.0×109 세포/kg 체중을 1회 또는 수회로 나누어 투여할 수 있다. 그러나, 유효성분의 실제 투여량은 치료하고자 하는 질환, 질환의 중증도, 투여경로, 환자의 체중, 연령 및 성별 등의 여러 관련 인자에 비추어 결정되어야 하는 것으로 이해되어야 하며, 따라서, 상기 투여량은 어떠한 면으로든 본 발명의 범위를 한정하는 것은 아니다.The daily dose of the stem cells may be administered 1.0 × 10 4 to 1.0 × 10 10 cells / kg body weight, preferably 1.0 × 10 5 to 1.0 × 10 9 cells / kg body weight divided into one or several times . However, it should be understood that the actual dosage of the active ingredient should be determined in light of several relevant factors such as the disease to be treated, the severity of the disease, the route of administration, the patient's weight, age and gender, and therefore, the dosage may be It is not intended to limit the scope of the present invention in terms of aspects.
본 발명의 또 다른 실시 양태로서, 본 발명은 본 발명의 부갑상선 조직 세포를 포함하는 세포치료제를 부갑상선 기능 저하증 또는 골다공증이 의심되는 개체에게 투여하는 단계를 포함하는, 부갑상선 기능 손상 관련 질병 또는 골다공증의 치료 방법을 제공한다.In another embodiment of the present invention, the present invention provides a method for treating parathyroid function-related diseases or osteoporosis, comprising administering a cell therapy comprising parathyroid tissue cells to an individual suspected of hypothyroidism or osteoporosis. Provide a method.
본 발명의 세포치료제의 투여는 임의의 동물에 적용가능하며, 동물은 인간 및 영장류뿐만 아니라, 소, 돼지, 양, 말, 개, 쥐, 랫트 및 고양이 등의 가축을 포함한다. Administration of the cell therapy of the present invention is applicable to any animal, and animals include humans and primates, as well as domestic animals such as cattle, pigs, sheep, horses, dogs, mice, rats, and cats.
본 발명에서 용어, "투여"는 어떠한 적절한 방법으로 환자에게 본 발명의 세포치료제를 도입하는 것을 의미하며, 분화된 세포의 이식을 포함한다. 본 발명의 세포치료제의 투여 경로는 목적 조직에 도달할 수 있는 한 다양한 경로를 통하여 투여될 수 있다.As used herein, the term "administration" means introducing the cell therapy of the invention into a patient in any suitable manner and includes the transplantation of differentiated cells. Routes of administration of the cell therapy products of the invention can be administered via a variety of routes as long as they can reach the desired tissue.
이하 본 발명을 실시예를 통하여 보다 상세하게 설명한다. 그러나 이들 실시예는 본 발명을 예시적으로 설명하기 위한 것으로 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail with reference to Examples. However, these examples are for illustrative purposes only and the scope of the present invention is not limited to these examples.
실시예 1: 편도 중간엽 줄기세포의 수득Example 1: Obtaining Amygdala Mesenchymal Stem Cells
이비인후-두경부외과의 환자 중에서 10세 미만의 9명의 환자(소년 5인 및 소녀 4인, 평균연령=6.2세)를 대상으로 편도적출술을 시행하였다. 이대목동병원에서 시행하였으며 임상윤리위원회의 심의를 통과하였다(ECT 11-53-02). 적출된 편도의 3분의 2는 조직검사에 이용하였으며 나머지 3분의 1을 가지고 실험에 사용하였다.Nine patients under 10 years of age (5 boys and 4 girls, mean age = 6.2 years) underwent otolaryngology-head and neck surgery. It was conducted at Ewha Womans University Mokdong Hospital and passed the deliberation of the Clinical Ethics Committee (ECT 11-53-02). Two-thirds of the extracted tonsils were used for histology and one third were used for the experiment.
획득한 편도선 조직으로부터 줄기세포를 분리 및 배양하는 방법은 도 5와 같다.5 is a method for isolating and culturing stem cells from the obtained tonsil tissue.
구체적으로, 채취한 편도조직을 생리식염수로 세척한 후, 효소혼합액 210 U/㎖의 콜라겐분해효소 I(collagenase type I)(Invitrogen)과 10㎍/㎖ DNAse (Sigma-Aldrich, St. Louis, MO)가 혼합된 세포배양액 RPMI-1640 (Roswell Park Memorial Institute medium 1640, Invitrogen Corporation, Carlsbad, CA)에서 37℃의 조건하에서 30분 동안 편도조직내의 결합조직을 분쇄하였다. 부유된 조직을 원심분리, 여과하여 세포를 획득하였다. 획득한 세포를 RPMI 1640/20% NHS (normal human serum) (PAA Laboratories, GmbH, Paching, Austria)로 2회 세척하고, RPMI 1640/10% NHS로 1회 세척하였다. 세척된 세포를 Ficoll-Paque (GE Healthcare, Little Chalfont, Buckinghamshire, UK) 구배 원심분리에 적용하여 단핵 세포들을 수득하였다. 상기 단핵세포 108개를 10% 우태혈청(Fetal bovine serum, FBS, Invitrogen), 100㎍/㎖ 스트렙토마이신 및 100U/㎖ 암피실린을 함유하는 DMEM-HG (Dulbecco’s modified Eagle's medium-High Glucose) (Invitrogen)배지에 접종하고 48시간 동안 배양하였다. 배양이 종료된 후, 부착되지 않은 세포는 제거하고, 편도조직 유래 중간엽 줄기세포 (tonsillar mesenchymal stem cells, T-MSC)로 간주되는 부착된 세포는 4주에 걸쳐 3-5세대까지 계대 배양하여 사용하였다.Specifically, the collected tonsils were washed with physiological saline, and the enzyme mixture 210 U / mL collagenase type I (Invitrogen) and 10 μg / mL DNAse (Sigma-Aldrich, St. Louis, MO). ) Was mixed in the culture medium RPMI-1640 (Roswell Park Memorial Institute medium 1640, Invitrogen Corporation, Carlsbad, Calif.) For 30 minutes at 37 ℃ conditions connective tissue in the amygdala. The suspended tissue was centrifuged and filtered to obtain cells. The obtained cells were washed twice with RPMI 1640/20% NHS (normal human serum) (PAA Laboratories, GmbH, Paching, Austria) and once with RPMI 1640/10% NHS. Washed cells were subjected to Ficoll-Paque (GE Healthcare, Little Chalfont, Buckinghamshire, UK) gradient centrifugation to obtain mononuclear cells. Dulbecco's modified Eagle's medium-High Glucose (DMEM-HG) containing 10 8 fetal mononuclear cells (Fetal bovine serum, FBS, Invitrogen), 100 μg / ml streptomycin and 100 U / ml ampicillin (Invitrogen) The medium was inoculated and incubated for 48 hours. After incubation, unattached cells are removed, and adherent cells, considered tonsillar mesenchymal stem cells (T-MSCs), are passaged to 3-5 generations over 4 weeks. Used.
상기에서 배양된 T-MSC를 PBS(pH 7.4)로 세척하고, 세척된 T-MSC에 동결보존액(cryomedium, 50% FBS, 40% DMEM 및 10% DMSO)을 가하여 현탁시킨 다음, 상기 현탁액을 각 용기당 2 x 106개의 세포를 포함하도록 분주하였다. 상기 현탁액이 분주된 용기를 -70℃ 에서 24시간 동안 동결시키고, -200℃ 액체 질소에서 10일 동안 보관하였다(Liu Y, et al., Biotechnol. Prog., 26:1635-1643, 2010). 공지된 방법을 이용하여 상기 보관된 용기를 37℃ 물에서 2분에 걸쳐 해동시키고, 해동된 세포현탁액를 원심분리하여 T-MSC를 수득하였다. The T-MSCs incubated above were washed with PBS (pH 7.4), and suspended by adding cryodium (50% FBS, 40% DMEM and 10% DMSO) to the washed T-MSCs. Aliquots were included to contain 2 × 10 6 cells per vessel. The suspension dispensed vessel was frozen at −70 ° C. for 24 hours and stored in −200 ° C. liquid nitrogen for 10 days (Liu Y, et al., Biotechnol. Prog., 26: 1635-1643, 2010). The stored vessel was thawed in 37 ° C. water for 2 minutes using a known method, and the thawed cell suspension was centrifuged to obtain T-MSC.
실시예 2: 접착능과 CFU-F(colony-forming unit-fibroblast) 분석Example 2 Adhesion and Colony-forming Unit-Fibroblast (CFU-F) Analysis
냉동되지 않은 T-MSC와 냉동 및 해동시킨 T-MSC의 접착능과 CFU-F 분석을 위하여, 먼저, 냉동시키지 않은 T-MSC(freshly-culture cells, FC)를 14일 동안 배양하고, 배양된 세포를 PBS로 세척하며, 100% 메탄올로 5분간 고정한 다음, 메탄올을 제거하고, 5분동안 공기중에서 건조시켰다. 건조된 세포를 대상으로 Giemsa 염색을 수행하고, 섬유아세포-유사 세포의 콜로니를 위상차 현미경을 사용하여 확인하였다(Castro-Malaspina H, et al., Blood, 56:289-301, 1980; Baksh D, et al., Exp. Hematol., 31:723-732, 2003).For adhesion and CFU-F analysis of frozen and thawed T-MSCs and frozen T-MSCs, freshly-cultured fresh T-MSCs (FCs) were incubated for 14 days and then cultured. The cells were washed with PBS, fixed in 100% methanol for 5 minutes, then the methanol was removed and dried in air for 5 minutes. Giemsa staining was performed on the dried cells and colonies of fibroblast-like cells were identified using phase contrast microscopy (Castro-Malaspina H, et al., Blood, 56: 289-301, 1980; Baksh D, et al., Exp. Hematol., 31: 723-732, 2003).
아울러, 냉동 및 해동과정을 거친 T-MSC(cryopreserved-and-thawed cell, CTC)를 7일 동안 배양하고, 상술한 바와 동일한 방법에 적용하였다.In addition, cryopreserved-and-thawed cells (CTCs), which have undergone freezing and thawing, were cultured for 7 days and applied to the same method as described above.
한편, FC와 CTC의 세포 분화능을 확인하기 위하여, 각각 3 X 103개의 세포를 배양액이 담겨진 96-웰 플레이트에 접종하고, 배양하면서, 0, 3, 7 및 14일이 경과한 시점에서 MTT(3-(4,5-dimethylthizol-2-yl)2,5-diphenyl tetrazolium bromide)(Sigma-Aldrich)를 이용하여 MTT 분석법을 수행하였다.On the other hand, in order to confirm the cell differentiation capacity of FC and CTC, 3 X 10 3 cells were inoculated into 96-well plates containing culture medium, and cultured with MTT (at 0, 3, 7 and 14 days). MTT assay was performed using 3- (4,5-dimethylthizol-2-yl) 2,5-diphenyl tetrazolium bromide) (Sigma-Aldrich).
그 결과, 각 편도조직(2cm x 1.5cm x 1.5cm)으로부터 수득한 단핵세포의 수는 약 108 내지 109이고, FC과 CTC는 각각 유사한 수준의 접착능을 나타내었으며(도 1), 유사한 수준의 CFU-F 결과를 나타내었다(도 1a).As a result, the number of mononuclear cells obtained from each tonsil (2 cm x 1.5 cm x 1.5 cm) was about 10 8 to 10 9 , and FC and CTC each exhibited similar levels of adhesion (FIG. 1). Levels of CFU-F results are shown (FIG. 1A).
또한, 증식능의 측면에서도 단일 공여자로부터 얻어진 FC 배양과 여러 공여자로부터 얻어진 혼합된 FC 배양 사이에서는 차이를 보이지 않았다(도 1b). 그러나, 여러 공여자로부터 얻어진 혼합된 CTC 배양시에는 상대적으로 높은 수준의 증식능을 나타내었다.In addition, there was no difference between FC cultures obtained from a single donor and mixed FC cultures obtained from several donors in terms of proliferative capacity (FIG. 1B). However, mixed CTC cultures obtained from several donors showed relatively high levels of proliferative capacity.
실시예 3: 세포표현형 분석Example 3: Cell Phenotyping Analysis
FACSCalibur system(Becton Dickinson, Franklin Lakes, NJ)을 이용하여, 상기 FC와 CTC의 표면항원의 발현여부를 확인함으로써, T-MSC의 세포표현형을 분석하였다. Cell phenotype of T-MSC was analyzed by using FACSCalibur system (Becton Dickinson, Franklin Lakes, NJ) to confirm the expression of the surface antigens of the FC and CTC.
