WO2020121800A1 - Mesenchymal stem cell, therapeutic agent for immune disease, and anti-inflammatory agent - Google Patents

Mesenchymal stem cell, therapeutic agent for immune disease, and anti-inflammatory agent Download PDF

Info

Publication number
WO2020121800A1
WO2020121800A1 PCT/JP2019/046098 JP2019046098W WO2020121800A1 WO 2020121800 A1 WO2020121800 A1 WO 2020121800A1 JP 2019046098 W JP2019046098 W JP 2019046098W WO 2020121800 A1 WO2020121800 A1 WO 2020121800A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
mesenchymal stem
kynurenine
stem cells
culture
Prior art date
Application number
PCT/JP2019/046098
Other languages
French (fr)
Japanese (ja)
Inventor
輝 黒木
真代 湯本
登紀子 長村
Original Assignee
ロート製薬株式会社
国立大学法人東京大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by ロート製薬株式会社, 国立大学法人東京大学 filed Critical ロート製薬株式会社
Priority to JP2020559070A priority Critical patent/JP7464956B2/en
Priority to CN201980081889.1A priority patent/CN113164770A/en
Publication of WO2020121800A1 publication Critical patent/WO2020121800A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology

Definitions

  • the present invention relates to mesenchymal stem cells, therapeutic agents for immune diseases and anti-inflammatory agents.
  • Immunodeficiency diseases In immunodeficiency diseases, the inability of the immune system to function normally causes infectious diseases, recurrent episodes, severe symptoms, and prolonged symptoms. Immunodeficiency disorders impair the ability of the immune system to protect the body from invasion by external enemies such as bacteria, viruses, and fungi, and attack of abnormal cells such as cancer cells. As a result, they suffer from infectious diseases and cancers caused by bacteria, viruses, and fungi, which would not occur if their immune functions were normal.
  • Immunodeficiency diseases include congenital (primary) immunodeficiency diseases that are already present at birth, and acquired (secondary) immunodeficiency diseases that are developed due to some disease in later years. .. Acquired immunodeficiency disease often develops as a result of long-term severe disease, such as cancer, aplastic anemia, leukemia, myelofibrosis, renal failure, diabetes, liver disease, splenic disease. are exemplified.
  • autoimmune diseases are more likely to occur.
  • various autoimmune diseases and various cells and tissues are targeted for attack.
  • suppression of the immune system to control an autoimmune reaction has been performed.
  • many of the drugs used to control the autoimmune reaction also impede the body's ability to fight infectious diseases and the like, and therefore there is a demand for the development of a new therapeutic drug having only the action of controlling the autoimmune reaction.
  • Mesenchymal stem cells are pluripotent progenitor cells first isolated from bone marrow by Friedenstein in 1982 (see Non-Patent Document 1). It has been revealed that mesenchymal stem cells are present in various tissues such as bone marrow, umbilical cord, and fat, and mesenchymal stem cell transplantation is expected as a new therapeutic method for various intractable diseases ( See Patent Documents 1 and 2). Recently, it is known that stromal cells such as adipose tissue, placenta, umbilical cord and egg membrane have cells having equivalent functions. Therefore, the mesenchymal stem cells are sometimes referred to as stromal cells (Mesenchymal Stromal Cell).
  • the present invention aims to provide a novel therapeutic agent for an immune disease and a novel therapeutic agent for an anti-inflammatory agent in the above situation.
  • mesenchymal stem cells are characterized by high kynurenine production or kynurenine secretion. They have found that they are effective in treating immune diseases and inflammatory diseases, and completed the present invention.
  • a therapeutic agent effective for treating immune diseases and inflammatory diseases can be provided. That is, the gist of the present invention is as follows.
  • a mesenchymal stem cell having a high kynurenine production amount or a high kynurenine secretion amount [2] A mesenchymal stem cell characterized by having a high kynurenine production amount or kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFN ⁇ treatment. [3] A mesenchymal stem cell characterized in that the JAK/STAT pathway is activated in a steady state. [4] The mesenchymal stem cell according to any one of [1] to [3], which is derived from umbilical cord tissue.
  • a therapeutic agent for an immune disease containing the mesenchymal stem cell according to any one of [1] to [4].
  • a method for treating an immune disease which comprises using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount.
  • a method for treating an immune disease which comprises using mesenchymal stem cells having a high kynurenine production amount or a kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFN ⁇ treatment.
  • a method for treating an immune disease which comprises using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state.
  • the method for treating an immune disease according to any of [8] to [10], wherein the mesenchymal stem cells are derived from umbilical cord tissue.
  • a method for treating an inflammatory disease which comprises using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount.
  • a method for treating an inflammatory disease which comprises using mesenchymal stem cells which have a high kynurenine production amount or a kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFN ⁇ treatment.
  • a method for treating an inflammatory disease which comprises using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state.
  • a method for treating GVHD which comprises using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount.
  • a method for treating GVHD which comprises using mesenchymal stem cells that have a high kynurenine production amount or a kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFN ⁇ treatment.
  • a method for treating GVHD which comprises using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state.
  • the method for treating GVHD according to any of [16] to [18], wherein the mesenchymal stem cells are derived from umbilical cord tissue.
  • a novel therapeutic agent for immune diseases and anti-inflammatory agent and a novel method for treating immune diseases and a method for treating inflammatory diseases can be provided.
  • FIG. 1 is a diagram showing a comparison of intracellular kynurenine amounts of flat culture cells (ADH) and suspension culture cells (SUS) of mesenchymal stem cells.
  • FIG. 2 is a diagram showing a comparison of the amount of kynurenine in the culture supernatants of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS).
  • FIG. 3 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS).
  • FIG. 1 is a diagram showing a comparison of intracellular kynurenine amounts of flat culture cells (ADH) and suspension culture cells (SUS) of mesenchymal stem cells.
  • FIG. 2 is a diagram showing a comparison of the amount of kynurenine in the culture supernatants of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells
  • FIG. 4 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatants of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS).
  • FIG. 5 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatants of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS).
  • FIG. 6 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS).
  • FIG. 7 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS).
  • FIG. 8 is a view showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of flat culture cells.
  • FIG. 9 is a diagram showing the analysis results (ratio of the expression level in SUS to the expression level in ADH) of gene expression by microarray of flat culture cells (ADH) of mesenchymal stem cells and suspension culture cells (SUS).
  • FIG. 10 is a diagram showing the effect of UC-MSC (EXP-SUS) administration in GVHD model mice.
  • FIG. 11 is a diagram showing the effect of UC-MSC (EXP-SUS) administration in GVHD model mice.
  • FIG. 12 is a graph showing the anti-inflammatory effect of mesenchymal stem cell planar cultured cells (ADH) and suspension cultured cells (SUS).
  • FIG. 13 is a diagram showing the JAK/STAT signal pathway.
  • the mesenchymal stem cells, therapeutic agents for immune diseases and anti-inflammatory agents of the present invention will be described in detail below.
  • mesenchymal stem cells are characterized by high kynurenine production or kynurenine secretion.
  • Kynurenine is a metabolite produced by the enzyme indoleamine 2,3-dioxygenase (IDO) when niacin is synthesized from tryptophan in cells. It is known that tryptophan is important for T cell proliferation, and tryptophan is converted to kynurenine by IDO activity and depleted to suppress T cell proliferation and induce immunological tolerance.
  • IDO indoleamine 2,3-dioxygenase
  • the amount of kynurenine produced or the amount of kynurenine secreted is the kynurenine released from the outside of cells after being synthesized from tryptophan intracellularly by indolamine 2,3-dioxygenase (IDO) induced by TNF ⁇ or IFN ⁇ treatment under inflammatory conditions. Means quantity.
  • IDO indolamine 2,3-dioxygenase
  • the mesenchymal stem cells of the present invention need only express kynurenine at a higher level than other cells.
  • the mesenchymal stem cells of the present invention are IDO that is induced intracellularly by IFN ⁇ treatment. Therefore, the amount of kynurenine produced from tryptophan or the amount of kynurenine secreted is high.
  • the mesenchymal stem cells of the present invention need only highly express kynurenine as compared with mesenchymal stem cells obtained by culturing in a conventional medium (for example, MEM ⁇ medium containing 10% serum), and preferably 10 1.1 times or more, more preferably 1.2 times or more, still more preferably 1.5 times or more, kynurenine production amount or kynurenine secretion compared with mesenchymal stem cells obtained under culture conditions using MEM ⁇ medium containing 10% serum The higher the amount, the better.
  • a conventional medium for example, MEM ⁇ medium containing 10% serum
  • the mesenchymal stem cells of the present invention need only highly express kynurenine as compared with other cells, and examples thereof include MRC-5 cells, human neonatal skin fibroblasts (HDF), or human umbilical vein veins. It is sufficient that kynurenine is highly expressed as compared with endothelial cells (HUVEC), preferably 5 times or more, more preferably 10 times or more kynurenine production amount or kynurenine secretion amount is higher than MRC-5 cells, HDF or HUVEC. Good.
  • kynurenine is highly expressed, and preferably mesenchymal stem cells obtained under conventional culture conditions. 2 times or more, more preferably 5 times or more, further preferably 10 times or more, particularly preferably 100 times or more, as long as the amount of kynurenine production or the amount of kynurenine secretion is high.
  • the planar culture method may be performed by static culture in which cells are adhered to a flat surface such as a flask or a petri dish, and does not particularly specify a container, a medium or the like.
  • the high amount of kynurenine produced or the amount of kynurenine secreted above includes high expression or high activity of IDO involved in kynurenine synthesis.
  • the high expression or high activity of IDO is also defined in the same manner as above, and the mesenchymal stem cells of the present invention are mesenchymal cells obtained by culturing in a conventional medium (for example, 10% FBS-containing MEM- ⁇ medium). It is sufficient that IDO is highly expressed and/or IDO is highly active as compared to the lineage stem cell. It is preferably 1.1 times or more, more preferably 1.2 times or more, further preferably 1.5 times or more, as compared with mesenchymal stem cells obtained under conventional culture conditions, with high expression and/or activity of IDO ing.
  • the mesenchymal stem cells of the present invention have higher IDO expression and/or activity than other cells such as MRC-5 cells, human neonatal skin fibroblasts (HDF), and human umbilical vein endothelial cells (HUVEC). Is increasing. Higher expression or activity of IDO than MRC-5 cells, HDF or HUVEC, preferably higher than MRC-5 cells, HDF or HUVEC by 5 times or more, more preferably 10 times or more high expression of IDO and/or The activity is increased.
  • the IDO of the mesenchymal stem cells of the present invention is highly expressed and/or the activity thereof is higher than that of the mesenchymal stem cells obtained by a conventional culture method (for example, culture by a flat culture method). It is preferably 2 times or more, more preferably 5 times or more, further preferably 10 times or more, particularly preferably 100 times or more compared to mesenchymal stem cells obtained under conventional culture conditions, and high expression and/or activity of IDO is enhanced. doing.
  • IDO is known to be induced by the The Janus kinase/signal transceivers and activators of transcription (JAK/STAT) signaling pathways. It is known that this JAK/STAT signal pathway is triggered by various cytokines and growth factors and is involved in various actions such as cell proliferation, differentiation, migration, apoptosis and cell survival.
  • JAK/STAT The Janus kinase/signal transceivers and activators of transcription
  • the mesenchymal stem cell of the present invention is characterized by high expression of the following factors in addition to kynurenine.