구체적으로, FC와 CTC를 각각 14일 및 7일동안 배양하고, 표면항원과 반응할 수 있는 각각의 항체(2㎍/㎖)를 사용하여 30분간 4℃에서 염색하고 FACSCaliur system을 이용하여 분석하였다. 이때, 사용된 항체는 다음과 같다; 조혈모세포 표지(hematopoietic cell markers)(PE-CD14, PE-CD34 및 PerCP-CD45), 시원 세포 표지(primitive cell markers)(PE-CD73, PE-CD90, 및 PE-CD105), class II MHC와 보조자극 인자 표지(co-stimulatory molecules markers)(PE-HLA-DR, FITC-CD40, PE-CD80 및 FITC-CD86) 및 모낭수지상 세포 표지(follicular dendritic cell markers)(PE-CD11b, PE-CD21, PE-CD23, PE-CD35 및 PE-CD54). Specifically, FC and CTC were incubated for 14 days and 7 days, respectively, and stained at 4 ° C. for 30 minutes using respective antibodies (2 μg / ml) capable of reacting with surface antigen and analyzed using the FACSCaliur system. . In this case, the antibodies used are as follows; Hematopoietic cell markers (PE-CD14, PE-CD34 and PerCP-CD45), primitive cell markers (PE-CD73, PE-CD90, and PE-CD105), class II MHCs and auxiliaries Co-stimulatory molecules markers (PE-HLA-DR, FITC-CD40, PE-CD80 and FITC-CD86) and follicular dendritic cell markers (PE-CD11b, PE-CD21, PE -CD23, PE-CD35 and PE-CD54).
아울러, 대조군으로서 비특이적 결합을 수행할 수 있는 항체인 IgG1 혹은 IgG2a(Becton Dickinson) isotype 항체를 사용하였다.In addition, an IgG1 or IgG2a (Becton Dickinson) isotype antibody, which is an antibody capable of performing nonspecific binding, was used as a control.
그 결과, T-MSC의 면역표현형은 전형적인 줄기세포의 표현형을 나타내었다. 즉, CD14, CD34 및 CD45의 표현은 약한 반면 CD73, CD95, CD105 표현이 증가하여, T-MSC가 조혈모세포와는 상이함을 나타내었다(도 2a). 또한, 면역관련 표현 항체(HLA-DR, CD40, CD80, CD86)도 음성을 나타내었다(도 2b). As a result, the immunophenotype of T-MSC showed a typical stem cell phenotype. That is, the expression of CD14, CD34 and CD45 was weak while the expression of CD73, CD95, CD105 was increased, indicating that T-MSC was different from hematopoietic stem cells (FIG. 2A). In addition, immune-related expressing antibodies (HLA-DR, CD40, CD80, CD86) were also negative (FIG. 2B).
편도조직에서 유래되며 T-MSC와 비슷한 접착성에 가변성을 가지며 증식하는 것으로 알려진 모낭수지상 세포(follicular dendritic cells, FDC)와 비교분석한 결과, 도 2c에서 보듯이 T-MSC는 FDC와는 다른 세포임을 알 수 있었다. As a result of comparison with follicular dendritic cells (FDC), which are derived from tonsils and are known to proliferate with adhesion similar to T-MSC, as shown in FIG. 2C, T-MSC is a different cell from FDC. Could.
아울러, FC와 CTC 군 사이에는 표현형에 차이가 없었다.In addition, there was no difference in phenotype between FC and CTC groups.
실시예 4: T-MSC의 중배엽성 세포로의 분화능 확인Example 4 Confirmation of Differentiation Capacity of T-MSCs into Mesodermal Cells
T-MSC가 중배엽성 세포로 분화가능한지를 확인하기 위하여, T-MSC를 이용하여 중배엽성 세포인 지방세포, 골세포 및 연골세포로의 분화를 유도하였다.To determine whether T-MSCs can differentiate into mesodermal cells, T-MSCs were used to induce differentiation into mesodermal cells, adipocytes, bone cells and chondrocytes.
실시예 4-1: 지방세포로의 분화Example 4-1 Differentiation into Adipocytes
3-이소부틸-1-메틸-젠틴, 덱사메타손, 인도메타신 및 인슐린을 함유한 지방세포 배양액(Invtorgen)이 담겨진 12-웰 플레이트에 각 웰당 104개의 T-MSC를 분주하고 상기 배양액을 3-4일마다 교체하면서 3주간 배양하였다. 배양이 종료된 후, 배양액을 제거하고, 각 웰의 바닥에 부착된 세포를 PBS로 세척한 다음, 4% 파라포름 알데히드를 사용하여 5분동안 고정하며, 각 웰을 건조시키고, Oil red O(Sigma-Aldrich)로 10분간 상온에서 염색하였다. 염색이 종료된 후, Oil red O를 제거하고 즉시 증류수로 4회 세척한 다음, 위상차 현미경으로 관찰하여 대조군(분화를 유도하지 않고 배양한 T-MSC)과 비교하였다(Yang Z, et al., Methods Mol. Biol., 698:353-366, 2011; Maxson S, et al., J. Tissue Eng. Regen. Med., 2:147-154, 2008).Dispense 10 4 T-MSCs per well into a 12-well plate containing adipocyte culture (Invtorgen) containing 3-isobutyl-1-methyl-gentine, dexamethasone, indomethacin and insulin, and add the culture 3- Incubate for 3 weeks with replacement every 4 days. After the incubation was completed, the culture solution was removed, the cells attached to the bottom of each well were washed with PBS, fixed for 5 minutes using 4% paraformaldehyde, each well was dried, and oil red O ( Sigma-Aldrich) was stained at room temperature for 10 minutes. After staining, oil red O was removed and immediately washed 4 times with distilled water, and then compared with a control group (T-MSC incubated without inducing differentiation) by observing with a phase contrast microscope (Yang Z, et al., Methods Mol. Biol., 698: 353-366, 2011; Maxson S, et al., J. Tissue Eng. Regen. Med., 2: 147-154, 2008).
실시예 4-2: 골세포로의 분화Example 4-2 Differentiation into Osteoblasts
아스코빅산 2-포스페이트, 덱사메타손 및 β-글리세로포스페이트를 함유한 골세포 배양액(Invtorgen)이 담겨진 12-웰 플레이트에 각 웰당 104개의 T-MSC를 분주하고 상기 배양액을 3-4일마다 교체하면서 3주간 배양하였다. 배양이 종료된 후, 배양액을 제거하고, 각 웰의 바닥에 부착된 세포를 PBS로 세척한 다음, 60% 이소프로필알콜을 사용하여 5분동안 고정하며, 증류수로 2회 세척하고, 2% Alizarin red O 수용액(pH 4.2)로 3분간 염색하였다. 염색이 종료된 후, Alizarin red O 수용액를 제거하고 즉시 증류수로 3회 세척한 다음, 위상차 현미경으로 관찰하여 대조군(분화를 유도하지 않고 배양한 T-MSC)과 비교하였다(Yang Z, et al., Methods Mol. Biol., 698:353-366, 2011; Maxson S, et al., J. Tissue Eng. Regen. Med., 2:147-154, 2008).Dispense 10 4 T-MSCs per well into 12-well plates containing bone cell culture (Invtorgen) containing ascorbic acid 2-phosphate, dexamethasone and β-glycerophosphate, and replace the culture every 3-4 days. Incubated for 3 weeks. After the incubation was terminated, the culture solution was removed, the cells attached to the bottom of each well were washed with PBS, fixed for 5 minutes using 60% isopropyl alcohol, washed twice with distilled water, 2% Alizarin Staining was performed with a red O aqueous solution (pH 4.2) for 3 minutes. After staining was completed, the Alizarin red O aqueous solution was removed and immediately washed three times with distilled water, and then compared with a control group (T-MSC cultured without inducing differentiation) by observation under a phase contrast microscope (Yang Z, et al., Methods Mol. Biol., 698: 353-366, 2011; Maxson S, et al., J. Tissue Eng. Regen. Med., 2: 147-154, 2008).
실시예 4-3: 연골세포로의 분화Example 4-3 Differentiation into Chondrocytes
덱사메타손, 아스코빅 산, L-프롤린, TGF-3, BMP-6 및 ITS를 함유한 연골세포 배양액(Invtorgen)이 담겨진 14㎖ 코니칼 튜브에 2 X 106개의 T-MSC를 접종하고 상기 배양액을 3-4일마다 교체하면서 3주간 배양하였다. 배양이 종료된 후, 배양된 세포를 수거하고, 수거된 세포를 4% 파라포름 알데히드로 고정하고 파라핀으로 임베드(embedded)하여 파라핀 블럭을 수득하였다. 상기 수득한 파라핀 블럭을 5㎛ 두께로 세절하여 박편을 수득하고, 상기 수득한 박편을 슬라이드 글라스 위에 올린 후, 파라핀을 제거하여, 슬라이드 글라스상에 세포절편을 고정하였다. 상기 슬라이드 글라스에 고정된 세포절편에 포함된 황화단백클리칸(sulfated preteoglycan)을 Alcian blue(pH 2.3)으로 염색하고, 현미경으로 관찰하여 대조군(분화를 유도하지 않고 배양한 T-MSC)과 비교하였다(Yang Z, et al., Methods Mol. Biol., 698:353-366, 2011).2 x 10 6 T-MSCs were inoculated into a 14 ml conical tube containing chondrocyte culture medium (Invtorgen) containing dexamethasone, ascorbic acid, L-proline, TGF-3, BMP-6 and ITS. Incubate for 3 weeks with replacement every 3-4 days. After the incubation was terminated, the cultured cells were harvested, and the harvested cells were fixed with 4% paraformaldehyde and embedded with paraffin to obtain paraffin blocks. The obtained paraffin block was cut to a thickness of 5 μm to obtain flakes. The obtained flakes were placed on a slide glass, and then paraffin was removed to fix cell sections on the slide glass. Sulfated preteoglycans contained in the cell sections fixed on the slide glass were stained with Alcian blue (pH 2.3), and compared with a control group (T-MSC cultured without inducing differentiation) under a microscope. (Yang Z, et al., Methods Mol. Biol., 698: 353-366, 2011).
실시예 4-4: 결과Example 4-4: Results
3주간의 지방분화 유도 후 세포내 지방 방울(lipid droplets)이 형성됨을 확인하였고(도 3a, 좌측), 골세포로의 분화와 칼슘의 침착이 발생됨을 확인하였으며(도 3a, 중앙), 연골세포로의 분화와 매트릭스 황화 단백클리칸(matrix sulfated preteoglycan)의 축적을 확인하였다(도 3a, 우측). 이에 반하여, 대조군에서는 분화특이적인 특성이 관찰되지 않았다.After 3 weeks of induction of fat differentiation, it was confirmed that intracellular lipid droplets were formed (FIG. 3A, left), and differentiation into bone cells and deposition of calcium occurred (FIG. 3A, center). Differentiation of the furnace and accumulation of matrix sulfated preteoglycan was confirmed (FIG. 3A, right). In contrast, no differentiation-specific characteristics were observed in the control group.
실시예 5: T-MSC의 내배엽성 세포로의 분화능 확인Example 5 Confirmation of Differentiation Ability of T-MSCs into Endoderm Cells
T-MSC가 내배엽성 세포로 분화가능한지를 확인하기 위하여, T-MSC를 내배엽성 세포인 부갑상선 조직세포(parathyroid cell-like cells)로의 분화를 유도하였다.To determine whether T-MSCs can differentiate into endoderm cells, T-MSCs were induced to differentiate into parathyroid cell-like cells, which are endoderm cells.
구체적으로, T-MSC를 5% FBS, 액티빈 A(activin A, 100 ng/㎖, R&D System, Inc. Minneapolis)와 sonic hedgehog(Shh)(100ng/㎖, R&D Systems)를 포함하는 RPMI 1640 배지에 접종하고, 상기 배양액을 4일마다 교체하면서 3주간 배양하였다. 배양이 종료된 후, 역전사 효소 연쇄 반응(RT-PCR) 방법으로 내배엽 특이적인 단백질의 발현을 확인하였고, 부갑상선 조직세포에 특이적인 GCM2(glial cells missing homolog 2), CaSR(calcium sensing receptor), CCL21(chemokine(C-C motif) ligand 21) 및 PTH(parathyroid hormone) 유전자의 발현을 확인하였다(Bingham EL, et al., Stem Cells Dev., 18:1071-1080, 2009). Specifically, T-MSC RPMI 1640 medium containing 5% FBS, activin A (activin A, 100 ng / ml, R & D System, Inc. Minneapolis) and sonic hedgehog (Shh) (100 ng / ml, R & D Systems) Inoculated and cultured for 3 weeks while replacing the culture every 4 days. After the incubation was completed, the expression of endoderm-specific protein was confirmed by reverse transcriptase chain reaction (RT-PCR) method. GCM2 (glial cells missing homolog 2), CaSR (calcium sensing receptor), and CCL21 specific for parathyroid tissue cells (chemokine (CC motif) ligand 21) and the expression of parathyroid hormone (PTH) genes were confirmed (Bingham EL, et al., Stem Cells Dev., 18: 1071-1080, 2009).