  • the mesenchymal stem cells of the present invention highly express CSF3, CSF2, LIF, IL6, IL6R or STAT4.
  • Colony stimulating factor 3 is a cytokine involved in the production and differentiation of granulocytes.
  • Colony stimulating factor 2 (CSF2) is a cytokine involved in the production and differentiation of granulocytes and macrophages.
  • the leukemia inhibitory factor (LIF) is a cytokine that plays an important role in the induction of cell differentiation in the hematopoietic system and the nervous system, the regulation of mesenchymal to epithelial conversion in the development of the kidney, and the maternal-fetal immune tolerance.
  • Interleukin 6 is a cytokine having various functions under various inflammatory conditions and B cell maturation.
  • interleukin 6 receptor (IL6R) is a receptor for IL6.
  • STAT4 The signal transducer and activator of transcription 4 (STAT4) functions as a transcription factor in response to cytokines and growth factors. It is known to play an important role in the response to IL12 in lymphocytes and the differentiation of helper T cells.
  • ciliary neurotrophic factor receptor growth hormone receptor
  • Cbl proto-oncogene B E3 ubiquitin protein ligase
  • interleukin 15 interleukin 6 signal transducer
  • interleukin 10 cardiotrophin-like cytokine factor 1
  • cyclin D2 interleukin 11 receptor
  • alpha, interleukin 15 receptor alpha
  • Pim-1 proto-oncogene serine / threonine kinase
  • suppressor of cytokine signaling 1 signal transducer and activator of transcription 2 113kDa
  • interleukin 19 interleukin 7, interleukin 24, interleukin 22 receptor
  • alpha 1 suppressor of cytokine signaling 3 Janus kinase 2
  • interferon (alpha, beta and omega) receptor 1 interferon (alpha, beta and omega) receptor 2
  • interferon gamma receptor 2 interferon gamma transducer
  • the mesenchymal stem cells have the ability to differentiate into one or more cells belonging to mesenchymal cells (osteocytes, cardiomyocytes, chondrocytes, tendon cells, adipocytes, etc.) and maintain the ability.
  • mesenchymal stem cells used in the present invention means the same cells as stromal cells, and does not particularly distinguish between the two. Further, it may be simply referred to as a mesenchymal cell.
  • tissue containing mesenchymal stem cells for example, adipose tissue, umbilical cord, bone marrow, cord blood, endometrium, placenta, amniotic membrane, chorion, decidua, dermis, skeletal muscle, periosteum, dental follicle, periodontal ligament, Examples include dental pulp and tooth germ.
  • adipose tissue-derived mesenchymal stem cells mean mesenchymal stem cells contained in adipose tissue, and may be referred to as adipose tissue-derived stromal cells.
  • adipose tissue-derived mesenchymal stem cells adipose tissue-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, placenta-derived mesenchymal cells, from the viewpoint of efficacy for treatment of neuropathic diseases, availability, etc.
  • Stem cells and dental pulp-derived mesenchymal stem cells are preferable
  • adipose tissue-derived mesenchymal stem cells and umbilical cord-derived mesenchymal stem cells are more preferable
  • umbilical cord-derived mesenchymal stem cells are most preferable.
  • the mesenchymal stem cells in the present invention may be derived from the same species as the subject (subject) to be treated or may be derived from different species.
  • Examples of the mesenchymal stem cells of the present invention include humans, horses, cows, sheep, pigs, dogs, cats, rabbits, mice, and rats, preferably cells derived from the same species as the subject (subject) to be treated. ..
  • the mesenchymal stem cells in the present invention may be derived from a subject (subject) to be treated, that is, autologous cells, or may be derived from another subject of the same species, that is, allogeneic cells. Allogeneic cells are preferred.
  • mesenchymal stem cells are unlikely to cause rejection even in an allogeneic subject, cells of a previously prepared donor are expanded and cryopreserved, and the mesenchymal stem cells in the therapeutic agent for diseases of the present invention are used. It can be used as a stem cell. Therefore, compared to the case of preparing and using autologous mesenchymal stem cells, the mesenchymal stem cells of the present invention are easy to commercialize, and from the viewpoint that stable and constant effects are easily obtained, It is more preferable that they are allogeneic.
  • the mesenchymal stem cells mean any cell population containing mesenchymal stem cells.
  • the cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98. % Or 99% are mesenchymal stem cells.
  • the umbilical cord is a white tubular tissue that connects the fetus and the placenta, and is composed of umbilical vein, umbilical artery, glue-like tissue (Wharton's Jelly), umbilical cord matrix itself, and the like, and mesenchymal stem cells. Including a lot.
  • the umbilical cord is preferably obtained from an animal of the same species as the subject (administration subject) using the disease therapeutic agent of the present invention, and more preferably human in consideration of administration of the disease therapeutic agent of the present invention to human.
  • Umbilical cord is a white tubular tissue that connects the fetus and the placenta, and is composed of umbilical vein, umbilical artery, glue-like tissue (Wharton's Jelly), umbilical cord matrix itself, and the like, and mesenchymal stem cells. Including a lot.
  • the umbilical cord is preferably obtained from an animal of the same species as the subject (administration subject) using the disease therapeutic agent of the present
  • the adipose tissue means a tissue containing stromal cells including adipocytes and microvascular cells, and is, for example, a tissue obtained by surgically excising or aspirating subcutaneous fat of a mammal.
  • Adipose tissue can be obtained from subcutaneous fat. It is preferably obtained from an animal of the same species as the subject to which the adipose tissue-derived mesenchymal stem cells described below are administered, and in view of administration to humans, human subcutaneous fat is more preferred.
  • the subcutaneous fat supplying individual may be alive or dead
  • the adipose tissue used in the present invention is preferably a tissue collected from a living individual.
  • liposuction When collected from an individual, examples of liposuction include PAL (power assist) liposuction, Eruconia laser liposuction, body jet liposuction, and the like, and ultrasonic waves are used from the viewpoint of maintaining the state of cells. Is preferably not used.
  • the bone marrow refers to a soft tissue that fills the inner cavity of a bone and is a hematopoietic organ.
  • Bone marrow fluid is present in the bone marrow, and the cells present therein are called bone marrow cells.
  • Bone marrow cells include erythrocytes, granulocytes, megakaryocytes, lymphocytes, adipocytes and the like, as well as mesenchymal stem cells, hematopoietic stem cells, vascular endothelial precursor cells and the like.
  • Bone marrow cells can be obtained, for example, from human iliac bone, long bone bone, or other bone.
  • each tissue-derived mesenchymal stem cell such as adipose tissue-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, bone marrow-derived mesenchymal stem cell is adipose tissue-derived mesenchymal stem cell, umbilical cord-derived mesenchymal system, respectively. It means any cell population containing mesenchymal stem cells derived from each tissue such as stem cells and bone marrow-derived mesenchymal stem cells. The cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98.
  • tissue-derived mesenchymal stem cell such as adipose tissue-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, bone marrow-derived mesenchymal stem cell.
  • the mesenchymal stem cells of the present invention have a high kynurenine production amount or a high kynurenine secretion amount, and, for example, growth characteristics (for example, population doubling ability from passage to aging, doubling time), karyotype analysis (for example, , Normal karyotype, maternal or neonatal lineage, surface marker expression by flow cytometry (eg FACS analysis), immunohistochemistry and/or immunocytochemistry (eg epitope detection), gene expression profiling (eg genes Chip array; reverse transcription PCR, real-time PCR, polymerase chain reaction such as conventional PCR), miRNA expression profiling, protein array, protein secretion such as cytokine (eg, plasma coagulation analysis, ELISA, cytokine array), metabolite (metabolome analysis) ), by other methods known in the art, and the like.
  • growth characteristics for example, population doubling ability from passage to aging, doubling time
  • karyotype analysis for example, Normal kary
  • the method for preparing mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount is not particularly limited, but can be prepared, for example, as follows. That is, from tissues such as fat, umbilical cord and bone marrow, according to a method known to those skilled in the art, mesenchymal stem cells are separated and cultured, cells with high kynurenine production amount or kynurenine secretion amount, cell sorter with IDO expression as an index, It can be obtained by separating with magnetic beads or the like.
  • mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount in mesenchymal stem cells can be obtained by culturing using a specific medium.
  • 80% or more is a cell having a high kynurenine production amount or a high kynurenine secretion amount, and more preferably 90% or more is a cell having a high kynurenine production amount or a high kynurenine secretion amount. Most preferably, it is a uniform cell population of cells having a high kynurenine production amount or a high kynurenine secretion amount.
  • a method for preparing mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount will be specifically described.
  • the mesenchymal stem cells can be prepared by a method well known to those skilled in the art. The method for preparing umbilical cord tissue-derived mesenchymal stem cells and adipose tissue-derived mesenchymal stem cells will be described below as an example.
  • the umbilical cord can be collected by appropriately removing the placenta from the postpartum tissue including the placenta and umbilical cord delivered by vaginal delivery and cesarean section.
  • Aseptic or bacteriostatic treatment may be performed after removing the cord blood from the collected umbilical cord. Removal of cord blood is accomplished by rinsing with an anticoagulant solution such as a heparin-containing solution.
  • Aseptic or bacteriostatic treatment is not particularly limited, but is a medium supplemented with povidone-iodine or a medium supplemented with one or more antibiotics and/or antifungal agents such as penicillin, streptomycin, amphotericin B, gentamicin, and nystatin.
  • red blood cells may be included if necessary.
  • a method for selectively lysing red blood cells a method well known in the art can be used, for example, incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride.
  • the umbilical cord-derived cell of the present invention means a cell population prepared using umbilical cord as a raw material, and may be obtained by a known production method, for example, a method including the following steps (i) to (iii) can do: (I) cutting the umbilical cord; (Ii) a step of culturing the umbilical cord obtained in the step (i); and (iii) a step of subculture.
  • the step of cutting the umbilical cord instead of (i) the step of cutting the umbilical cord, (i′) the step of dissociating the tissue by enzymatic treatment of the umbilical cord may be included. Further, in addition to (i) the step of cutting the umbilical cord, (i') the step of dissociating the tissue by enzymatic treatment of the umbilical cord may be included.
  • the umbilical cord obtained by the above method is cut by mechanical force (shredding force or shearing force) in a state including amniotic membrane, blood vessel, perivascular tissue and Walton jelly. It can be done by Although not particularly limited, examples of the umbilical cord section obtained by cutting have a size of 1 to 10 mm 3 , 1 to 5 mm 3 , 1 to 4 mm 3 , 1 to 3 mm 3 or 1 to 2 mm 3 .
  • the umbilical cord obtained by the above-mentioned method is treated with the enzyme in a state containing amniotic membrane, blood vessel, perivascular tissue and Walton jelly to remove the tissue. It can be performed in the step of dissociating.
  • the enzyme treatment is not particularly limited, but an enzyme treatment using one or more enzymes such as collagenase, dispase and hyaluronidase is exemplified.
  • the umbilical cord obtained in the step (ii) (i) of the present invention is used at an appropriate cell density and culture conditions by using an appropriate cell culture medium. Incubate.
  • the umbilical cord tissue-derived mesenchymal stem cells of the present invention can be produced using a suspension culture production method.
  • a suspension culture production method a method of stirring culture of sphere-shaped cell clusters formed by any method in a culture container, stirring culture in a culture tank by adhering cells on the microcarrier and stirring the microcarrier There are ways to do it.