또한, T-MSC에서 분화된 내배엽성 세포에서 총 RNA를 추출하고, 추출된 RNA 1㎍을 RT system(Promega, Madison, WI)에 적용하여 cDNA를 합성하였다. 이때, 사용된 프라이머의 서열은 표 1에 기재하였고, 전체적인 방법은 TRIZOL(Invitrogen) 사의 사용자 매뉴얼에 따라 수행하였다.In addition, total RNA was extracted from endoderm cells differentiated from T-MSC, and cDNA was synthesized by applying 1 μg of the extracted RNA to an RT system (Promega, Madison, Wis.). At this time, the sequence of the primer used is described in Table 1, the overall method was performed according to the user's manual of TRIZOL (Invitrogen).
표 1
Figure PCTKR2013003644-appb-T000001
Table 1
Figure PCTKR2013003644-appb-T000001
FoxA2: forkhead box A2, SOX17: SRY(sex determining region Y)-box 17, CXCR4: chemokine(C-X-C motif) receptor 4, Eya1: eyes absent homolog 1, Six1: SIX homeobox 1, Pax1: paired box 1, BMP4: bone morphogenic protein 4, GCM2: glial cells missing homolog 2, CaSR: calcium sensing receptor, CCL21: chemokine(C-C motif) ligand 21, PTH: parathyroid hormone, and GAPDH: glyceraldehyde-3-phosphate dehydrogenaseFoxA2: forkhead box A2, SOX17: sex determining region Y (SRY) -box 17, CXCR4: chemokine (CXC motif) receptor 4, Eya1: eyes absent homolog 1, Six1: SIX homeobox 1, Pax1: paired box 1, BMP4: bone morphogenic protein 4, GCM2: glial cells missing homolog 2, CaSR: calcium sensing receptor, CCL21: chemokine (CC motif) ligand 21, PTH: parathyroid hormone, and GAPDH: glyceraldehyde-3-phosphate dehydrogenase
그 결과, RT-PCR 결과에서 내배엽성 세포의 표지 유전자인 FoxA2, Sox17 및 CXCR4의 발현을 확인하였고(도 3b), 흉선과 부갑성 마커(thymus/parathyroid primordium markers) 인 Eya1, Six1, Pax1, Noggin 및 BMP4의 발현도 확인하였으며, 부갑상선의 결정적 마커인 GCM2, CaSR, CCL21 및 PTH의 발현도 확인하였다. As a result, the expression of FoxA2, Sox17 and CXCR4, which are marker genes of endoderm cells, was confirmed in RT-PCR results (Fig. 3b), and thymus and parathyroid markers (thymus / parathyroid primordium markers) Eya1, Six1, Pax1, Noggin Expression of BMP4 was also confirmed, and expression of GCM2, CaSR, CCL21 and PTH, which are crucial markers of the parathyroid gland, was also confirmed.
상기 결과로부터 T-MSC가 내배엽성 세포인 부갑상선 조직세포로 분화될 수 있음을 알 수 있었다.The results show that T-MSCs can be differentiated into parathyroid tissue cells, which are endoderm cells.
실시예 6: T-MSC의 면역조절능 분석Example 6: Analysis of immunomodulatory capacity of T-MSCs
인간 CD4+ T 세포를 건강한 공여자의 말초 혈액으로부터 수득하였고, PMA(Phorbol 12- myristate 13-acetate)(Sigma-Aldrich)를 유사분열 촉진제로 사용하였다. Human CD4 + T cells were obtained from the peripheral blood of healthy donors and PMA (Phorbol 12- myristate 13-acetate) (Sigma-Aldrich) was used as a mitosis promoter.
공지된 방법에 따라, PMA-자극 T 세포 분열의 억제능을 측정하였다(Rasmusson I, et al., Exp. Cell Res., 305:33-41, 2005). In accordance with known methods, inhibition of PMA-stimulated T cell division was measured (Rasmusson I, et al., Exp. Cell Res., 305: 33-41, 2005).
CellTraceTM CFSE Cell Proliferation Kit(Invitrogen)을 이용하여, CD4+ T 세포를 CFSE(carboxyfluorescein diacetate, succinimidyl ester)로 표지하였다. 구체적인 방법은 다음과 같다: 세포를 미리 가온한 0.1% BSA를 포함하는 PBS에 106세포/㎖의 농도로 현탁한 다음, 10μM CFSE를 가하였다. 이어, 37℃에서 10분간 배양하고, 배양된 세포를 다시 5배의 ice-cold 배양액에 정치시켜서 염색을 종료시킨 다음, 원심분리하여, CFSE로 표지된 CD4+ T 세포를 수득하였다. Using CellTraceTM CFSE Cell Proliferation Kit (Invitrogen), CD4 + T cells were labeled with carboxyfluorescein diacetate (succinimidyl ester) (CFSE). The specific method is as follows: The cells were suspended in PBS containing 0.1% BSA pre-warmed at a concentration of 10 6 cells / ml, followed by the addition of 10 μM CFSE. Subsequently, the cells were incubated at 37 ° C. for 10 minutes, and the cells were left in 5 times ice-cold culture to terminate staining, followed by centrifugation to obtain CD4 + T cells labeled with CFSE.
상기 수득한 CFSE로 표지된 CD4+ T 세포(3 x 104 세포)를 배양액에 재현탁하여 3회 세척하고, 세척된 세포를 배양된 T-MSC가 존재하는 웰(104 세포) 또는 존재하지 않는 웰에 접종하였다. 그런 다음, 상기 세포가 접종된 각 웰에 최종농도 1 내지 200 ng/㎖의 PMA를 가하고, 3일 동안 배양한 다음, CFSE+ CD+ T 세포를 유세포분석법(flow cytometry)으로 증식정도를 측정하고, ModiFitLT 소프트웨어를 이용하여 CFSE 양성세포의 감소정도를 분석하였다.The obtained CFSE-labeled CD4 + T cells (3 × 10 4 cells) were resuspended in culture and washed three times, and the washed cells were wells (10 4 cells) with or without cultured T-MSCs or not. The wells were inoculated. Then, PMA at a final concentration of 1 to 200 ng / ml was added to each well inoculated with the cells, and cultured for 3 days, and then the proliferation of CFSE + CD + T cells was measured by flow cytometry, and ModiFitLT The reduction of CFSE positive cells was analyzed using the software.
분석 결과, PMA는 그의 투여랑에 비례하는 방식으로 CD4+ T 세포의 증식을 유도하였으나, T-MSC의 처리에 의하여 PMA에 의한 CD4+ T 세포의 증식유도가 억제됨을 확인하였고, 이는 FC와 CTC 양쪽 모두 유사한 수준을 나타내었다(도 4a).As a result, PMA induced the proliferation of CD4 + T cells in a proportional manner to its administration, but it was confirmed that the induction of proliferation of CD4 + T cells by PMA was inhibited by treatment with T-MSC. Similar levels were shown (FIG. 4A).
실시예 7: 다수 공여자로부터 추출한 T-MSC의 STR(Short tandem repeats) 분석Example 7: Short tandem repeats (STR) analysis of T-MSCs extracted from multiple donors
3명의 독립적인 공여자(2명의 소년과 1명의 소녀)로부터 얻은 각각의 T-MSC를 각각 개별적으로 또는 함께 혼합하여 4주동안 배양하였다. 배양이 종료된 후, 각 배양된 세포에서 게놈 DNA를 추출하여 공지된 방법으로 STR 분석을 수행함으로써 chimerism을 확인하였다(Kristt DM, et al., J. Biomol. Tech., 16:380-391, 2005; Kim DW, et al., Blood, 103:1941-1948, 2004). 이때, 추출된 게놈 DNA는 PCR로 증폭하여 사용하고, STR 분석은 capillary electrophoresis(ABI 3130Xl Genetic analyzer, Carlsbad, CA)를 이용하여 수행하였으며, 결과분석은 GeneMapper software를 이용하여 수행하였다. Each T-MSC from three independent donors (two boys and one girl) was incubated for four weeks, either individually or mixed together. After the incubation was completed, chimerism was confirmed by extracting genomic DNA from each cultured cell and performing STR analysis by a known method (Kristt DM, et al., J. Biomol. Tech., 16: 380-391, 2005; Kim DW, et al., Blood, 103: 1941-1948, 2004). At this time, the extracted genomic DNA was amplified by PCR, STR analysis was performed using capillary electrophoresis (ABI 3130Xl Genetic analyzer, Carlsbad, CA), and the result analysis was performed using GeneMapper software.
STR 분석을 통하여, 총 15개의 부위중에서 D8S1179, D3S1358 및 D18S51의 3개의 부위에서 키메라(이형접합체)를 형성함을 확인하였다(도 4b). 상기 결과에서 보듯이, 다양한 기원의 편도로부터 유래된 T-MSC는 혼합배양시 키메라를 형성할 수 있음을 확인하였으므로, 상기 T-MSC는 기원에 상관없이 동등하고 유사한 수준의 세포특성을 나타내어, 혼합배양을 통해 하나의 중간엽 줄기세포군을 형성할 수 있음을 알 수 있었다.Through STR analysis, chimeras (heterozygote) were formed at three sites of D8S1179, D3S1358 and D18S51 among the total 15 sites (FIG. 4B). As shown in the results, it was confirmed that T-MSCs derived from amygdala of various origins can form chimeras in mixed cultures, and thus, T-MSCs exhibited similar and similar levels of cellular characteristics regardless of origin, resulting in mixed Through culture, it was found that one mesenchymal stem cell group could be formed.
실시예 8: 편도줄기세포의 부갑상선조직으로의 분화유도Example 8: Induction of Differentiation of Tonsil Stem Cells into Parathyroid Tissues
편도줄기세포의 부갑상선 조직으로의 분화에 대한 추가 연구를 위하여, 단일 개체로부터 유래한 T-MSC를 부갑상선 조직세포(parathyroid cell-like cells)로의 분화를 유도하였으며, 구체적으로, T-MSC를 5% FBS, 액티빈 A (activin A, 100 ng/㎖, R&D System, Inc. Minneapolis)와 sonic hedgehog (Shh, 100 ng/㎖, R&D Systems)를 포함하는 DMEM 배지에 접종하고, 상기 배양액을 4일마다 교체하면서 21-25일간 배양하였다. For further study of the differentiation of amygdala stem cells into parathyroid tissue, differentiation of T-MSCs from single individuals into parathyroid cell-like cells was induced. Specifically, 5% of T-MSCs were induced. Inoculate DMEM medium containing FBS, activin A (activin A, 100 ng / ml, R & D System, Inc. Minneapolis) and sonic hedgehog (Shh, 100 ng / ml, R & D Systems), and incubate the culture every 4 days Incubate for 21-25 days with replacement.
배양 결과, 분화기간이 경과함에 따라 편도줄기세포는 도 6과 같이 여러 층으로 누적되는 multi-layer를 형성함과 동시에 입방형의 세포 모양을 형성하는 것이 관찰되었다. As a result of the culture, as the differentiation period elapsed, the amygdala stem cells formed a multi-layer accumulating into several layers as shown in FIG.
실시예 9: 분화기간에 따른 PTH의 분비정도 조사Example 9 Investigation of PTH Secretion by Differentiation Period
분화기간별로 분화유도제가 함유된 세포배양액으로 교환해줄 때마다 (0, 4, 7, 10, 15, 20일), 기존의 세포배양액을 모아서 부갑상선조직으로의 분화과정에 따라 세포외부로 분비되는 PTH의 농도를 분석하기 위한 시료로 사용하였다. 각 시기별로 모은 세포배양액을 0.45 ㎛크기의 시린지 필터로 여과하여 세포부유물이나 세포파편 등을 제거한 뒤, 여과된 세포배양액을 동결건조기 (Freeze Dryer, Operon)를 사용하여 동결건조, 농축하였다. Whenever exchanged with cell culture medium containing differentiation inducer by differentiation period (0, 4, 7, 10, 15, 20 days), PTH is collected and secreted into the extracellular cells during differentiation into parathyroid tissue. It was used as a sample for analyzing the concentration of. Cell cultures collected at each time were filtered through a 0.45 μm syringe filter to remove cell suspensions and cell debris, and the filtered cell cultures were freeze-dried and concentrated using a freeze dryer (Operon).
동결건조한 파우더형태의 시료는 Phosphate buffered saline (pH 7.4)으로 재현탁하여 면역화학법으로 PTH 농도를 분석하였다. 즉, PTH 단백질에 대한 항체와의 결합력을 이용한 ELCIA 방법으로 배출되는 PTH 농도를 조사하였다. Lyophilized powder samples were resuspended in phosphate buffered saline (pH 7.4) and analyzed for PTH concentration by immunochemistry. That is, the concentration of PTH released by the ELCIA method using the binding force with the antibody to the PTH protein was investigated.
그 결과, 도 7a와 같이, 분화 유도액을 처리한 세포에서는 분화하지 않은 세포에 비해 통계적으로 유의적으로 많은 양의 PTH가 세포배양액으로 분비되었다. 이를 분화기간별로 조사한 결과, 분화유도액 처리 7일 이후부터 10일까지 현저히 증가함을 알 수 있었고, 그 양은 최대 40 pg/ml 농도로 이는 생체내 정상 PTH 농도범위인 15-60 pg/ml의 범위에 포함됨을 알 수 있었다 (도 7b). 이를 누적 PTH 농도 값으로 조사한 결과, 분화유도액을 처리한 후 10일 이후의 누적 PTH 분비량은 100 pg/ml의 농도로 조사되어, 분화한 편도줄기세포를 지속적으로 배양할 경우, 100 pg/ml의 PTH도 획득 가능함을 알 수 있었다 (도 7c).As a result, as shown in Fig. 7a, the cells treated with the differentiation inducing liquid secreted statistically significant amount of PTH into the cell culture fluid compared with the cells that did not differentiate. As a result of the investigation by differentiation period, it was found that the increase was significantly increased from 7 days to 10 days after the differentiation induction solution, and the amount was up to 40 pg / ml, which was 15-60 pg / ml of normal PTH concentration range in vivo. It was found to be included in the range (FIG. 7B). As a result of investigating the cumulative PTH concentration value, the cumulative PTH secretion after 10 days after the differentiation induction solution was investigated at a concentration of 100 pg / ml, 100 pg / ml when continuously cultured differentiated tonsil cells It can be seen that the PTH of can also be obtained (Fig. 7c).
실시예 10: 웨스턴 블롯 방법에 의한 PTH 단백질 및 관련 단백질 발현조사Example 10 PTH Protein and Related Protein Expression by Western Blot Method
각 분화단계별로 분화과정 중의 T-MSC를 모은 뒤, 세포표면을 PBS (Phosphate buffered saline, pH 7.4)로 2회 세척한 뒤, RIPA 버퍼 (25mM Tris·HCl pH 7.6, 150mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS)를 이용하여 세포내 단백질 부분을 추출하였다. 추출된 단백질은 알부민을 표준용액으로 한 BCA (bicinchoninic acid)방법으로 정량하여, 세포내 PTH 단백질 발현조사에 사용하였다. After the differentiation step, T-MSCs were collected during differentiation, the cell surface was washed twice with PBS (Phosphate buffered saline, pH 7.4), and then RIPA buffer (25 mM Tris-HCl pH 7.6, 150 mM NaCl, 1% NP- 40, 1% sodium deoxycholate, 0.1% SDS) was used to extract the intracellular protein portion. The extracted protein was quantified by BCA (bicinchoninic acid) method using albumin as a standard solution, and used for intracellular PTH protein expression.
10-12 %의 SDS (sodium deoxycholate)-폴리아크릴아미드 젤 (SDS-PAGE)을 제조하고, 분화기간 각 단계별로 정량한 단백질 시료 30㎍씩을 미리 준비한 SDS-PAGE에 로딩하여 전기영동함으로써, 추출한 세포내 단백질들이 각각의 크기별로 분리되도록 하였다. 전기영동 후, 젤상에서 분리한 각 크기별 단백질들을 니트로셀룰로오스 멤브레인으로 전기적인 방법을 이용하여 옮겨주었다. 10-12% SDS (sodium deoxycholate) -polyacrylamide gel (SDS-PAGE) was prepared, and 30 ㎍ of protein samples quantified at each stage of differentiation period were loaded on pre-prepared SDS-PAGE, followed by electrophoresis. My proteins were separated by their size. After electrophoresis, proteins of each size separated on the gel were transferred to the nitrocellulose membrane using an electric method.
멤브레인상으로 전달된 각 크기별 단백질 중, PTH 단백질 크기에 해당하는 멤브레인 부분만을 취하고, 취한 멤브레인에 PTH 특이 항체 (Ab frontier, LF-MA0140), 칼슘 감응 수용체(calcium sensing receptor) (CaSR, Abcam ab18200), 크로마그라닌(chromagranin) A (CHGA, AVIVA ARP-41930-T100), 시스테인 농후 단백질(Cystein rich protein) 61 (Cyr61,Santa Cruz SC-13100) 및 오스테오칼신(osteocalcin) (OC, Santa Cruz SC-74495) 를 첨가하여 14-16시간 동안 4℃ 저온상태에서 항체와 반응시켰다. 1차 항체와의 반응 후 (1:1000 비율로 희석), 1차 항체 특이적인 2차 항체인 마우스-IgG-HRP (horse radish peroxidase) conjugate을 추가 반응시켜주었다 (1:3000 비율로 희석). 세포내 PTH 단백질 항원과 결합한 PTH 항체의 반응은 ECL (enhanced chemiluminescence)방법을 통해 결합한 정도를 조사하였다. 즉, 1차 항체인 PTH 항체와 결합한 2차 항체내의 HRP효소에 의한 ECL 반응 정도를 감지함으로써 각 단계별로 추출된 단백질내에 존재하는 PTH 및 CaSR, CHGA, Cyr61의 발현정도를 조사하였다. Of the proteins of each size delivered on the membrane, only the membrane portion corresponding to the PTH protein size is taken, and the PTH specific antibody (Ab frontier, LF-MA0140), calcium sensing receptor (CaSR, Abcam ab18200) , Chromagranin A (CHGA, AVIVA ARP-41930-T100), Cystein rich protein 61 (Cyr61, Santa Cruz SC-13100) and osteocalcin (OC, Santa Cruz SC-74495 ) Was reacted with the antibody at 4 ° C. low temperature for 14-16 hours. After the reaction with the primary antibody (diluted at 1: 1000 ratio), the primary antibody-specific secondary antibody mouse-IgG-HRP (horse radish peroxidase) conjugate was further reacted (diluted at 1: 3000 ratio). The response of the PTH antibody bound to the intracellular PTH protein antigen was examined by ECL (enhanced chemiluminescence) method. That is, the expression level of PTH, CaSR, CHGA, and Cyr61 present in the extracted protein was examined by detecting the degree of ECL reaction by HRP enzyme in the secondary antibody bound to the PTH antibody as the primary antibody.
동시에, 각 시료별로 동일한 양의 단백질을 사용하였으며, 전체 웨스턴 블롯 방법이 일관적으로 수행되었음을 증명하기 위해 각 단계별로 추출된 단백질내에서의 GAPDH (glyceraldehyde-3- phosphate dehydrogenase) 항체를 사용한 발현정도를 조사하였다. At the same time, the same amount of protein was used for each sample, and the expression level using the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) antibody in the protein extracted at each step was verified to demonstrate that the entire western blot method was performed consistently. Investigate.
각각의 특정 단백질의 발현정도는 Image J 프로그램을 이용하여 정량분석하였다. The expression level of each specific protein was quantitatively analyzed using the Image J program.
그 결과, 도 8에 나타난 대로, 분화기간이 경과함에 따라, 편도세포내 PTH 단백질의 발현이 증가하는 것으로 조사되었다. 특히 분화유도 4일이 경과한 후, 유의적으로 증가하여 7일까지 유지되는 것을 알 수 있었다. As a result, as shown in Figure 8, as the differentiation period, the expression of PTH protein in tonsil cells increased. In particular, after 4 days of eruption induction, it was found that significantly increased until 7 days.
실시예 11: 공초점 현미경을 이용한 PTH 및 PTH 분비 과립 단백질 분포 조사Example 11: Investigation of PTH and PTH Secretory Granular Protein Distribution Using Confocal Microscopy
부갑상선조직으로 분화되는 과정동안 T-MSC 세포 내부에서 PTH 및 PTH 분비 조절과 관련된 단백질의 세포내 분포양상을 공초점현미경 (Confocal Lazer Scanning Microscope, LSM-5 Pascal EXCITER, Carl Zeiss)을 이용하여 조사하였다. The intracellular distribution of proteins related to PTH and PTH secretion control in T-MSC cells during the differentiation of parathyroid tissue was investigated using a confocal lazer scanning microscope (LSM-5 Pascal EXCITER, Carl Zeiss). .
공초점현미경 관찰에 사용할 세포를 처리하기 위해, 분화하지 않은 세포와 분화한 세포를 여러 분화기간별 (7, 14, 21일)로 다양하게 준비하여 미리 cover-glass를 각각의 웰 바닥에 준비해둔 6-웰 플레이트에 각 웰 당 104 개씩 분주하였다. In order to process the cells to be used for confocal microscopy, a variety of undifferentiated and differentiated cells were prepared at various differentiation periods (7, 14, 21 days), and cover-glass was prepared at the bottom of each well in advance. 10 4 aliquots were dispensed in each well plate.
각 분화기간별 세포는 PBS로 세포표면을 세척한 후, 10% 포르말린용액으로 세포를 고정하였다. 고정된 세포는 2% BSA (bovine serum albumin)용액으로 비특이적인 단백질들을 불활성화 시킨 후, 1차 항체 (PTH, CHGA, Cyr61)를 넣은 2% BSA용액을 각 세포에 처리하여 37℃에서 4시간동안 반응시켰다. 1차 항체와의 반응 후, 반응하지 않은 항체를 PBS로 2회 세척한 후, 1차 항체에 상응하는 형광이 달린 2차 항체 (mouse IgG-FITC, rabbit-IgG-Rhodamine, Invitrogen)가 포함된 2% BSA용액을 37℃에서 1시간동안 반응시켰다. 이때 핵에 특이적으로 염색되는 DAPI (4',6-Diamidino-2-Phenylindole, Dihydrochloride) (Molecular Probes®)를 첨가하여 각 세포내의 핵을 동시에 염색하였다. 2차 항체와의 반응 종료 후, 세포표면을 다시 PBS로 2회 세척한 후, 세포가 부착되어 있는 커버글래스를 떼어내고, 형광현미경 용 글래스에 얹은 뒤, mounting 용액으로 글래스에 부착시킨 뒤, 형광현미경상에서의 각각 항체의 발현 및 세포내 분포양상을 조사하였다. After differentiation, the cells were washed with PBS, and the cells were fixed with 10% formalin solution. The fixed cells were inactivated nonspecific proteins with 2% BSA (bovine serum albumin) solution, and then treated with 2% BSA solution containing primary antibody (PTH, CHGA, Cyr61) to each cell for 4 hours at 37 ° C. Reacted for a while. After reaction with the primary antibody, the unreacted antibody was washed twice with PBS, and then the fluorescent secondary antibody (mouse IgG-FITC, rabbit-IgG-Rhodamine, Invitrogen) corresponding to the primary antibody was included. The 2% BSA solution was reacted at 37 ° C. for 1 hour. At this time, DAPI (4 ', 6-Diamidino-2-Phenylindole, Dihydrochloride) (Molecular Probes®), which is specifically stained in the nucleus, was added to simultaneously stain the nuclei in each cell. After completion of the reaction with the secondary antibody, the cell surface was washed twice with PBS, and then the cover glass to which the cells were attached was removed, placed on a glass for fluorescence microscopy, and then attached to the glass with a mounting solution. The expression and intracellular distribution of each antibody on the microscope were examined.