  • the stirring can be carried out by rotating a stirring blade in a container with a stirrer, placing a bag containing the culture solution and cells on a shaker, and shaking the bag to suspend the culture solution.
  • the medium used in the suspension culture production method is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, and the medium as described above is exemplified.
  • the microcarrier is not particularly limited as long as it can be used in suspension culture, and examples thereof include polyester, polystyrene, glass, dextran, gelatin, collagen and the like. Specific examples of available microcarriers include GE Healthcare's Cytodex1, Cytodex3, cytopore1, Cytopore2, Cultisphere, Cytoline1, Cytoline2, CornBing's positive concentration, low concentration SynthemaxII and high concentration. Examples thereof include microcarriers, collagen-coated microcarriers, soluble microcarriers, and SoloHill microcarriers manufactured by Pall.
  • the material of the microcarrier preferably has properties such as solubility and biodegradability, and more preferably xeno-free and animal-free in addition to these.
  • the adipose tissue-derived mesenchymal stem cells may be obtained by, for example, the production method described in US Pat. No. 6,777,231, and may be produced by a method including the following steps (i) to (iii), for example. it can: (I) a step of obtaining a cell suspension by digesting adipose tissue with an enzyme; (Ii) sedimenting the cells and resuspending the cells in a suitable medium; and (iii) culturing the cells on a solid surface to remove cells that do not show binding to the solid surface.
  • washing can be accomplished by using a physiologically compatible saline solution (eg, phosphate buffered saline (PBS)) and stirring vigorously to allow precipitation.
  • PBS physiologically compatible saline solution
  • impurities also called debris, for example, damaged tissue, blood, red blood cells
  • washing and sedimentation are generally repeated until the debris have been totally removed from the supernatant.
  • Remaining cells exist as clumps of various sizes, and in order to dissociate them while minimizing damage to the cells themselves, the washed cell clumps are weakened by an enzyme that weakens intercellular bonds or destroys them (for example, Treatment with collagenase, dispase or trypsin) is preferred.
  • the amount of such enzymes and the duration of treatment vary depending on the conditions used, but are known in the art.
  • a method of using cells that naturally exude from the cell mass instead of or in combination with such enzyme treatment is also preferable (improved explant method and the like).
  • the cell mass can be decomposed by other treatment methods such as mechanical stirring, ultrasonic energy, and heat energy, but it is preferable to perform only the enzyme treatment in order to minimize damage to the cells.
  • the cell suspension obtained in step (i) contains a slurry or suspension of aggregated cells, and various types of contaminant cells such as erythrocytes, smooth muscle cells, endothelial cells, and fibroblasts. Therefore, the cells in an aggregated state and these contaminating cells may be subsequently separated and removed, but since they can be removed by adhesion and washing in step (iii) described later, the separation and removal are omitted. May be.
  • the separation and removal of contaminating cells can be accomplished by centrifugation, which forces the cells into a supernatant and a precipitate.
  • the resulting precipitate containing contaminating cells is suspended in a physiologically compatible solvent.
  • the suspended cells may contain red blood cells, but the red blood cells are excluded by the selection by adhesion to the surface of the solid body, which will be described later, and thus the lysis step is not always necessary.
  • a method for selectively lysing red blood cells methods well known in the art can be used, for example, incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride. After lysis, the lysate may be separated from the desired cells by, for example, filtration, centrifugation, or density fractionation.
  • step (ii) in order to increase the purity of mesenchymal stem cells in the cells in suspension, they may be washed once or continuously several times, centrifuged, and resuspended in the medium. Alternatively, cells may be separated based on cell surface marker profile or based on cell size and granularity.
  • the medium used for resuspension is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, but such a medium includes serum added to a basal medium, and/or albumin, transferrin, fatty acid, It may be prepared by adding one or more serum substitutes such as insulin, sodium selenite, cholesterol, collagen precursor, trace elements, 2-mercaptoethanol, 3′-thiolglycerol. If necessary, lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular weight compounds, antibiotics, antioxidants, pyruvic acid, buffers, inorganic salts, and other substances are added to these media, if necessary. May be.
  • the “solid surface” means any material capable of binding and adhering the adipose tissue-derived mesenchymal stem cells of the present invention.
  • such material is a plastic material that has been treated to promote the attachment and adhesion of mammalian cells to its surface.
  • the shape of the culture vessel having a solid surface is not particularly limited, but a petri dish, a flask and the like are preferably used. The cells are washed after incubation to remove unbound cells and cell debris.
  • cells that finally remain bound and adhered to a solid surface can be selected as a cell population of adipose tissue-derived mesenchymal stem cells.
  • surface antigens may be analyzed by a conventional method using flow cytometry or the like.
  • the ability to differentiate into each cell lineage may be tested and such differentiation may be performed by conventional methods.
  • the medium used for culturing the cells of the present invention is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, but such a medium includes serum added to a basal medium and/or albumin and transferrin. , Serum fatty acid, insulin, sodium selenite, cholesterol, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiol glycerol, etc. If necessary, lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular weight compounds, antibiotics, antioxidants, pyruvic acid, buffers, inorganic salts, and other substances are added to these media, if necessary. May be.
  • basal medium examples include IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, MEM- ⁇ medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's medium, and Ham's medium.
  • EMEM Eagle's Minimum Essential Medium
  • DMEM Dulbecco's modified Eagle's Medium
  • Ham's medium examples include RPMI 1640 medium, Fischer's medium, MCDB201 medium and mixed medium thereof.
  • serum examples include, but are not limited to, human serum, fetal bovine serum (FBS), bovine serum, calf serum, goat serum, horse serum, pig serum, sheep serum, rabbit serum, rat serum and the like. .. When using serum, 5 v/v% to 15 v/v%, preferably 10 v/v% may be added to the basal medium.
  • fatty acids include, but are not limited to, linoleic acid, oleic acid, linoleic acid, arachidonic acid, myristic acid, palmitic acid, palmitic acid, stearic acid and the like.
  • lipid include, but are not limited to, phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine.
  • Amino acids include, but are not limited to, for example, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine and the like.
  • Examples of the protein include, but are not limited to, ecotin, reduced glutathione, fibronectin, ⁇ 2-microglobulin and the like.
  • Glycosaminoglycans are exemplified as the polysaccharide, and hyaluronic acid, heparan sulfate, and the like are particularly exemplified among the glycosaminoglycans, but are not limited thereto.
  • the growth factor is, for example, platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor beta (TGF- ⁇ ), hepatocyte growth factor (HGF), epidermal growth factor (EGF).
  • PDGF platelet-derived growth factor
  • bFGF basic fibroblast growth factor
  • TGF- ⁇ transforming growth factor beta
  • HGF epidermal growth factor
  • EGF epidermal growth factor
  • Connective tissue growth factor CGF
  • VEGF vascular endothelial cell growth factor
  • a (Zeno-free) medium containing no xeno-derived components such as serum.
  • Such a medium is provided as a medium prepared in advance for mesenchymal stem cells (stromal cells) from PromoCell, Lonza, Biological Industries, Veritas, R&D Systems, Corning, and Rohto, for example. Has been done.
  • the mesenchymal stem cells of the present invention can be prepared as described above, but may be defined as cells having the following characteristics; (1) Adhesiveness to plastic under culture conditions in standard medium, (2) Surface antigens CD44, CD73, and CD90 are positive, CD31 and CD45 are negative, and (3) Differentiation into osteocytes, adipocytes, and chondrocytes is possible under culture conditions.
  • mesenchymal stem cells obtained by the above step cells having a high IDO expression amount, kynurenine production amount, or kynurenine secretion amount are selectively separated by an immunological method using a cell sorter, magnetic beads, or the like, It is possible to obtain mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount.
  • kynurenine production or a specific culture method capable of inducing kynurenine secretion, or by culturing with a specific medium or the like, induces kynurenine production in mesenchymal stem cells, or kynurenine secretion, efficiently produces kynurenine, or It is also possible to obtain mesenchymal stem cells with high kynurenine secretion.
  • a specific method of selective separation by an immunological method using a cell sorter a method of adhering cells to a microcarrier and stirring the microcarrier to perform stirring culture in a culture tank will be described below.
  • the above-prepared mesenchymal stem cells are treated with a trypsin/EDTA solution or the like, and the cell suspension obtained is centrifuged (room temperature, 400 G, 5 minutes) to remove the supernatant.
  • Staining Buffer 1% BSA-PBS
  • a primary antibody (anti-IDO antibody, anti-kynurenine antibody, etc.) is added to the dispensed cell suspension at a concentration of 5 to 20 ⁇ g/mL and suspended, and then reacted for 30 minutes to 1 hour under light-shielding and refrigeration. After washing 3 times with 1 mL of Staining Buffer, add 50 ⁇ L of Staining Buffer to make the volume 50 ⁇ L, and add the secondary antibody at a concentration of 1-10 ⁇ g/mL to suspend, then react for 30 minutes to 1 hour under light-shielding and refrigeration. ..
  • the cell suspension containing the prepared mesenchymal stem cells and a microcarrier are mixed, added to a culture tank, stirred culture is performed using a serum-free medium for mesenchymal stem cells, and suspension culture is performed for a certain period.
  • stirred culture is performed using a serum-free medium for mesenchymal stem cells, and suspension culture is performed for a certain period.
  • the mesenchymal stem cells of the present invention may be cells that have been repeatedly cryopreserved and thawed as long as they have an immune disease therapeutic effect and an inflammatory disease therapeutic effect.
  • cryopreservation can be performed by suspending mesenchymal stem cells in a cryopreservation solution well known to those skilled in the art and cooling.
  • Suspension can be performed by detaching cells with a detaching agent such as trypsin if necessary, transferring to a cryopreservation container, appropriately treating and then adding a cryopreservation liquid.
  • the cryopreservation liquid may contain DMSO (Dimethyl sulfoxide) as a cryoprotective agent, but DMSO has the property of inducing differentiation of mesenchymal stem cells in addition to cytotoxicity, so the DMSO content is Is preferably reduced.
  • DMSO Dimethyl sulfoxide
  • glycerol, propylene glycol or polysaccharides are exemplified.
  • DMSO contains a concentration of 5% to 20%, preferably a concentration of 5% to 10%, more preferably a concentration of 10%.
  • the additives described in WO2007/058308 may be included.
  • cryopreservation liquid for example, cryopreservation provided by BioVerde, Japan Genetics, Reprocell, Xenoac, Cosmo Bio, Kojin Bio, Thermo Fisher Scientific, etc. A liquid may be used.
  • the suspended cells described above When the suspended cells described above are cryopreserved, they may be frozen at a temperature between ⁇ 80° C. and ⁇ 100° C. (for example, ⁇ 80° C.), using any freezer capable of attaining the temperature. obtain.
  • the cooling rate may be appropriately controlled using a program freezer in order to avoid a rapid temperature change.
  • the cooling rate may be appropriately selected depending on the components of the cryopreservation liquid, and may be performed according to the manufacturer's instruction of the cryopreservation liquid.
  • the storage period is not particularly limited as long as the cells that have been cryopreserved under the above conditions are thawed and retain the same properties as those before freezing, for example, 1 week or more, 2 weeks or more, 3 weeks or more, 4 weeks or more, 2 months or more, 3 months or more, 4 months or more, 5 months or more, 6 months or more, 1 year or more, or more.