그 결과, 도 9에 나타난 바와 같이, 총 분화기간동안 조사한 결과, 세포내 PTH의 발현은 분화유도액을 처리하지 않은 세포군 (Control cell, C로 표기)에서는 아래의 그림처럼 PTH 분포에 큰 변화가 나타나지 않는 것으로 조사되었다. 반면, 분화유도액을 처리한 경우 (Differentiated cell, D로 표기)에서는 분화기간이 경과함에 따라, PTH의 발현이 현저히 증가함을 알 수 있었다. 그 양상은 14일에서 현저히 높은 발현 분포를 보여주어, 세포내 PTH 단백질의 발현양상 및 분비되는 PTH 농도 변화 양상과 동일한 경향을 보여주었다.As a result, as shown in Figure 9, as a result of the investigation during the total differentiation, the expression of intracellular PTH was significantly changed in PTH distribution in the cell group (labeled as Control cell, C) not treated with differentiation-inducing fluid as shown below It was investigated not to appear. On the other hand, when the differentiation induction solution (differentiated cells, denoted as D), the expression of PTH was significantly increased as the differentiation period elapsed. The pattern showed a significantly higher expression distribution at 14 days, showing the same trend as the expression pattern of intracellular PTH protein and the pattern of changes in secreted PTH concentration.
또한, 세포질내에 분산되어 분포하는 PTH의 발현양상은 과립의 형태로 나타나, 생성된 PTH의 세포 내 존재형태가 다른 분비되는 호르몬과 같이 분비과립의 형성 및 이와 유사한 기작을 통해 호르몬 저장 및 분비과정이 이뤄질 것임을 예측할 수 있었다. In addition, the expression pattern of PTH dispersed and distributed in the cytoplasm appears in the form of granules, and the hormonal storage and secretion process is performed through the formation of secretory granules and similar mechanisms like other secretory hormones. It could be predicted that it would come true.
세포 내에서의 PTH 분포와 관련하여, CHGA의 세포내 분포양상을 조사한 결과, 도 10에서와 같이 분화하지 않은 편도세포에서는 발현되지 않는 것으로 조사되었으나, 분화한 편도세포에서는 분화기간이 경과함에 따라, CHGA의 발현이 증가하고, 그 정도도 세포질내에 고르게 분포하는 양상을 보여주었다. In relation to the distribution of PTH in the cells, the intracellular distribution patterns of CHGA were found not to be expressed in undifferentiated tonsil cells as shown in FIG. 10, but as the differentiation period elapsed in the differentiated tonsil cells, The expression of CHGA was increased and evenly distributed in the cytoplasm.
PTH 단백질과 CHGA를 동시에 이중염색하여 분포양상을 조사한 결과, PTH와 CHGA의 발현부위가 일치하는 것으로 조사되어, 생성된 PTH를 저장하고 분비하는 과정에 CHGA와 직접적인 연관성이 있음을 알 수 있었다(도 11). As a result of double staining of PTH protein and CHGA at the same time, it was found that the expression sites of PTH and CHGA matched, indicating that there is a direct correlation with CHGA in the process of storing and secreting the generated PTH (Fig. 11).
실시예 12: 세포 외부 칼슘 농도에 따른 PTH 분비조절능 조사Example 12: Investigation of PTH Secretion Capacity According to Extracellular Calcium Concentration
PTH는 세포외부 칼슘 농도의 변화를 인지하여, 생체내 칼슘의 항상성을 유지하는데 작용하는 주요 호르몬이다. 편도세포에서 분화된 부갑상선 조직세포에서 생성된 PTH를 생체내 적용, 사용하기 위해서는 PTH가 PTH 고유의 기능을 정상적으로 수행하는지 여부를 조사하는 것이 중요하다. PTH is a major hormone that works to maintain homeostasis of calcium in vivo by recognizing changes in extracellular calcium concentrations. In vivo application and use of PTH produced from parathyroid tissue cells differentiated from tonsil cells, it is important to investigate whether PTH normally performs its own functions.
따라서, 편도세포를 부갑상선조직으로 분화시킨 후, 세포외부 칼슘 농도를 다양하게 조절하여 외부의 칼슘 농도 변화에 따른 PTH 분비 조절능의 변화를 조사하였다.Therefore, after the amygdala was differentiated into parathyroid tissue, the extracellular calcium concentration was variously controlled to investigate the change of PTH secretion capacity according to the external calcium concentration.
먼저, T-MSC에 분화유도제를 7일간 처리하여 부갑상선조직으로의 분화를 유도하였다. 7일간 분화를 유도한 후, 부갑상선으로 분화된 세포를 세포 배양액 내의 칼슘 농도를 정상농도 (1.5 mM), 저농도 (0.09 mM), 고농도 (3.0 mM)로 달리한 배양액으로 배양하였다. 다양한 칼슘 농도를 함유한 배양액에 배양한 T-MSC를 3, 8, 24, 48시간동안 노출시킨 뒤, 시간대 별로 세포배양액을 수집하여 외부 칼슘 농도에 따른 세포배양액으로의 PTH 분비량을 실시예 9의 ELCIA 방법으로 조사하였다. First, differentiation inducing agent was treated with T-MSC for 7 days to induce differentiation into parathyroid tissue. After inducing differentiation for 7 days, the cells differentiated into parathyroid glands were cultured in culture medium in which the calcium concentration in the cell culture medium was varied at normal concentration (1.5 mM), low concentration (0.09 mM) and high concentration (3.0 mM). After exposing the cultured T-MSC to a culture solution containing various calcium concentrations for 3, 8, 24, and 48 hours, the cell culture fluid was collected for each time period, and the amount of PTH secreted into the cell culture fluid according to the external calcium concentration was measured. It was investigated by ELCIA method.
그 결과, 도 12와 같이, 저칼슘 상태에서는 정상 칼슘 농도에 비해 고농도의 PTH 분비량을 보여주었으며, 특히 24시간 노출 이후에 그 분비량이 더욱 증가하는 것으로 조사되었다. 반면, 고칼슘 상태에서는 PTH의 분비량이 정상 칼슘농도 상태에서 보다 현저히 낮은 것으로 조사되었다. 이는 분화된 부갑상선조직 역시 세포외부 칼슘농도에 따라 PTH 분비 정도를 조절가능하고, PTH 분비 조절을 통해 칼슘의 흡수/재흡수를 조절하여 혈중 칼슘농도 항상성 유지에 기여하는 PTH 고유의 기능을 수행할 수 있음을 보여주는 결과이다. As a result, as shown in Figure 12, in the low calcium state showed a high concentration of PTH secretion compared to the normal calcium concentration, in particular, the secretion after 24 hours exposure was further increased. On the other hand, in the high calcium state, the secretion of PTH was significantly lower than the normal calcium concentration. The differentiated parathyroid tissue can also control the level of PTH secretion according to extracellular calcium concentration, and can control PTH secretion to perform the unique function of PTH, which contributes to maintaining homeostasis of calcium concentration by controlling calcium absorption / resorption. The result shows that there is.
특히, 저칼슘 상태에서는 편도세포의 세포모양 역시 분비과립 형태의 입자가 다수 형성되는 것을 보여주어, 분비과립형성에 따른 세포형태의 변화 역시 유발될 수 있음을 보여주고 있다(도 13).Particularly, in the low calcium state, the cell appearance of tonsils also shows that a large number of particles of secretory granules are formed, thereby showing that a change in cell morphology according to secretory granulation can also be induced (FIG. 13).
또한, 세포외부 칼슘농도를 인지하고, 저장된 PTH의 분비를 조절하는 것으로 알려진 인자로는 칼슘 감응 수용체 (CaSR)가 있다. CaSR는 부갑상선세포의 표면에 다수 존재하는 수용체로, 세포외부의 칼슘농도를 인지하면서 혈중 칼슘농도의 항상성을 유지, 조절하는 역할을 한다. 칼슘농도가 낮은 상태인 경우, 이를 인지하고 저장된 PTH의 세포외부로의 분비를 촉진하고, 이때 분비된 PTH는 혈액순환을 통해 골조직으로 이동하여 조골세포 표면에 존재하는 PTH 수용체 (PTH receptor, PTHR)와 결합, 반응하면서 저장된 칼슘이 혈액으로 방출되도록 함으로써 혈중 칼슘의 항상성이 유지되는 작용이 생체내에서 일어나게 된다. 이러한 작용을 하는 CaSR는 수용체 자체의 양에 의해 그 활성이 조절되는 것으로 알려져 있다. 이에, 다양한 세포외부 칼슘농도에 노출된 편도세포를 이용하여 CaSR의 발현정도 및 PTH 발현정도를 조사하여, 생성된 PTH의 CaSR와의 작용과의 연관성을 조사하였다. In addition, a factor known to recognize extracellular calcium concentrations and to control the secretion of stored PTH is calcium sensitive receptor (CaSR). CaSR is a receptor that exists on the surface of parathyroid cells, and it recognizes the extracellular calcium concentration and plays a role in maintaining and controlling the homeostasis of the calcium concentration in the blood. When the calcium concentration is low, it recognizes and promotes the secretion of the stored PTH to the outside of the cell, where the secreted PTH moves to bone tissue through blood circulation and is present on the surface of osteoblasts (PTH receptor, PTHR) In combination with the reaction, the stored calcium is released into the blood to maintain the homeostasis of calcium in the blood occurs in vivo. It is known that CaSR having such a function is regulated by the amount of the receptor itself. Therefore, the expression level of CaSR and the expression level of PTH were examined using tonsil cells exposed to various extracellular calcium concentrations, and the correlation between the generated PTH and CaSR was investigated.
그 결과, 도 14와 같이, 저칼슘 상태일때는 CaSR의 발현정도가 증가하고, 고칼슘 상태에서는 발현량이 현저히 감소하여, 편도세포에서 생성된 PTH의 정상적인 칼슘농도 항상성 조절과정에도 CaSR가 주요 인자로 작용할 수 있음을 알 수 있었다. As a result, as shown in Fig. 14, the expression level of CaSR is increased in the low calcium state, and the expression level is significantly decreased in the high calcium state, and CaSR may act as a major factor in the normal homeostasis control process of PTH produced in tonsil cells. I could see that.
실시예 13: 부갑상선조직으로 분화된 편도줄기세포의 세포내 미세구조 확인Example 13: Confirmation of intracellular microstructure of tonsil stem cells differentiated into parathyroid tissue
줄기세포를 이용하여 특정 장기로의 분화를 유도한 경우, 특정 장기 고유의 기능을 수행하기 위해서는 분화된 조직이 실제 장기 조직이나 세포와 구조적인 면에서 유사성을 갖게 된다. When stem cells are used to induce differentiation into specific organs, differentiated tissues have structural similarities with actual organ tissues or cells in order to perform specific organ specific functions.
이에, 부갑상선조직으로 분화된 편도줄기세포의 세포내 미세구조를 전자현미경을 이용하여 조사함으로써 정상 부갑상선조직과의 유사성을 조사하였다.In this regard, the intracellular microstructure of amygdala stem cells differentiated into parathyroid tissue was examined by electron microscopy to investigate similarity with normal parathyroid tissue.