  • cell damage can be suppressed by storing it at a lower temperature, it may be stored in a gas phase on liquid nitrogen (from about ⁇ 150° C. or lower to ⁇ 180° C. or higher).
  • the gas phase on liquid nitrogen it can be performed using a storage container well known to those skilled in the art.
  • the thawed mesenchymal stem cells may be appropriately cultured until the next cryopreservation.
  • Culturing of mesenchymal stem cells is performed using a medium capable of culturing the above-mentioned mesenchymal stem cells, and is not particularly limited, but at a culture temperature of about 30 to 40° C., preferably about 37° C., CO 2 -containing air is used. It may be done in an atmosphere.
  • the CO 2 concentration is about 2-10%, preferably about 5-10%.
  • the cells are detached with an exfoliating agent such as trypsin.
  • the culture may be continued by seeding it in a separately prepared culture container at an appropriate cell density.
  • typical cell densities are 100 cells/cm 2 to 100,000 cells/cm 2 , 500 cells/cm 2 to 50,000 cells/cm 2 , 1,000 to 10,000 cells. /Cm 2 , 2,000 to 10,000 cells/cm 2, and the like.
  • the cell density is 2,000-10,000 cells/cm 2 . It is preferable to adjust the time until reaching appropriate confluency to 3 to 7 days. During the culturing, the medium may be appropriately replaced as necessary.
  • Thawing of cryopreserved cells can be performed by a method well known to those skilled in the art. For example, a method of carrying out by standing or shaking in a 37° C. constant temperature bath or in a hot water bath is exemplified.
  • the mesenchymal stem cells of the present invention may be cells in any state, for example, cells recovered by peeling cells in culture, or cells frozen in a cryopreservation solution Good. It is preferable to use cells of the same lot obtained by expansion culture that have been divided into small portions and frozen and stored, because the same effects and advantages can be stably obtained, and handling is excellent.
  • the cryopreserved mesenchymal stem cells may be thawed immediately before use and may be directly mixed with a solution such as an infusion solution or a medium while being suspended in the cryopreservation solution. Alternatively, the cryopreservation solution may be removed by a method such as centrifugation and then suspended in an infusion solution or a solution such as a medium.
  • the “infusion” in the present invention refers to a solution used in the treatment of humans, and is not particularly limited, and examples thereof include physiological saline, Japanese Pharmacopoeia physiological saline, 5% glucose solution, and Japanese Pharmacopoeia.
  • Glucose injection Ringer's solution, Japanese Ringer's solution, Lactated Ringer's solution, Acetate Ringer's solution, 1st solution (starting solution), 2nd solution (dehydration replenishment solution), 3rd solution (maintenance solution), 4th solution (postoperative recovery solution) Etc.
  • the therapeutic agent for an immune disease or the anti-inflammatory agent of the present invention contains the mesenchymal stem cells of the present invention, which have a high kynurenine production amount or a high kynurenine secretion amount.
  • the therapeutic agent for immune diseases or the anti-inflammatory agent of the present invention can be used for prevention, suppression or treatment.
  • the mesenchymal stem cells containing the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention the above description of the mesenchymal stem cells can be applied.
  • the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention are pharmaceutically acceptable according to a conventional method according to their use and morphology, in addition to the above mesenchymal stem cells, as long as the effects of the present invention are not impaired.
  • a carrier or an additive may be included. Examples of such carriers and additives include isotonic agents, thickeners, sugars, sugar alcohols, preservatives (preservatives), bactericides or antibacterial agents, pH regulators, stabilizers, chelating agents.
  • Oily base Oily base, gel base, surfactant, suspending agent, binder, excipient, lubricant, disintegrating agent, foaming agent, fluidizing agent, dispersing agent, emulsifying agent, buffering agent, solubilizing agent , Antioxidants, sweeteners, acidulants, coloring agents, flavoring agents, flavoring agents, and cooling agents, but are not limited thereto.
  • Typical components include, for example, the following carriers and additives.
  • an aqueous carrier such as water or hydrous ethanol
  • the tonicity agent inorganic salt
  • the polyhydric alcohol for example, , Glycerin, propylene glycol, polyethylene glycol, etc.
  • the thickener include carboxyvinyl polymer, hydroxyethyl cellulose, hydroxypropylmethyl cellulose, methyl cellulose, alginic acid, polyvinyl alcohol (completely or partially saponified product), polyvinylpyrrolidone, macrogol.
  • Etc. as sugars, for example, cyclodextrin, glucose, etc.; as sugar alcohols, for example, xylitol, sorbitol, mannitol, etc. (these may be d-form, l-form or dl-form); preservatives
  • the bactericide or antibacterial agent for example, dibutylhydroxytoluene, butylhydroxyanisole, alkyldiaminoethylglycine hydrochloride, sodium benzoate, ethanol, benzalkonium chloride, benzethonium chloride, chlorhexidine gluconate, chlorobutanol, sorbic acid, sorbin Potassium acid, trometamol, sodium dehydroacetate, methyl paraoxybenzoate, ethyl paraoxybenzoate, propyl paraoxybenzoate, butyl paraoxybenzoate, oxyquinoline sulfate, phenethyl alcohol, benzyl alcohol,
  • Potassium hydroxide calcium hydroxide, magnesium hydroxide, sodium hydrogen carbonate, sodium carbonate, borax, triethanolamine, monoethanolamine, diisopropanolamine, sulfuric acid, magnesium sulfate, phosphoric acid, polyphosphoric acid, propionic acid, shu Acid, gluconic acid, fumaric acid, lactic acid, tartaric acid, malic acid, succinic acid, gluconolactone, ammonium acetate and the like; as the stabilizer, for example, dibutyl hydroxytoluene, trometamol, sodium formaldehyde sulfoxylate (Rongalit), Tocopherol, sodium pyrosulfite, monoethanolamine, aluminum monostearate, glyceryl monostearate, Sodium bisulfite, sodium sulfite, etc.; as the oily base, for example, vegetable oil such as olive oil, corn oil, soybean oil, sesame oil, cottonseed oil, medium-chain fatty acids,
  • the therapeutic agent for immune diseases or the anti-inflammatory agent of the present invention can be provided in various forms depending on the purpose, for example, various dosage forms such as solid dosage forms, semi-solid dosage forms and liquid dosage forms.
  • various dosage forms such as solid dosage forms, semi-solid dosage forms and liquid dosage forms.
  • solid agents tablets, powders, powders, granules, capsules, etc.
  • semi-solid agents [ointments (hard ointments, ointments, etc.), creams]
  • liquids [lotions, extracts, suspensions]
  • injections including infusions, embedded injections, continuous injections, ready-to-use injections
  • dialysis agents aerosols, soft capsules, drinks, etc.
  • patches It can be used in the form of agents, po
  • the therapeutic agent for immune diseases or anti-inflammatory agent of the present invention can also be used in the form of a solution or emulsion in an oily or aqueous vehicle. Further, the therapeutic agent or anti-inflammatory agent for immune diseases of the present invention can be applied to the affected area by spraying, and the therapeutic agent or anti-inflammatory agent for immune diseases of the present invention is gelated or formed into a sheet at the affected area after spraying. But it can be used.
  • the immune disease therapeutic agent or anti-inflammatory agent of the present invention can be applied to the affected area after the mesenchymal stem cells are formed into a sheet or a three-dimensional structure.
  • the immunological disease therapeutic agent or anti-inflammatory agent of the present invention is physiological saline, Japanese saline solution, 5% glucose solution, Japanese glucose injection solution, Ringer's solution, Japanese Ringer's solution, lactated Ringer's solution, acetate Ringer's solution, bicarbonate Ringer's solution, Solution 1 (starting solution), solution 2 (dehydration replenisher), solution 3 (maintenance solution), solution 4 (postoperative recovery solution), or a cell culture medium such as DMEM, It can be used by suspending or diluting, and is preferably suspended in physiological saline, 5% glucose solution, No. 1 solution (starting solution), more preferably in 5% glucose solution, No. 1 solution (starting solution), or It can be diluted and used.
  • the pH of the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention is within a pharmaceutically, pharmacologically (pharmaceutically) or physiologically acceptable range. It is not particularly limited as long as it is present, but an example is a range of 2.5 to 9.0, preferably 3.0 to 8.5, and more preferably 3.5 to 8.0.
  • the osmotic pressure of the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention is not particularly limited as long as it is within the range that is acceptable for the living body.
  • An example of the osmotic pressure ratio of the composition of the present invention is a range of preferably 0.7 to 5.0, more preferably 0.8 to 3.0, and further preferably 0.9 to 1.4.
  • the osmotic pressure can be adjusted by a method known in the art using an inorganic salt, a polyhydric alcohol, a sugar alcohol, a saccharide or the like.
  • the osmotic pressure ratio is the ratio of the osmotic pressure of the sample to the osmotic pressure of 286 mOsm (0.9 w/v% sodium chloride aqueous solution) based on the 15th revised Japanese Pharmacopoeia, and the osmotic pressure is the osmotic pressure measurement method described in the Japanese Pharmacopoeia ( Freezing point depression method)
  • the standard solution for measuring the osmotic pressure ratio (0.9 w/v% sodium chloride aqueous solution) was dried in sodium chloride (Japanese Pharmacopoeia standard reagent) at 500 to 650° C. for 40 to 50 minutes, and then in a desiccator (silica gel). Allow to cool, measure exactly 0.900 g of it, dissolve in purified water to prepare exactly 100 mL, or use a commercially available standard solution for osmotic pressure ratio measurement (0.9 w/v% sodium chloride aqueous solution).
  • the route of administration of the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention to the subject is oral administration, subcutaneous administration, intramuscular administration, intravenous administration, intraarterial administration, umbilical cord intravenous administration, intracerebroventricular administration, intrathecal administration. , Intraperitoneal administration, sublingual administration, transrectal administration, transvaginal administration, intraocular administration, nasal administration, inhalation, transdermal administration, implant, direct administration by spraying on the surface of an organ and sticking a sheet, etc.
  • the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention is preferably intraarterial administration, intravenous administration and intraventricular administration, and more preferably intravenous from the viewpoint of reducing the burden on the subject.
  • Inner administration, intramuscular administration, intranasal administration are preferred.
  • the dose depends on the patient's condition (weight, age, symptoms, physical condition, etc.), and the dosage form of the therapeutic agent for anti-inflammatory diseases and anti-inflammatory agent of the present invention. Although it may vary depending on the like, from the viewpoint of exhibiting sufficient therapeutic effect of the therapeutic agent for immune diseases and anti-inflammatory agent, the larger amount tends to be preferable, while the amount from the viewpoint of suppressing the occurrence of side effects is A smaller amount tends to be preferable.
  • the number of cells is 1 ⁇ 10 3 to 1 ⁇ 10 12 cells/time, preferably 1 ⁇ 10 4 to 1 ⁇ 10 11 cells/time, more preferably 1 ⁇ 10 5 to It is 1 ⁇ 10 10 pieces/time, more preferably 5 ⁇ 10 6 to 1 ⁇ 10 9 pieces/time.
  • the dose per body weight of the patient is 1 ⁇ 10 to 5 ⁇ 10 10 cells/kg, preferably 1 ⁇ 10 2 to 5 ⁇ 10 9 cells/kg, and more preferably 1 ⁇ 10 3 to 5 ⁇ 10 5. 8 pieces/kg, more preferably 1 ⁇ 10 4 to 5 ⁇ 10 7 pieces/kg.
  • the number of cells When administered to a newborn, the number of cells is 1 ⁇ 10 3 to 1 ⁇ 10 11 cells/time, preferably 1 ⁇ 10 4 to 1 ⁇ 10 10 cells/time, more preferably 1 ⁇ 10 5 to 1 ⁇ .
  • the number is 10 9 pieces/time, more preferably 5 ⁇ 10 5 to 5 ⁇ 10 8 pieces/time.
  • the dose per body weight of the patient is 1 ⁇ 10 to 5 ⁇ 10 10 cells/kg, preferably 1 ⁇ 10 2 to 5 ⁇ 10 9 cells/kg, and more preferably 1 ⁇ 10 3 to 5 ⁇ 10 5. 8 pieces/kg, more preferably 1 ⁇ 10 4 to 5 ⁇ 10 7 pieces/kg. It should be noted that this dose may be a single dose and may be administered in multiple doses, or the dose may be administered in multiple doses.
  • the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention may be administered together with one or more other drugs.
  • the other drug includes any drug that can be used as a therapeutic agent for immune disorders or an anti-inflammatory agent, and examples thereof include abciximab, adalimumab, alemtuzumab, basiliximab, bavacizumab, cetuximab, daclizumab, efalizumab, gemtuzumab, infliximab, infliximab, infliximab, and infliximab.
  • Monoclonal antibodies such as mepolizumab, OKT3, omaluzumab, palivizumab, pexelizumab, rituximab, tositumomab, trastuzumab, alefacept, fusion protein such as denileukin diftitox, etanercept, soluble cytokine receptors such as anakinra, IFN-IF, IFN- ⁇ , IFN-IF.
  • cytokines azatadine maleate, brompheniramine maleate, chlorpheniramine maleate, clemastine fumarate, cyproheptadine hydrochloride, d chlorphene maleate Lamin, diphenhydramine hydrochloride, diphenylpyraline hydrochloride, hydroxyzine hydrochloride, metzirazine hydrochloride, promethazine hydrochloride, trimeprazine tartrate, loeiperenamine citrate, triperenamine hydrochloride, triprrolidine hydrochloride, acrivastine, cetirizine, desloratadine, ebastine, fexofenadine , H1 blockers such as levocetirizine, loratadine and mizolastine, H2 blockers such as cimetidine, beclomethasone dipropionate, bude
  • ⁇ -agonists such as albuterol
  • cytotoxic drugs such as daunomycin, etoposide, 6-mercaptopurine
  • antihistamines such as daunomycin, etoposide, 6-mercaptopurine
  • NSAIDs such as amino-2-amino-2-amino-2-amino-2-amino-2-amino-2-amino-2-amino-2-aminoethyline, aminotyline, and aspirin.
  • cryotherapy can be combined with administration of the therapeutic agent for immune diseases or anti-inflammatory agent of the present invention.
  • the mesenchymal stem cells of the present invention can be used for various autoimmune diseases and inflammatory diseases.
  • Specific diseases include GVHD, cartilage degradation, rheumatoid arthritis, systemic lupus erythematosus, psoriatic arthritis, spondyloarthritis, and deformity.
  • Gastroesophageal reflux disease Barrett's esophagus, aplastic anemia, graft-versus-host disease, sickle cell disease, CVID, high IgM syndrome, IgA deficiency, transient hypogammaglobulinemia, X-linked agammaglobulinemia Disease, chronic cutaneous mucosal candidiasis, Di George syndrome, X-linked lymphoproliferative syndrome, ataxia-telangiectasia, cartilage dysplasia, combined immunodeficiency, high IgE syndrome, MHC deficiency, severe combined immunity Deficiency, Viscott-Aldrich syndrome, Chediak-East syndrome, chronic granulomatous disease, leukocyte adhesion deficiency, IFN- ⁇ receptor deficiency, interleukin (IL)-12 deficiency, IL-12 receptor ⁇ 1 deficiency, Examples include ZAP-70 deficiency and angioedema.
  • the present invention is characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, a method for treating an immune disease, kynurenine synthesized from tryptophan by IDO induced intracellularly by IFN ⁇ treatment.
  • a method for treating an immune disease characterized by using mesenchymal stem cells having high production or kynurenine secretion, characterized by using mesenchymal stem cells in which JAK/STAT pathway is activated in a steady state
  • methods of treating immune disorders are also included.
  • the present invention is characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, a method for treating an inflammatory disease, and IDO induced intracellularly by IFN ⁇ treatment to synthesize from tryptophan.
  • Method for treating inflammatory diseases characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, and using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state
  • a method for treating an inflammatory disease which is characterized in that Furthermore, the present invention is characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, a method for treating GVHD, and IDO induced intracellularly by IFN ⁇ treatment to synthesize from tryptophan.
  • a method for treating GVHD characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, characterized by using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state.
  • the method of treating GVHD is also included.
  • the description regarding the mesenchymal stem cells, the therapeutic agent for immune diseases, the anti-inflammatory agent, and the therapeutic agent for GVHD can be applied.
  • Umbilical cord-derived mesenchymal stem cells (UCMSC-EXP, UCMSC-CP)] Umbilical cord-derived cells were collected by the method described in Cytotherapy, 18, 229-241, 2016. Briefly, with the approval of the Ethics Committee of the Institute of Medical Science, the University of Tokyo, the umbilical cord collected with the consent of the donor was shredded into 1 to 2 mm 3 pieces and seeded on a culture dish. Then, cell amigo (Tsubakimoto Chain Co., Ltd.) is covered, and the cells are cultured in ⁇ -minimal essential medium (MEM- ⁇ ) supplemented with 10% fetal bovine serum (FBS) and antibiotics by the “improved explant method”. Umbilical cord-derived mesenchymal stem cells (hereinafter referred to as "UCMSC-EXP”) were obtained.
  • MEM- ⁇ ⁇ -minimal essential medium
  • FBS fetal bovine serum
  • umbilical cord-derived cells were collected by the method described in Cytotherapy, 18, 229-241, 2016, and after obtaining the approval of the Ethics Committee of the Institute of Medical Science, University of Tokyo, with the consent of the donor.
  • the collected umbilical cord was cut into a size of 1 to 2 mm 3 , treated with collagenase, and then centrifuged (400 ⁇ g for 5 minutes) to obtain cells, which were then used as serum-free medium for mesenchymal stem cells (Rohto Umbilical cord-derived mesenchymal stem cells (hereinafter referred to as "UCMSC-CP”) were obtained by "collagenase method" in which the cells were adherently cultured in a culture flask.
  • the cells obtained from the umbilical cord tissue by the above-mentioned improved explant method or collagenase method were used as the first passage cells (P1).
  • Cell (P2) the cells obtained from the umbilical cord tissue by the above-mentioned improved explant method or collagenase method were used as the first passage cells (P1).
  • Cell (P2) the cells obtained from the umbilical cord tissue by the above-mentioned improved explant method or collagenase method were used as the first passage cells (P1).
  • Cell (P2) Cell (P2), third passage cell (P3), etc.
  • the obtained UCMSC-EXP and UCMSC-CP were detached using a cell detachment solution (TrypLE Select (1X)), transferred to a centrifuge tube, and centrifuged at 400 ⁇ g for 5 minutes to obtain a cell precipitate. .. After removing the supernatant, an appropriate amount of cell cryopreservation solution (STEM-CELLBANKER (Zenoac)) was added and suspended. The cell suspension solution was dispensed into a cryotube and stored at -80°C in a freezer. Then, it moved to the vapor phase on liquid nitrogen, and continued preservation.
  • a cell detachment solution TrypLE Select (1X)
  • UMCSC-EXP which is the umbilical cord tissue-derived frozen cells obtained by the improved explant method described above, were put to sleep, and the cell suspension containing the cells and the microcarriers were mixed, added to the culture tank, and the mesenchyme was added.
  • Stirring culture was performed using a serum-free medium for line stem cells (Rohto) to obtain suspension-cultured cells (hereinafter referred to as “EXP-SUS”) for 8 days.
  • EXP-ADH flat culture cells
  • EXP-SUS suspension culture cells
  • the intracellular kynurenine accumulation amount was higher in EXP-SUS than in EXP-ADH (FIG. 1).
  • the amount of kynurenine in the culture supernatant was quantified by an ELISA method (Immundiagnostic GmbH, product number K7728). This quantification method also resulted in a higher intracellular kynurenine accumulation amount after 8 days of culture in EXP-SUS than in EXP-ADH (FIG. 2). This revealed that intracellular kynurenine production was enhanced by suspension culture in the steady state.
  • the aforementioned UCMSC-CP was put to sleep, the cells were seeded in a cell culture flask, and the cells were subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto), and cultured for 4 days to obtain flat culture cells ( Hereinafter, referred to as "CP-ADH").
  • IFN ⁇ product number: AF-300-02, manufactured by PeproTeck, Inc.
  • IFN ⁇ concentration of 100 ng/mL group and 200 ng/mL group was determined using the Ehrlich reagent.
  • IFN ⁇ 100 ng/mL
  • an IDO inhibitor (30 nM, Epacadostat, CAS No: 1204669-58-8) was added.
  • a group inhibitor group
  • kynurenine in the culture supernatant was also quantified for a control group to which IFN ⁇ was not added.
  • WST-8 Cell Counting Kit-8, Dojindo Laboratories
  • CP-SUS released more kynurenine in response to IFN ⁇ stimulation into the culture supernatant than CP-ADH under all conditions. From this, it was found that CP-SUS was higher than CP-ADH in the induction and activation of IDO by IFN ⁇ stimulation, the accompanying production of kynurenine in the cell, and the higher secretion ability into the culture supernatant (Fig. 3).
  • umbilical cord tissue-derived frozen cells (UCMSC-EXP) were put to sleep, and the cells were seeded in a cell culture flask and subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto). After culturing for one day, the cells were subcultured and further cultivated for 4 days to obtain flat-cultured cells (hereinafter referred to as "EXP-ADH").
  • IFN ⁇ (product number: AF-300-02, manufactured by PeproTeck, Inc.) was added in an amount to give final concentrations of 50 ng/mL and 100 ng/mL, and the amount of kynurenine in the cell supernatant after 64 hours was determined by the Ehrlich reagent. (IFN ⁇ concentration of 50 ng/mL group and 100 ng/mL group). As a control, kynurenine in the culture supernatant was also quantified for a control group to which IFN ⁇ was not added.
  • the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity. Was evaluated.
  • the IDO activity ratio was calculated by setting the IDO/WST8 value of the EXP-ADH control group to 1 (FIG. 4).
  • EXP-SUS has higher IDO activity under any condition than EXP-ADH. From this, it was found that EXP-SUS was higher than EXP-ADH in the induction and activation of IDO by IFN ⁇ stimulation, the accompanying production of kynurenine in the cell, and the higher secretion ability into the culture supernatant.
  • UCMSC-CP was put to sleep, the cells were seeded in a cell culture flask, and the cells were subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto), and cultured for 8 days. , "CP-ADH" was obtained.
  • IFN ⁇ product number: AF-300-02, manufactured by PeproTeck, Inc.
  • IFN ⁇ concentration of 50 ng/mL group and 100 ng/mL group was determined by the Ehrlich reagent.