부갑상선으로 분화한 T-MSC의 세포내 미세구조의 변화를 조사하기 위해 전자현미경을 이용하여 관찰하였다. 가장 최대의 PTH 분비능을 보이는 시점인 분화 7일째의 세포를 이용하여 세포내 미세구조의 변화를 조사하였다. 이때, 대조군으로는 분화유도제인 액티빈 A와 Shh를 처리하지 않은 7일째의 세포를 사용하였다. 대조군 (Control, C)과 분화 7일째 (Differentiation, D)의 세포를 PBS 를 이용하여 세포 표면을 세척한 후, trypsin을 처리하여 부착된 세포를 떼어낸 뒤, 원심분리기를 이용하여 1200 rpm에서 5분간 원심분리하여 떨어진 세포를 수집하였다. 펠렛 형태로 모은 세포는 2.5 % glutaraldehyde 용액에 보관하여 4℃에서 고정하여 보관하였다. Electron microscopy was used to investigate changes in intracellular microstructure of T-MSCs differentiated into parathyroid glands. The intracellular microstructure changes were investigated using cells at day 7 of differentiation, the point of maximum PTH secretion. At this time, as a control group, cells on day 7 which were not treated with differentiation-inducing activin A and Shh were used. After washing the cell surface of the control group (Control, C) and the 7th day of differentiation (Differentiation, D) using PBS, remove the attached cells by treatment with trypsin, and then using a centrifuge at 5 Centrifuged for a minute to collect the fallen cells. The cells collected in pellet form were stored in 2.5% glutaraldehyde solution and fixed at 4 ° C.
세포내 미세구조를 관찰하기 위해 투과전자현미경 (MODEL H-7650 (2006), H-600 (1982) / HITACHI)을 이용하여 관찰 조사하였다. 관찰한 배율은 4,000배율, 12,000배율을 이용하였다.In order to observe the intracellular microstructure, it was examined using a transmission electron microscope (MODEL H-7650 (2006), H-600 (1982) / HITACHI). The observed magnification was 4,000 times and 12,000 times.
분화하지 않은 편도줄기세포와 부갑상선으로 분화한 편도줄기세포를 비교, 관찰하였을 때, 분화하지 않은 편도줄기세포는 다른 일반세포와 마찬가지로 핵, 미토콘드리아, 골지체의 기본 구조를 보이고 있는 것으로 조사되었다 (도 15a). Comparing and observing undifferentiated tonsils and parathyroid tonsils, the undifferentiated tonsils showed the basic structure of the nucleus, mitochondria and Golgi like other normal cells (Fig. 15a). ).
반면, 부갑상선으로 분화유도된 세포에서는 기본 세포 미세구조 외에, 세포과립으로 간주되는 구조가 다수 형성되어 있음이 조사되었다 (도 15b). 또한, 과립전구체 (pro-secretory granule) 와 유사한 형태를 보이는 구조 역시 관찰되어(도 15b), PTH를 분비하는 호르몬 분비조직으로의 분화가 잘 진행되었음을 구조적으로도 확인할 수 있었다. On the other hand, in the cells induced by differentiation of the parathyroid gland, it was investigated that in addition to the basic cellular microstructures, a number of structures regarded as cell granules were formed (FIG. 15B). In addition, a structure similar to that of the pro-secretory granule (pro-secretory granule) was also observed (FIG. 15b), and structurally confirmed that the differentiation into the hormone-secreting tissue secreting PTH proceeded well.
실시예 14: 전조골세포를 이용한 조골기능 조사Example 14: Osteoblast function investigation using progenitor osteoblasts
PTH는 조골세포 표면의 PTH 수용체 (parathyroid hormone receptor, PTHR)를 통해 골형성능을 갖는다는 연구결과가 보고된 이래로, PTH 자체가 갖는 직접적인 골형성능 유도를 통한 새로운 골다공증치료제로서의 역할이 제시되어 오고 있다. Since PTH has been reported to have bone formation ability through the PTH receptor (PTHR) on the surface of osteoblasts, the role of PTH itself as a new osteoporosis treatment through direct induction of bone formation ability has been suggested.
이를 토대로, 부갑상선조직으로 분화된 편도줄기세포로부터 생성, 분비된 PTH가 실제로 PTH 고유의 골형성 능력을 갖는지 여부를 조사하였다.Based on this, we investigated whether PTH produced and secreted from amygdala stem cells differentiated into parathyroid tissues actually has PTH-specific bone formation ability.
전조골세포인 (Preosteoblast) MC3T3-E1 세포를 이용하여 PTH를 배출하는 부갑상선조직으로 분화된 T-MSC에서 얻어진 세포배양액에 의한 조골기능을 조사하였다. 즉, MC3T3-E1 세포는 24-웰 플레이트에 각 웰 당 104 개씩 분주하고, 각 웰당 아스코빅산 2-포스페이트, 덱사메타손 및 β-글리세로포스페이트를 함유한 골세포배양액 (Invtorgen)을 2주간 처리하여 조골기능을 조사하였다. 상기 배양액을 3-4일마다 교체하였으며, 세포외부로 분비된 PTH를 함유하고 있는 세포배양액 (conditioned medium, CM)을 다양한 농도로 처리하여 (세포배양액 내의 PTH함유량으로 10, 50, 100, 1000 pg/ml), 분비된 PTH의 골형성 능력을 조사하였다. 유전자재조합 방법으로 합성, 시판하는 PTH와 골다공증의 호르몬 요법으로 임상에서 사용중인 17-beta estradiol (10 μM)을 양성대조약물로 비교 조사하였다. Osteoblast function by cell culture fluid obtained from T-MSCs differentiated into parathyroid tissue releasing PTH using preosteoblast MC3T3-E1 cells. That is, MC3T3-E1 cells were seeded 10 4 per each well in 24-well plates, and processes each per well biksan ascorbyl 2-phosphate, dexamethasone and a bone cell culture medium containing a phosphate β- glycerophosphate (Invtorgen) 2 jugan Osteopathic function was investigated. The culture medium was replaced every 3-4 days, and treated with various concentrations of cell culture medium (conditioned medium, CM) containing PTH secreted out of the cell (10, 50, 100, 1000 pg in PTH content in the cell culture solution). / ml), the osteogenic ability of secreted PTH was investigated. We compared and compared 17-beta estradiol (10 μM) in clinical trials with PTH and osteoporosis hormone therapy synthesized and marketed by genetic recombination method.
골형성능을 조사하기 위해, 웨스턴 블롯 방법으로 골형성의 대표적인 표지자인 오스테오칼신 (OC) 단백질의 세포질내 발현 및 세포질내 칼슘의 축적정도를 염색하는 Alizarin red S 방법을 이용하여 조사하였다. To investigate the osteogenic ability, Western blot analysis was performed using the Alizarin red S method, which stains the cytoplasmic expression and cytoplasmic calcium accumulation of osteocalcin (OC) protein, a representative marker of bone formation.
즉, 배양이 종료된 후, 배양액을 제거하고, 각 웰의 바닥에 부착된 세포를 PBS로 세척한 다음, 60% 이소프로필알콜을 사용하여 5분동안 고정하며, 증류수로 2회 세척하고, 2% Alizarin red S 수용액(pH 4.2)로 3분간 염색하였다. 염색이 종료된 후, Alizarin red S 수용액을 제거하고 즉시 증류수로 3회 세척한 다음, 위상차 현미경으로 관찰하여 대조군 (분화를 유도하지 않고 배양한 MC3T3-E1)과 비교하였다 That is, after the incubation is completed, the culture solution is removed, and the cells attached to the bottom of each well are washed with PBS, and then fixed with 60% isopropyl alcohol for 5 minutes, washed twice with distilled water, 2 It was stained for 3 minutes with% Alizarin red S aqueous solution (pH 4.2). After staining was completed, the Alizarin red S aqueous solution was removed and immediately washed three times with distilled water, and then compared with a control group (MC3T3-E1 cultured without inducing differentiation) by observing with a phase contrast microscope.
대표적인 골형성 표지자인 osteocalcin 단백질의 발현 정도를 조사한 결과, CM의 농도가 증가함에 따라, 오스테오칼신의 발현 역시 증가하는 양상을 보였으며, 그 정도는 호르몬인 에스트로겐 보다는 낮은 발현정도를 보였으나, 분화유도액을 처리한 경우와 유사한 정도의 골형성능을 보여주었다 (도 16). 또한, 시판 PTH (104 ng/ml)를 처리한 경우와 비교할 때, 시판 PTH 농도의 1/100에 해당하는 농도의 CM (1000 pg/ml = 1 ng/ml)을 처리한 경우에서도 시판 PTH와 유사한 골형성능을 보여주어, 편도줄기세포에서 생성, 분비된 PTH가 높은 생리활성을 보유하고 있음을 확인하였다. As a result of examining the expression level of osteocalcin protein, which is a representative bone formation marker, the expression of osteocalcin also increased as the concentration of CM increased, which was lower than that of the hormone estrogen. It showed a similar degree of bone formation ability as treated (Fig. 16). In addition, compared to the case of treatment with the commercial PTH (104 ng / ml), even when treated with a CM (1000 pg / ml = 1 ng / ml) at a concentration corresponding to 1/100 of the commercial PTH concentration It showed similar bone formation ability, and it was confirmed that PTH produced and secreted in tonsil stem cells has high physiological activity.
Alizarin red-S 염색법을 이용하여 세포내 석회화된 부분을 염색한 결과에서도, CM에 의해 골형성이 유도되었으며, 석회화되어 염색된 부분이 현저히 증가하였음을 확인할 수 있었다.As a result of staining the intracellular calcified portion using Alizarin red-S staining, bone formation was induced by CM, and the calcified portion was markedly increased.
상기 결과는 본 발명의 방법에 의해 생산된 PTH는 생체내에 적용시 PTH 고유의 생리학적 기능 수행이 가능하여, 치료제로 활용될 수 있음을 보여준다. The results show that PTH produced by the method of the present invention is capable of performing physiological functions unique to PTH when applied in vivo, and thus can be used as a therapeutic agent.
모든 데이터는 평균 ± 표준편차(S.D.)로 표시하고, 통계적으로 유의성 검증은 GRAPHPAD PRISM 소프트웨어(GraphPad Software Inc., San Diego, CA)를 이용하여 Student’s t-test로 분석하였다. 모든 분석에서, P<0.05인 경우를 통계적으로 유의한 것으로 간주하였다. 모든 실험은 최소 3회 반복 수행하였다.All data are expressed as mean ± standard deviation (S.D.), and statistically significant tests were analyzed by Student's t-test using GRAPHPAD PRISM software (GraphPad Software Inc., San Diego, Calif.). In all analyzes, cases with P <0.05 were considered statistically significant. All experiments were performed at least three times.
이상의 설명으로부터, 본 발명이 속하는 기술분야의 당업자는 본 발명이 그 기술적 사상이나 필수적 특징을 변경하지 않고서 다른 구체적인 형태로 실시될 수 있다는 것을 이해할 수 있을 것이다. 이와 관련하여, 이상에서 기술한 실시 예들은 모든 면에서 예시적인 것이며 한정적인 것이 아닌 것으로서 이해해야만 한다. 본 발명의 범위는 상기 상세한 설명보다는 후술하는 특허 청구범위의 의미 및 범위 그리고 그 등가 개념으로부터 도출되는 모든 변경 또는 변형된 형태가 본 발명의 범위에 포함되는 것으로 해석되어야 한다.From the above description, those skilled in the art will appreciate that the present invention can be implemented in other specific forms without changing the technical spirit or essential features. In this regard, the embodiments described above are to be understood in all respects as illustrative and not restrictive. The scope of the present invention should be construed that all changes or modifications derived from the meaning and scope of the following claims and equivalent concepts rather than the detailed description are included in the scope of the present invention.