  • the IDO activity was also measured for a control group to which IFN ⁇ was not added as a comparison target.
  • the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity.
  • WST-8 Cell Counting Kit-8, Dojindo Laboratories
  • the IDO activity ratio was calculated by setting the IDO/WST8 value of the CP-ADH control group to 1 (FIG. 5).
  • CP-SUS has higher IDO activity under all conditions than CP-ADH. From this, it was found that CP-SUS was higher than CP-ADH in the induction and activation of IDO by IFN ⁇ stimulation, the accompanying production of kynurenine in cells, and the secretion ability into the culture supernatant.
  • umbilical cord tissue-derived frozen cells were put to sleep, and the cells were seeded in a cell culture flask and subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto). The cells were cultured for one day to obtain flat culture cells (hereinafter referred to as "CP-ADH").
  • IFN ⁇ product number: AF-300-02, manufactured by PeproTeck, Inc.
  • IDO activity was also measured for a control group to which IFN ⁇ was not added as a comparison target.
  • the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity.
  • WST-8 Cell Counting Kit-8, Dojindo Laboratories
  • the IDO activity ratio was calculated by setting the IDO/WST8 value of the CP-ADH control group to 1 (FIG. 6).
  • CP-SUS has higher IDO activity under all conditions than CP-ADH. From this, it was found that CP-SUS was higher than CP-ADH in the induction and activation of IDO by IFN ⁇ stimulation, the accompanying production of kynurenine in cells, and the secretion ability into the culture supernatant.
  • umbilical cord tissue-derived frozen cells were put to sleep, and the cells were seeded in a cell culture flask and subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto) for 8 days.
  • the cells were cultured to obtain flat culture cells (hereinafter referred to as "EXP-ADH").
  • IFN ⁇ product number: AF-300-02, manufactured by PeproTeck, Inc.
  • the IDO activity was also measured for a control group to which IFN ⁇ was not added as a comparison target.
  • the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity.
  • the IDO activity ratio was calculated by setting the IDO/WST8 value of the EXP-ADH control group to 1 (FIG. 7).
  • EXP-SUS has higher IDO activity under any condition than EXP-ADH. From this, it was found that EXP-SUS was higher than EXP-ADH in the induction and activation of IDO by IFN ⁇ stimulation, the accompanying production of kynurenine in the cell, and the higher secretion ability into the culture supernatant.
  • MRC-5 cells ATCC
  • human newborn-derived skin fibroblasts HDF, ScienceCell Research Laboratories
  • MEM ⁇ medium containing 10% serum Obtained.
  • human umbilical vein endothelial cells (HUVEC, PromoCell) were thawed and subjected to planar culture for 3 days using Endotherial cell growth medium (PromoCell, product number C-22210) to obtain planar cultured cells.
  • UCMSC-SF, UCMSC-MEM, MRC-5, HDF and HUVEC were each cultured at 30,000 cells/cm 2 for 4 days, and the culture supernatant was recovered.
  • the amount of kynurenine in the culture supernatant was measured by a colorimetric method using Ehrlich reagent.
  • the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity. Was evaluated (FIG. 8).
  • UCMSC-SF was found to have higher IDO activity than UCMSC-MEM. Further, it was revealed that UCMSC-SF and UCMSC-MEM have higher IDO activity than MRC-5, HDF and HUVEC.
  • umbilical cord tissue-derived mesenchymal stem cells cultured in a serum-free medium for mesenchymal stem cells produced kynurenine in cells and cultured compared to umbilical cord tissue-derived mesenchymal stem cells cultured in MEM ⁇ containing 10% serum. It was found that the secretory ability into the supernatant was high. Further, it was found that umbilical cord tissue-derived mesenchymal stem cells have higher intracellular kynurenine production and higher secretory ability into the culture supernatant than MRC-5, HDF and HUVEC.
  • the expressed genes were comprehensively analyzed by 3.0.
  • 33 out of 155 genes related to the JAK-STAT signal pathway registered in Kyoto Encyclopedia of Genes and Genomes (KEGG) were compared to EXP-ADH in the steady state, and in EXP-SUS, 1. The expression was higher than 5-fold.
  • FIG. 9 shows the expression level ratios of the top 9 genes.
  • real-time PCR was performed using SYBR (registered trademark) Premix Ex Taq (trademark) II (part number RR081A, manufacturer Takara) with PikoReal96 (manufacturer Thermo), and analyzed with PikoReal Software 2.0 (manufacturer Thermo).
  • the IDO expression level of SUS was calculated when the IDO expression level of ADH in each culture period was set to 1 (Table 1).
  • the primer sequences used are as follows.
  • GAPDH AGC CTC AAG ATC ATC AGC AAT G (F; SEQ ID NO: 3) and ATG GAC TGT GGT CAT GAG TCC TT (R; SEQ ID NO: 4)
  • EXP-SUS was higher than EXP-ADH in the expression of IDO by IFN ⁇ stimulation in cells of any culture days.
  • EXP-SUS suspension culture cells
  • the primer sequences used are as follows.
  • CCL2 AAGAAGCTGTGATCTTCAAGAC (F; SEQ ID NO: 5) and CCATGGAATCCTGAACCCA (R; SEQ ID NO: 6)
  • LPS stimulation increased CCL2 mRNA expression (LPS group), but its expression was suppressed by co-culture with EXP-SUS and EXP-ADH. Furthermore, the result that EXP-SUS had a larger inhibitory ability was obtained (FIG. 12). From this, it was found that EXP-SUS and EXP-ADH are effective against inflammation, and especially EXP-SUS has an excellent anti-inflammatory effect.
  • a therapeutic agent for immune diseases and an anti-inflammatory agent can be provided.

Abstract

The present invention addresses the problem of providing a novel therapeutic agent for an immune disease and a novel therapeutic agent for an anti-inflammatory agent. The present invention which can solve the problem is a mesenchymal stem cell characterized by being capable of producing kynurenine in a large amount or capable of secreting kynurenine in a large amount. The present invention also includes a mesenchymal stem cell which is characterized by being capable of producing kynurenine, which is synthesized from tryptophan with IDO induced in a cell by the treatment with IFNγ, in a large amount or being capable of secreting the kynurenine in a large amount, and a mesenchymal stem cell which is characterized in that the JAK/STAT pathway thereof is enhanced in a steady state.

Description

間葉系幹細胞、免疫疾患治療剤及び抗炎症剤Mesenchymal stem cells, therapeutic agents for immune diseases and anti-inflammatory agents
 本発明は、間葉系幹細胞、免疫疾患治療剤及び抗炎症剤に関する。 The present invention relates to mesenchymal stem cells, therapeutic agents for immune diseases and anti-inflammatory agents.
 免疫不全疾患では、免疫システムが正常に働かないことにより、感染症が起こったり、何度も再発したり、症状が重くなったり、長引いたりする。免疫不全疾患にかかると、細菌、ウィルス、真菌などの外敵による侵襲や癌細胞のような異常細胞の攻撃から体を守る免疫システムの能力が損なわれる。その結果、免疫機能が正常であればかからないような細菌、ウィルス、真菌による感染症や癌に罹患する。  In immunodeficiency diseases, the inability of the immune system to function normally causes infectious diseases, recurrent episodes, severe symptoms, and prolonged symptoms. Immunodeficiency disorders impair the ability of the immune system to protect the body from invasion by external enemies such as bacteria, viruses, and fungi, and attack of abnormal cells such as cancer cells. As a result, they suffer from infectious diseases and cancers caused by bacteria, viruses, and fungi, which would not occur if their immune functions were normal.
 免疫不全疾患には、出生時に既に羅患している先天性(1次性)の免疫不全疾患と、後年何らかの病気の結果などによって発症する後天性(2次性)の免疫不全疾患がある。後天性免疫不全疾患は、長期間の重症疾患の結果発症することが多く、このような重症疾患としては、癌、再生不良貧血、白血病、骨髄線維症、腎不全、糖尿病、肝疾患、脾疾患などが例示される。 Immunodeficiency diseases include congenital (primary) immunodeficiency diseases that are already present at birth, and acquired (secondary) immunodeficiency diseases that are developed due to some disease in later years. .. Acquired immunodeficiency disease often develops as a result of long-term severe disease, such as cancer, aplastic anemia, leukemia, myelofibrosis, renal failure, diabetes, liver disease, splenic disease. Are exemplified.
 また、年をとると共に免疫システムも衰弱し、徐々に、自己と異物を区別できなくなり、その結果、自己免疫疾患が起こりやすくなる。自己免疫疾患は種々あり、様々な細胞や組織が攻撃の対象となる。このような自己免疫疾患の治療としては、免疫システムを抑制して自己免疫反応を制御すること等が行われている。しかし、自己免疫反応の制御に使われる薬物の多くは、体が感染症などと戦う能力も妨げてしまうため自己免疫反応の制御作用のみを有する新規治療薬の開発が望まれている。 Also, as you get older, your immune system becomes weaker, and you gradually lose the ability to distinguish yourself from foreign bodies, and as a result, autoimmune diseases are more likely to occur. There are various autoimmune diseases, and various cells and tissues are targeted for attack. As a treatment for such an autoimmune disease, suppression of the immune system to control an autoimmune reaction has been performed. However, many of the drugs used to control the autoimmune reaction also impede the body's ability to fight infectious diseases and the like, and therefore there is a demand for the development of a new therapeutic drug having only the action of controlling the autoimmune reaction.
 間葉系幹細胞は、1982年にFriedensteinによって初めて骨髄から単離された多分化能を有する前駆細胞である(非特許文献1参照)。間葉系幹細胞は、骨髄、臍帯、脂肪等の様々な組織に存在することが明らかにされており、間葉系幹細胞移植は、様々な難治性疾患に対する新しい治療方法として、期待されている(特許文献1~2参照)。最近では、脂肪組織、胎盤、臍帯、卵膜等の間質細胞に同等の機能を有する細胞が存在することが知られている。そのため、間葉系幹細胞を間質細胞(Mesenchymal Stromal Cell)と称することもある。 Mesenchymal stem cells are pluripotent progenitor cells first isolated from bone marrow by Friedenstein in 1982 (see Non-Patent Document 1). It has been revealed that mesenchymal stem cells are present in various tissues such as bone marrow, umbilical cord, and fat, and mesenchymal stem cell transplantation is expected as a new therapeutic method for various intractable diseases ( See Patent Documents 1 and 2). Recently, it is known that stromal cells such as adipose tissue, placenta, umbilical cord and egg membrane have cells having equivalent functions. Therefore, the mesenchymal stem cells are sometimes referred to as stromal cells (Mesenchymal Stromal Cell).
特開2012-157263号公報JP 2012-157263 A 特表2012-508733号公報Special table 2012-508733 bulletin
 本発明は、上述のような状況の中、新規の免疫疾患治療剤及び抗炎症剤の新規治療剤を提供することを目的とする。 The present invention aims to provide a novel therapeutic agent for an immune disease and a novel therapeutic agent for an anti-inflammatory agent in the above situation.
 上記課題を解決するために鋭意研究した結果、本発明者らは、キヌレニン産生量、もしくはキヌレニン分泌量が高いことを特徴とする、間葉系幹細胞(mesenchymal stem(stromal) cell; MSC)が、免疫疾患及び炎症性疾患の治療に有効であることを見出し、本発明を完成させた。本発明によれば、免疫疾患及び炎症性疾患の治療のために有効な治療剤を提供できる。すなわち本発明の要旨は、以下の通りである。 As a result of intensive research to solve the above-mentioned problems, the present inventors have found that mesenchymal stem cells (mesc) are characterized by high kynurenine production or kynurenine secretion. They have found that they are effective in treating immune diseases and inflammatory diseases, and completed the present invention. According to the present invention, a therapeutic agent effective for treating immune diseases and inflammatory diseases can be provided. That is, the gist of the present invention is as follows.
[1]キヌレニン産生量、もしくはキヌレニン分泌量が高いことを特徴とする、間葉系幹細胞。
[2]IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高いことを特徴とする、間葉系幹細胞。
[3]JAK/STAT経路が、定常状態で活性化していることを特徴とする、間葉系幹細胞。
[4]臍帯組織由来である、[1]から[3]のいずれかに記載の間葉系幹細胞。
[5][1]から[4]のいずれかに記載の間葉系幹細胞を含有する免疫疾患治療剤。
[6][1]から[4]のいずれかに記載の間葉系幹細胞を含有する抗炎症剤。
[7][1]から[4]のいずれかに記載の間葉系幹細胞を含有するGVHD治療剤。
[8]キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、免疫疾患の治療方法。
[9]IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、免疫疾患の治療方法。
[10]JAK/STAT経路が、定常状態で活性化している間葉系幹細胞を用いることを特徴とする、免疫疾患の治療方法。
[11]上記間葉系幹細胞が、臍帯組織由来である、[8]から[10]のいずれかに記載の、免疫疾患の治療方法。
[12]キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、炎症性疾患の治療方法。
[13]IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、炎症性疾患の治療方法。
[14]JAK/STAT経路が、定常状態で活性化している間葉系幹細胞を用いることを特徴とする、炎症性疾患の治療方法。
[15]上記間葉系幹細胞が、臍帯組織由来である、[12]から[14]のいずれかに記載の、炎症性疾患の治療方法。
[16]キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、GVHDの治療方法。
[17]IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、GVHDの治療方法。
[18]JAK/STAT経路が、定常状態で活性化している間葉系幹細胞を用いることを特徴とする、GVHDの治療方法。
[19]上記間葉系幹細胞が、臍帯組織由来である、[16]から[18]のいずれかに記載の、GVHDの治療方法。
[1] A mesenchymal stem cell having a high kynurenine production amount or a high kynurenine secretion amount.
[2] A mesenchymal stem cell characterized by having a high kynurenine production amount or kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFNγ treatment.
[3] A mesenchymal stem cell characterized in that the JAK/STAT pathway is activated in a steady state.
[4] The mesenchymal stem cell according to any one of [1] to [3], which is derived from umbilical cord tissue.
[5] A therapeutic agent for an immune disease containing the mesenchymal stem cell according to any one of [1] to [4].
[6] An anti-inflammatory agent containing mesenchymal stem cells according to any one of [1] to [4].
[7] A therapeutic agent for GVHD containing mesenchymal stem cells according to any one of [1] to [4].
[8] A method for treating an immune disease, which comprises using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount.
[9] A method for treating an immune disease, which comprises using mesenchymal stem cells having a high kynurenine production amount or a kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFNγ treatment.
[10] A method for treating an immune disease, which comprises using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state.
[11] The method for treating an immune disease according to any of [8] to [10], wherein the mesenchymal stem cells are derived from umbilical cord tissue.
[12] A method for treating an inflammatory disease, which comprises using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount.
[13] A method for treating an inflammatory disease, which comprises using mesenchymal stem cells which have a high kynurenine production amount or a kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFNγ treatment.
[14] A method for treating an inflammatory disease, which comprises using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state.
[15] The method for treating an inflammatory disease according to any of [12] to [14], wherein the mesenchymal stem cells are derived from umbilical cord tissue.
[16] A method for treating GVHD, which comprises using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount.
[17] A method for treating GVHD, which comprises using mesenchymal stem cells that have a high kynurenine production amount or a kynurenine secretion amount synthesized from tryptophan by IDO induced intracellularly by IFNγ treatment.
[18] A method for treating GVHD, which comprises using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state.
[19] The method for treating GVHD according to any of [16] to [18], wherein the mesenchymal stem cells are derived from umbilical cord tissue.
 本発明によると、新規な免疫疾患治療剤及び抗炎症剤、並びに新規な免疫疾患の治療方法及び炎症性疾患の治療方法を提供することができる。 According to the present invention, a novel therapeutic agent for immune diseases and anti-inflammatory agent, and a novel method for treating immune diseases and a method for treating inflammatory diseases can be provided.
図1は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の細胞内キヌレニン量の比較を示す図である。FIG. 1 is a diagram showing a comparison of intracellular kynurenine amounts of flat culture cells (ADH) and suspension culture cells (SUS) of mesenchymal stem cells. 図2は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の培養上清中のキヌレニン量の比較を示す図である。FIG. 2 is a diagram showing a comparison of the amount of kynurenine in the culture supernatants of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS). 図3は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の培養上清中のキヌレニン量(IDO活性)の比較を示す図である。FIG. 3 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS). 図4は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の培養上清中のキヌレニン量(IDO活性)の比較を示す図である。FIG. 4 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatants of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS). 図5は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の培養上清中のキヌレニン量(IDO活性)の比較を示す図である。FIG. 5 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatants of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS). 図6は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の培養上清中のキヌレニン量(IDO活性)の比較を示す図である。FIG. 6 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS). 図7は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の培養上清中のキヌレニン量(IDO活性)の比較を示す図である。FIG. 7 is a diagram showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of mesenchymal stem cell flat culture cells (ADH) and suspension culture cells (SUS). 図8は、平面培養細胞培養上清中のキヌレニン量(IDO活性)の比較を示す図である。FIG. 8 is a view showing a comparison of the amount of kynurenine (IDO activity) in the culture supernatant of flat culture cells. 図9は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)のマイクロアレイによる遺伝子発現の解析結果(ADHにおける発現量対するSUSにおける発現量の比)を示す図である。FIG. 9 is a diagram showing the analysis results (ratio of the expression level in SUS to the expression level in ADH) of gene expression by microarray of flat culture cells (ADH) of mesenchymal stem cells and suspension culture cells (SUS). 図10は、GVHDモデルマウスにおけるUC-MSC(EXP-SUS)投与効果を示す図である。FIG. 10 is a diagram showing the effect of UC-MSC (EXP-SUS) administration in GVHD model mice. 図11は、GVHDモデルマウスにおけるUC-MSC(EXP-SUS)投与効果を示す図である。FIG. 11 is a diagram showing the effect of UC-MSC (EXP-SUS) administration in GVHD model mice. 図12は、間葉系幹細胞の平面培養細胞(ADH)と浮遊培養細胞(SUS)の抗炎症効果を示す図である。FIG. 12 is a graph showing the anti-inflammatory effect of mesenchymal stem cell planar cultured cells (ADH) and suspension cultured cells (SUS). 図13は、JAK/STATシグナル経路を示す図である。FIG. 13 is a diagram showing the JAK/STAT signal pathway.
 以下、本発明の間葉系幹細胞、免疫疾患治療剤及び抗炎症剤について詳細に説明する。 The mesenchymal stem cells, therapeutic agents for immune diseases and anti-inflammatory agents of the present invention will be described in detail below.
[間葉系幹細胞]
 本発明の間葉系幹細胞は、キヌレニン産生量、もしくはキヌレニン分泌量が高いことを特徴とする。
[Mesenchymal stem cells]
The mesenchymal stem cells of the present invention are characterized by high kynurenine production or kynurenine secretion.
 キヌレニン(Kynurenine)は、細胞内でトリプトファンよりナイアシンが合成される際に、酵素indoleamine 2,3-dioxygenase(IDO)により産生される代謝物である。T細胞の増殖にはトリプトファンが重要であり、IDO活性によりトリプトファンがキヌレニンに変換され、枯渇することでT細胞の増殖が抑制され、免疫寛容が引き起こされることが知られている。 Kynurenine is a metabolite produced by the enzyme indoleamine 2,3-dioxygenase (IDO) when niacin is synthesized from tryptophan in cells. It is known that tryptophan is important for T cell proliferation, and tryptophan is converted to kynurenine by IDO activity and depleted to suppress T cell proliferation and induce immunological tolerance.
 キヌレニン産生量、もしくはキヌレニン分泌量とは、炎症状態下においてTNFαやIFNγ処理により誘導されるindoleamine 2,3-dioxygenase(IDO)によりトリプトファンから細胞内にて合成された後に細胞外に放出されるキヌレニン量を意味する。 The amount of kynurenine produced or the amount of kynurenine secreted is the kynurenine released from the outside of cells after being synthesized from tryptophan intracellularly by indolamine 2,3-dioxygenase (IDO) induced by TNFα or IFNγ treatment under inflammatory conditions. Means quantity.
 本発明の間葉系幹細胞は、他の細胞に比べ、キヌレニンを高発現していればよいが、具体的には、本発明の間葉系幹細胞は、IFNγ処理により細胞内で誘導されるIDOによりトリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高ければよい。 The mesenchymal stem cells of the present invention need only express kynurenine at a higher level than other cells. Specifically, the mesenchymal stem cells of the present invention are IDO that is induced intracellularly by IFNγ treatment. Therefore, the amount of kynurenine produced from tryptophan or the amount of kynurenine secreted is high.
 本発明の間葉系幹細胞は、従来の培地(例えば、10%血清含有MEMα培地)を用いた培養で得られる間葉系幹細胞に比べて、キヌレニンを高発現していればよく、好ましくは10%血清含有MEMα培地を用いた培養条件下で得られる間葉系幹細胞に比べ1.1倍以上、より好ましくは1.2倍以上、さらに好ましくは1.5倍以上キヌレニン産生量、もしくはキヌレニン分泌量が高ければよい。 The mesenchymal stem cells of the present invention need only highly express kynurenine as compared with mesenchymal stem cells obtained by culturing in a conventional medium (for example, MEMα medium containing 10% serum), and preferably 10 1.1 times or more, more preferably 1.2 times or more, still more preferably 1.5 times or more, kynurenine production amount or kynurenine secretion compared with mesenchymal stem cells obtained under culture conditions using MEM α medium containing 10% serum The higher the amount, the better.
 また、本発明の間葉系幹細胞は、他の細胞に比べて、キヌレニンを高発現していればよく、例えば、MRC―5細胞、ヒト新生児由来皮膚線維芽細胞(HDF)、もしくはヒト臍帯静脈内皮細胞(HUVEC)に比べ、キヌレニンを高発現していればよく、好ましくはMRC―5細胞、HDFもしくはHUVECに比べ5倍以上、より好ましくは10倍以上キヌレニン産生量、もしくはキヌレニン分泌量が高ければよい。 Further, the mesenchymal stem cells of the present invention need only highly express kynurenine as compared with other cells, and examples thereof include MRC-5 cells, human neonatal skin fibroblasts (HDF), or human umbilical vein veins. It is sufficient that kynurenine is highly expressed as compared with endothelial cells (HUVEC), preferably 5 times or more, more preferably 10 times or more kynurenine production amount or kynurenine secretion amount is higher than MRC-5 cells, HDF or HUVEC. Good.