Claims (34)

  1. (i) 별개의 기증자로부터 분리된 각 편도조직으로부터 단핵세포를 각각 수득하는 단계; (i) obtaining mononuclear cells from each tonsil isolated from a separate donor;
    (ii) 상기 수득한 단핵세포를 배양하여 중간엽 줄기세포를 선별하는 단계; 및, (ii) culturing the obtained mononuclear cells to select mesenchymal stem cells; And,
    (iii) 상기 선별된 중간엽 줄기세포를 혼합하여 배양하는 단계를 포함하는, 키메라 중간엽 줄기세포군의 제조방법. (iii) mixing the selected mesenchymal stem cells and culturing, the method for producing a chimeric mesenchymal stem cell group.
  2. 제1항의 방법에 의하여 제조되고, 하기의 특성을 나타내는 키메라 중간엽 줄기세포군:A chimeric mesenchymal stem cell group prepared by the method of claim 1 and exhibiting the following characteristics:
    (i) 별개의 기증자로부터 수득한 별개의 편도조직으로부터 분리된 중간엽 줄기세포를 포함하고;(i) comprises mesenchymal stem cells isolated from separate tonsils obtained from separate donors;
    (ii) 단일 기증자로부터 수득한 편도조직 유래 중간엽 줄기세포와 동일한 세포활성을 나타내며;(ii) exhibit the same cellular activity as tonsil-derived mesenchymal stem cells obtained from a single donor;
    (iii) CD14, CD34 및 CD45의 발현이 저하되고;(iii) the expression of CD14, CD34 and CD45 is reduced;
    (iv) CD73, CD95 및 CD105의 발현이 증가하며;(iv) the expression of CD73, CD95 and CD105 is increased;
    (v) 면역관련 표현 항체에 대하여 음성을 나타내고;(v) negative for immunorelevant expressing antibodies;
    (vi) 중배엽성 세포로 분화될 수 있으며; 및(vi) can differentiate into mesodermal cells; And
    (vii) 내배엽성 세포로 분화될 수 있다. (vii) can differentiate into endoderm cells.
  3. 제2항에 있어서,The method of claim 2,
    상기 면역관련 표현 항체는 HLA-DR, CD40, CD80, CD86 또는 이들의 조합인 것인 키메라 중간엽 줄기세포군.The immunologically expressing antibody is HLA-DR, CD40, CD80, CD86 or a combination thereof chimeric mesenchymal stem cell population.
  4. 제2항에 있어서,The method of claim 2,
    상기 중배엽성 세포는 지방세포, 골세포, 연골세포 또는 이들의 조합인 것인 키메라 중간엽 줄기세포군.The mesoderm cells are adipocytes, bone cells, chondrocytes or a combination thereof chimeric mesenchymal stem cell group.
  5. 제2항에 있어서,The method of claim 2,
    상기 내배엽성 세포는 부갑상선 조직세포인 것인 키메라 중간엽 줄기세포군.The endoderm cells are parathyroid tissue cells chimeric mesenchymal stem cell group.
  6. 제2항의 키메라 중간엽 줄기세포군을 냉동 및 해동하는 단계를 포함하는 증식능이 향상된 키메라 중간엽 줄기세포군의 제조방법.The method of producing a chimeric mesenchymal stem cell group having improved proliferative capacity, comprising freezing and thawing the chimeric mesenchymal stem cell group of claim 2.
  7. 제6항에 있어서,The method of claim 6,
    상기 냉동은 냉각매체, 동결보호제 또는 이들의 조합을 이용하여 수행되는 것인 방법.Wherein the freezing is performed using a cooling medium, a cryoprotectant or a combination thereof.
  8. 제7항에 있어서,The method of claim 7, wherein
    상기 냉각매체는 액체질소인 것인 방법.The cooling medium is liquid nitrogen.
  9. 제7항에 있어서,The method of claim 7, wherein
    상기 동결보호제는 글라이세롤, 설탕, 글루코스 또는 이들의 조합인 것인 방법.The cryoprotectant is glycerol, sugar, glucose or a combination thereof.
  10. 제6항의 방법으로 제조되고, 하기의 특성을 나타내는 증식능이 향상된 키메라 중간엽 줄기세포군:A chimeric mesenchymal stem cell group prepared by the method of claim 6 and having an improved proliferative capacity having the following characteristics:
    (i) 냉동 및 해동을 수행하지 않은 키메라 중간엽 줄기세포군와 동등한 수준의 분화능을 나타내고; 및,(i) exhibits the same level of differentiation as the chimeric mesenchymal stem cell population that did not undergo freezing and thawing; And,
    (ii) 냉동 및 해동을 수행하지 않은 키메라 중간엽 줄기세포군보다 향상된 수준의 증식능을 나타낸다.(ii) show an enhanced level of proliferative capacity than the chimeric mesenchymal stem cell population that did not perform freezing and thawing.
  11. 제2항의 키메라 중간엽 줄기세포군 또는 제10항의 증식능이 향상된 키메라 중간엽 줄기세포군을 덱사메타손, 아스코빅 산, L-프롤린, TGF-3, BMP-6 및 ITS를 함유한 연골세포 분화배지에서 배양하는 단계를 포함하는 키메라 중간엽 줄기세포군을 연골세포로 분화시키는 방법.The chimeric mesenchymal stem cell group of claim 2 or the chimeric mesenchymal stem cell group having improved proliferative capacity of claim 10 are cultured in chondrocyte differentiation medium containing dexamethasone, ascorbic acid, L-proline, TGF-3, BMP-6 and ITS. Chimera mesenchymal stem cell group comprising the step of differentiating into chondrocytes.
  12. 제2항의 키메라 중간엽 줄기세포군 또는 제10항의 증식능이 향상된 키메라 중간엽 줄기세포군을 아스코빅산 2-포스페이트, 덱사메타손 및 β-글리세로포스페이트를 함유한 배양배지에 배양하는 단계를 포함하는 키메라 중간엽 줄기세포군을 골세포로 분화시키는 방법.A chimeric mesenchymal stem cell group according to claim 2 or a chimeric mesenchymal stem cell group having improved proliferative capacity according to claim 10, comprising a step of culturing a chimeric mesenchymal stem cell comprising a culture medium containing ascorbic acid 2-phosphate, dexamethasone and β-glycerophosphate. Method of differentiating cell population into osteocytes.
  13. 제2항의 키메라 중간엽 줄기세포군 또는 제10항의 증식능이 향상된 키메라 중간엽 줄기세포군을 3-이소부틸-1-메틸-젠틴, 덱사메타손, 인도메타신 및 인슐린을 함유한 배양배지에 배양하는 단계를 포함하는 키메라 중간엽 줄기세포군을 지방세포로 분화시키는 방법.The method of claim 2, comprising the step of culturing the chimeric mesenchymal stem cell group of claim 2 or the chimeric mesenchymal stem cell group having improved proliferative capacity of claim 10 in a culture medium containing 3-isobutyl-1-methyl-gentine, dexamethasone, indomethacin and insulin. Method of differentiating chimeric mesenchymal stem cell population to adipocytes.
  14. 제2항의 키메라 중간엽 줄기세포군 또는 제10항의 증식능이 향상된 키메라 중간엽 줄기세포군을 우태아혈청(FBS), 액티빈 A(activin A) 및 Shh(sonic hedgehog)를 함유한 배양배지에 배양하는 단계를 포함하는 키메라 중간엽 줄기세포군을 부갑상선 조직세포로 분화시키는 방법.Cultivating the chimeric mesenchymal stem cell group of claim 2 or the chimeric mesenchymal stem cell group having improved proliferative capacity of claim 10 in a culture medium containing fetal bovine serum (FBS), activin A (activin A) and Shh (sonic hedgehog) Method for differentiating chimeric mesenchymal stem cell group comprising a parathyroid tissue cells.
  15. 제2항의 키메라 중간엽 줄기세포군 또는 제10항의 증식능이 향상된 키메라 중간엽 줄기세포군으로부터 분리된 줄기세포, 또는 상기 키메라 중간엽 줄기세포군으로부터 분화된 세포를 유효성분으로 포함하는 세포 치료제.A cell therapeutic agent comprising stem cells isolated from the chimeric mesenchymal stem cell group of claim 2 or the chimeric mesenchymal stem cell group having improved proliferative capacity of claim 10, or cells differentiated from the chimeric mesenchymal stem cell group.
  16. 액티빈(Activin) A 및 소닉 헤지호그(Sonic hedgehog)(Shh)를 포함하는 배지에서 편도줄기세포를 배양하는 단계를 포함하는, 편도줄기세포로부터 부갑상선 조직세포를 분화시키는 방법. A method of differentiating parathyroid tissue cells from tonsils, comprising culturing tonsils in a medium comprising Activin A and Sonic hedgehog (Shh).
  17. 제16항에 있어서, 상기 액티빈 A 및 소닉 헤지호그는 각각 50 ng/ml 내지 300 ng/ml의 농도로 배지에 존재하는 것인 방법.The method of claim 16, wherein the activin A and sonic hedgehog are each present in the medium at a concentration of 50 ng / ml to 300 ng / ml.
  18. 제17항에 있어서, 상기 액티빈 A 및 소닉 헤지호그는 각각 100 ng/ml의 농도로 배지에 존재하는 것인 방법.18. The method of claim 17, wherein the activin A and sonic hedgehog are each present in the medium at a concentration of 100 ng / ml.
  19. 제16항에 있어서, 상기 배지는 DMEM(Dulbecco's modified Eagle medium) 배지인 방법.The method of claim 16, wherein the medium is Dulbecco's modified Eagle medium (DMEM) medium.
  20. 제16항에 있어서, 상기 배양은 4일 이상 실시되는 것인 방법.The method of claim 16, wherein the culturing is carried out for at least 4 days.
  21. 제16항에 있어서, 상기 편도줄기세포는 중간엽줄기세포인 방법.The method of claim 16, wherein the amygdala stem cells are mesenchymal stem cells.
  22. 제16항에 있어서, 배양이 4일 이상 이루어질 경우, 4 내지 5일 마다 신선한 배지로 교체하는 단계를 추가로 포함하는 방법.The method of claim 16, further comprising the step of replacing with fresh medium every 4 to 5 days if the cultivation is for 4 days or more.
  23. i) 액티빈(Activin) A 및 소닉 헤지호그(Sonic hedgehog)(Shh)를 포함하는 배지에서 편도줄기세포를 배양하여 부갑상선 조직세포로 분화시키는 단계 및 ii) i) 단계에서 수득한 배양액 또는 세포 용해액으로부터 부갑상선 호르몬을 분리하는 단계를 포함하는, 편도줄기세포를 이용하여 부갑상선 호르몬을 생산하는 방법.i) culturing tonsilocytes in a medium comprising Activin A and Sonic hedgehog (Shh) to differentiate into parathyroid tissue cells and ii) culture or cell lysis obtained in step i) A method of producing parathyroid hormone using tonsilocytes, comprising the step of separating parathyroid hormone from fluid.
  24. 제23항에 있어서, i) 단계 후에, 배양액의 칼슘 농도를 조절하여 부갑상선 호르몬을 세포 밖으로 분비시키는 단계를 추가로 포함하는 방법.The method of claim 23, further comprising, after step i) adjusting the calcium concentration of the culture to secrete parathyroid hormone out of the cell.
  25. 제24항에 있어서, i) 단계 후에, 배양액의 칼슘 농도를 0.5 mM 이하로 조절하는 단계를 추가로 포함하는 방법. The method of claim 24 further comprising, after step i), adjusting the calcium concentration of the culture to 0.5 mM or less.
  26. 제23항에 있어서, 상기 액티빈 A 및 소닉 헤지호그는 각각 50 ng/ml 내지 300 ng/ml의 농도로 배지에 존재하는 것인 방법.The method of claim 23, wherein the activin A and sonic hedgehog are each present in the medium at a concentration of 50 ng / ml to 300 ng / ml.
  27. 제23항에 있어서, 상기 액티빈 A 및 소닉 헤지호그는 각각 100 ng/ml 의 농도로 배지에 존재하는 것인 방법.The method of claim 23, wherein the activin A and sonic hedgehog are each present in the medium at a concentration of 100 ng / ml.
  28. 제23항에 있어서, 상기 배양은 4일 이상 실시되는 것인 방법.The method of claim 23, wherein the culturing is carried out for at least 4 days.
  29. 제23항에 있어서, 상기 배양은 7일 이상 실시되는 것인 방법.The method of claim 23, wherein the culturing is carried out for at least 7 days.
  30. 편도줄기세포로부터 분화된 부갑상선 조직세포.Parathyroid tissue cells differentiated from tonsil stem cells.
  31. 제30항에 있어서, 제16항 내지 제22항 중 어느 한 항의 방법에 의해 분화된 것인 부갑상선 조직세포.The parathyroid tissue cell according to claim 30, which is differentiated by the method of any one of claims 16 to 22.
  32. 제30항에 있어서, 부갑상선 호르몬을 생산하는 것인 부갑상선 조직세포.32. The parathyroid tissue cell of claim 30 which produces parathyroid hormone.
  33. 편도줄기세포로부터 분화된 부갑상선 조직세포를 포함하는 부갑상선 기능저하증 또는 골다공증의 치료를 위한 세포 치료제.A cell therapeutic agent for the treatment of hypothyroidism or osteoporosis comprising parathyroid tissue cells differentiated from tonsil stem cells.
  34. 편도줄기세포로부터 분화된 부갑상선 조직 세포를 포함하는 세포치료제를 부갑상선 기능저하증 또는 골다공증이 의심되는 개체에게 투여하는 단계를 포함하는, 부갑상선 기능저하증 또는 골다공증의 치료 방법.A method of treating parathyroid hypoplasia or osteoporosis, comprising administering a cell therapy comprising parathyroid tissue cells differentiated from tonsil stem cells to an individual suspected of hypothyroidism or osteoporosis.
PCT/KR2013/003644 2012-04-27 2013-04-26 Chimeric mesenchymal stem cell population and preparation method therefor, and method for producing parathyroid hormone using tonsil-derived stem cells WO2013162330A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20120044970A KR101508413B1 (en) 2012-04-27 2012-04-27 Chimeric mesenchymal stem cell population and process for preparing the same
KR10-2012-0044970 2012-04-27
KR10-2013-0045033 2013-04-23
KR1020130045033A KR101520531B1 (en) 2013-04-23 2013-04-23 Method of producing parathyroid hormone from tonsil-derived mesenchymal stem cell

Publications (1)

Publication Number Publication Date
WO2013162330A1 true WO2013162330A1 (en) 2013-10-31

Family

ID=49483535

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2013/003644 WO2013162330A1 (en) 2012-04-27 2013-04-26 Chimeric mesenchymal stem cell population and preparation method therefor, and method for producing parathyroid hormone using tonsil-derived stem cells

Country Status (1)

Country Link
WO (1) WO2013162330A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2581023C1 (en) * 2015-02-11 2016-04-10 Федеральное государственное бюджетное научное учреждение "Иркутский научный центр хирургии и травматологии" Method for correction of postoperative hypoparathyroidism
CN108990964A (en) * 2018-08-09 2018-12-14 华东理工大学 Cells frozen storing liquid
CN110387350A (en) * 2018-04-18 2019-10-29 江苏齐氏生物科技有限公司 A kind of isolation and culture of Marrow Mesenchymal Stem Cells
RU2806978C1 (en) * 2022-12-27 2023-11-08 Федеральное государственное бюджетное образовательное учреждение высшего образования "Сибирский государственный медицинский университет" Министерства здравоохранения Российской Федерации Method for intrathyroidal autotransplantation of parathyroid gland tissue

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060182724A1 (en) * 2005-02-15 2006-08-17 Riordan Neil H Method for expansion of stem cells
KR20090069013A (en) * 2007-12-24 2009-06-29 동국대학교 산학협력단 Chondrogenesis of bone marrow mesenchymal stem cells
US20120088272A1 (en) * 2009-06-10 2012-04-12 Bts Research International Pty Ltd Methods of generating hybrid/chimeric cells, and uses thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060182724A1 (en) * 2005-02-15 2006-08-17 Riordan Neil H Method for expansion of stem cells
KR20090069013A (en) * 2007-12-24 2009-06-29 동국대학교 산학협력단 Chondrogenesis of bone marrow mesenchymal stem cells
US20120088272A1 (en) * 2009-06-10 2012-04-12 Bts Research International Pty Ltd Methods of generating hybrid/chimeric cells, and uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JANJANIN, SASA ET AL.: "Human palatine tonsil: a new potential tissue source of multipotent mesenchymal progenitor cells", ARTHRITIS RESEARCH & THERAPY, vol. 10, no. 4, 28 July 2008 (2008-07-28), pages 1 - 12 *
RYU, KYUNG-HA ET AL.: "Tonsil-derived mesenchymal stromal cells: evaluation of biologic, immunologic and genetic factors for successful banking", CYTOTHERAPY, vol. 14, no. 10, 17 August 2012 (2012-08-17), pages 1193 - 1202 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2581023C1 (en) * 2015-02-11 2016-04-10 Федеральное государственное бюджетное научное учреждение "Иркутский научный центр хирургии и травматологии" Method for correction of postoperative hypoparathyroidism
CN110387350A (en) * 2018-04-18 2019-10-29 江苏齐氏生物科技有限公司 A kind of isolation and culture of Marrow Mesenchymal Stem Cells
CN108990964A (en) * 2018-08-09 2018-12-14 华东理工大学 Cells frozen storing liquid
CN108990964B (en) * 2018-08-09 2022-01-28 华东理工大学 Cell cryopreservation liquid
RU2806978C1 (en) * 2022-12-27 2023-11-08 Федеральное государственное бюджетное образовательное учреждение высшего образования "Сибирский государственный медицинский университет" Министерства здравоохранения Российской Федерации Method for intrathyroidal autotransplantation of parathyroid gland tissue
RU2817205C1 (en) * 2023-04-13 2024-04-11 Государственное бюджетное учреждение здравоохранения города Москвы городская клиническая больница имени С.П. Боткина департамента здравоохранения города Москвы (ГБУЗ ГКБ им. С.П. Боткина ДЗМ) Method of treating postoperative hypoparathyroidism after surgery in patients with secondary hyperparathyroidism

Similar Documents

Publication Publication Date Title
WO2013094988A1 (en) Method for producing natural killer cells, natural killer cells produced thereby, and composition for treating cancers and infectious diseases containing the same
WO2012026712A4 (en) Pharmaceutical composition for preventing or treating immune diseases or inflammatory diseases, containing stem cells treated with nod2 agonist or cultured product thereof
WO2019107913A1 (en) Method for improving quality of therapeutic cell through real-time glutathione measurement
WO2011049414A2 (en) Method for inducing migration of adult stem cells derived from adipose tissue
WO2018124642A9 (en) Composition for differentiation of dental pulp stem cells into odontoblast progenitor cells and igg or igm type monoclonal antibody specifically binding to surface of odontoblast progenitor cells
WO2014181954A1 (en) Culture medium composition for improving regenerative capacity of stem cells, and stem cell culturing method using same
WO2019083281A2 (en) Novel musculoskeletal stem cell
WO2021045374A1 (en) Medium composition for differentiation of proliferative liver organoid and method for preparing liver organoid using same
WO2013162330A1 (en) Chimeric mesenchymal stem cell population and preparation method therefor, and method for producing parathyroid hormone using tonsil-derived stem cells
WO2017155166A1 (en) Cell reprogramming method using imposition of physical stimulation-mediated environmental transition
WO2022025559A1 (en) Composition including stem cell-derived exosome, and method for producing same
WO2019216623A1 (en) Cellular vaccine having immune tolerance for treatment of diabetes and obesity and method for producing insulin-secreting cells
WO2021033990A1 (en) Composition comprising exosomes derived from induced pluripotent stem cell-derived mesenchymal stem cell precursor for prevention or treatment of non-alcoholic steatohepatitis
WO2022004938A1 (en) Method for preparing mesenchymal-like stem cells
WO2011102680A2 (en) Cd49f promoting proliferation, multipotency and reprogramming of adult stem cells through pi3k/akt/gsk3 pathway
WO2019050350A2 (en) Stem cell-derived sertoli-like cell, preparation method therefor, and use thereof
WO2016117960A1 (en) Mesenchymal stem cells over-expressed by grim19 efficacious in treating immune disease, and use thereof
Wang et al. Immunogenicity and immune modulation of osteogenic differentiated mesenchymal stem cells from Banna minipig inbred line
WO2022030973A1 (en) Cell therapeutic composition comprising t memory stem cells dedifferentiated from tumor-infiltrating lymphocytes, and method for producing composition
WO2015023165A1 (en) Inflammation-controlling composite and stabilized mesenchymal stem cells having optimized immunity control function by blocking stat3 signal molecule
WO2019059713A2 (en) Method for producing natural killer cell and use thereof
WO2018008941A1 (en) Complex comprising cartilage cell-free lysate and stem cell for promoting cartilage differentiation and use thereof
WO2021187911A1 (en) Cord blood plasma-derived exosome or mimetic thereof and pharmaceutical use thereof
WO2021091185A1 (en) Composition for cell culture, comprising paracrine factor
WO2020122498A1 (en) Pharmaceutical composition for treating pancreatitis, comprising clonal stem cells

Legal Events

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

Ref document number: 13781434

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13781434

Country of ref document: EP

Kind code of ref document: A1