 例えば、従来の培養方法(例えば、平面培養法による培養)で得られる間葉系幹細胞に比べて、キヌレニンを高発現していればよく、好ましくは従来の培養条件下で得られる間葉系幹細胞に比べ2倍以上、より好ましくは5倍以上、さらに好ましくは10倍以上、特に好ましくは100倍以上キヌレニン産生量、もしくはキヌレニン分泌量が高ければよい。なお、平面培養法とは、フラスコやシャーレといった平面に細胞を接着させて行う静置培養により培養が行われておればよく、特に容器、培地等を特定するものではない。 For example, compared to mesenchymal stem cells obtained by a conventional culture method (culture by a flat culture method), it is sufficient that kynurenine is highly expressed, and preferably mesenchymal stem cells obtained under conventional culture conditions. 2 times or more, more preferably 5 times or more, further preferably 10 times or more, particularly preferably 100 times or more, as long as the amount of kynurenine production or the amount of kynurenine secretion is high. In addition, the planar culture method may be performed by static culture in which cells are adhered to a flat surface such as a flask or a petri dish, and does not particularly specify a container, a medium or the like.
 なお、上記キヌレニン産生量、もしくはキヌレニン分泌量が高いこととは、キヌレニン合成に関与するIDOが高発現もしくは活性が高いことを含む。IDOが高発現もしくは活性が高いことについても、上記と同様に規定され、本発明の間葉系幹細胞は、従来の培地(例えば、10%FBS含有MEM-α培地)による培養で得られる間葉系幹細胞に比べて、IDOを高発現している及び/又はIDOが高活性であればよい。好ましくは従来の培養条件下で得られる間葉系幹細胞に比べ1.1倍以上、より好ましくは1.2倍以上、さらに好ましくは1.5倍以上IDOが高発現及び/又は活性が亢進している。 Note that the high amount of kynurenine produced or the amount of kynurenine secreted above includes high expression or high activity of IDO involved in kynurenine synthesis. The high expression or high activity of IDO is also defined in the same manner as above, and the mesenchymal stem cells of the present invention are mesenchymal cells obtained by culturing in a conventional medium (for example, 10% FBS-containing MEM-α medium). It is sufficient that IDO is highly expressed and/or IDO is highly active as compared to the lineage stem cell. It is preferably 1.1 times or more, more preferably 1.2 times or more, further preferably 1.5 times or more, as compared with mesenchymal stem cells obtained under conventional culture conditions, with high expression and/or activity of IDO ing.
 本発明の間葉系幹細胞は、他の細胞、例えば、MRC―5細胞、ヒト新生児由来皮膚線維芽細胞(HDF)、ヒト臍帯静脈内皮細胞(HUVEC)に比べ、IDOが高発現及び/又は活性が亢進している。MRC―5細胞、HDFもしくはHUVECに比べて、IDOが高発現もしくは活性が高く、好ましくはMRC―5細胞、HDFもしくはHUVECに比べ5倍以上、より好ましくは10倍以上IDOが高発現及び/又は活性が亢進している。 The mesenchymal stem cells of the present invention have higher IDO expression and/or activity than other cells such as MRC-5 cells, human neonatal skin fibroblasts (HDF), and human umbilical vein endothelial cells (HUVEC). Is increasing. Higher expression or activity of IDO than MRC-5 cells, HDF or HUVEC, preferably higher than MRC-5 cells, HDF or HUVEC by 5 times or more, more preferably 10 times or more high expression of IDO and/or The activity is increased.
 本発明の間葉系幹細胞は、従来の培養方法(例えば、平面培養法による培養)で得られる間葉系幹細胞に比べて、IDOが高発現及び/又は活性が亢進している。好ましくは従来の培養条件下で得られる間葉系幹細胞に比べ2倍以上、より好ましくは5倍以上、さらに好ましくは10倍以上、特に好ましくは100倍以上IDOが高発現及び/又は活性が亢進している。 The IDO of the mesenchymal stem cells of the present invention is highly expressed and/or the activity thereof is higher than that of the mesenchymal stem cells obtained by a conventional culture method (for example, culture by a flat culture method). It is preferably 2 times or more, more preferably 5 times or more, further preferably 10 times or more, particularly preferably 100 times or more compared to mesenchymal stem cells obtained under conventional culture conditions, and high expression and/or activity of IDO is enhanced. doing.
 IDOは、The Janus kinase/signal transducers and activators of transcription(JAK/STAT)シグナル経路により誘導されることが既知である。このJAK/STATシグナル経路は、種々のサイトカインや成長因子により惹起され、細胞の増殖、分化、遊走、アポトーシス、細胞の生存等様々な作用に関与することが知られている。 IDO is known to be induced by the The Janus kinase/signal transceivers and activators of transcription (JAK/STAT) signaling pathways. It is known that this JAK/STAT signal pathway is triggered by various cytokines and growth factors and is involved in various actions such as cell proliferation, differentiation, migration, apoptosis and cell survival.
 本発明の間葉系幹細胞は、キヌレニン以外にも、以下に挙げる因子が高発現であることを特徴とする。 The mesenchymal stem cell of the present invention is characterized by high expression of the following factors in addition to kynurenine.
 本発明の間葉系幹細胞は、CSF3、CSF2、LIF、IL6、IL6RもしくはSTAT4が高発現である。colony stimulating factor 3(CSF3)は、顆粒球の産生や分化に関与するサイトカインである。colony stimulating factor 2(CSF2)は、顆粒球やマクロファージの産生や分化に関与するサイトカインである。leukemia inhibitory factor(LIF)は、造血系、神経系の細胞分化誘導や、腎臓の発生においてはmesenchymal to epithelial conversionの制御、母体-胎児間の免疫寛容に重要な役割を果たすサイトカインである。interleukin 6(IL6)は、種々の炎症条件下やB細胞の成熟において様々な機能を有するサイトカインである。interleukin 6 receptor(IL6R)は、IL6の受容体である。signal transducer and activator of transcription 4(STAT4)は、サイトカインや成長因子に応答して転写因子として機能する。リンパ球におけるIL12への応答やヘルパーT細胞の分化において重要な役割を担うことが知られている。 The mesenchymal stem cells of the present invention highly express CSF3, CSF2, LIF, IL6, IL6R or STAT4. Colony stimulating factor 3 (CSF3) is a cytokine involved in the production and differentiation of granulocytes. Colony stimulating factor 2 (CSF2) is a cytokine involved in the production and differentiation of granulocytes and macrophages. The leukemia inhibitory factor (LIF) is a cytokine that plays an important role in the induction of cell differentiation in the hematopoietic system and the nervous system, the regulation of mesenchymal to epithelial conversion in the development of the kidney, and the maternal-fetal immune tolerance. Interleukin 6 (IL6) is a cytokine having various functions under various inflammatory conditions and B cell maturation. interleukin 6 receptor (IL6R) is a receptor for IL6. The signal transducer and activator of transcription 4 (STAT4) functions as a transcription factor in response to cytokines and growth factors. It is known to play an important role in the response to IL12 in lymphocytes and the differentiation of helper T cells.
 さらに、本発明の間葉系幹細胞は、以下の因子が高発現である。
 ciliary neurotrophic factor receptor、growth hormone receptor、Cbl proto-oncogene B, E3 ubiquitin protein ligase、interleukin 15、interleukin 6 signal transducer、interleukin 10、cardiotrophin-like cytokine factor 1、cyclin D2、interleukin 11 receptor, alpha、interleukin 15 receptor, alpha、Pim-1 proto-oncogene, serine/threonine kinase、suppressor of cytokine signaling 1、signal transducer and activator of transcription 2, 113kDa、interleukin 19、interleukin 7、interleukin 24、interleukin 22 receptor, alpha 1、suppressor of cytokine signaling 3、Janus kinase 2、interferon (alpha, beta and omega) receptor 1、interferon (alpha, beta and omega) receptor 2、interferon gamma receptor 2 (interferon gamma transducer 1)、prolactin、leukemia inhibitory factor receptor alpha、sprouty homolog 2 (Drosophila)、v-akt murine thymoma viral oncogene homolog 3、interleukin 4 receptor、interleukin 2 receptor, alpha、son of sevenless homolog 1 (Drosophila)、leptin receptor、v-akt murine thymoma viral oncogene homolog 3、interleukin 24、growth hormone 1、interleukin 7 receptor、phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit delta、interleukin 21およびprotein inhibitor of activated STAT 4;これらの因子は、JAK/STAT経路の構成や誘導に重要であることが知られている。本発明の間葉系幹細胞においては、JAK/STAT経路が、定常状態で活性化し、これらの因子が高発現となる。これらの因子は一般的にJAK/STAT経路において図13に示す位置関係を取る。
Furthermore, the following factors are highly expressed in the mesenchymal stem cells of the present invention.
ciliary neurotrophic factor receptor, growth hormone receptor, Cbl proto-oncogene B, E3 ubiquitin protein ligase, interleukin 15, interleukin 6 signal transducer, interleukin 10, cardiotrophin-like cytokine factor 1, cyclin D2, interleukin 11 receptor, alpha, interleukin 15 receptor , alpha, Pim-1 proto-oncogene, serine / threonine kinase, suppressor of cytokine signaling 1, signal transducer and activator of transcription 2, 113kDa, interleukin 19, interleukin 7, interleukin 24, interleukin 22 receptor, alpha 1, suppressor of cytokine signaling 3, Janus kinase 2, interferon (alpha, beta and omega) receptor 1, interferon (alpha, beta and omega) receptor 2, interferon gamma receptor 2 (interferon gamma transducer 1), prolactin, leukemia inhibitory factor receptor alpha, sprouty homolog 2 (Drosophila), v-akt murine thymoma viral oncogene homolog 3, interleukin 4 receptor, interleukin 2 receptor, alpha, son of sevenless homolog 1 (Drosophila), leptin receptor, v-akt murine thymoma viral oncogene homolog 3, interleukin 24, growth hormone 1, interleukin 7 receptor, phosphatide lininositol -4,5-bisphosphate 3-kinase, catalytic subunit AT ate, and interleukin 21 and protein inhito, and interleukin 21 and protein inbite. It is known. In the mesenchymal stem cells of the present invention, the JAK/STAT pathway is activated in the steady state, and these factors are highly expressed. These factors generally have the positional relationship shown in FIG. 13 in the JAK/STAT pathway.
 本発明において間葉系幹細胞とは、間葉系に属する一種以上の細胞(骨細胞、心筋細胞、軟骨細胞、腱細胞、脂肪細胞など)への分化能を有し、当該能力を維持したまま増殖できる細胞を意味する。本発明において用いる間葉系幹細胞なる用語は、間質細胞と同じ細胞を意味し、両者を特に区別するものではない。また、単に間葉系細胞と表記される場合もある。間葉系幹細胞を含む組織としては、例えば、脂肪組織、臍帯、骨髄、臍帯血、子宮内膜、胎盤、羊膜、絨毛膜、脱落膜、真皮、骨格筋、骨膜、歯小嚢、歯根膜、歯髄、歯胚等が挙げられる。例えば脂肪組織由来間葉系幹細胞とは、脂肪組織に含有される間葉系幹細胞を意味し、脂肪組織由来間質細胞と称してもよい。これらのうち、神経障害疾患の治療に対する有効性の観点、入手容易性の観点等から、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞、胎盤由来間葉系幹細胞、歯髄由来間葉系幹細胞が好ましく、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞がより好ましく、臍帯由来間葉系幹細胞が最も好ましい。 In the present invention, the mesenchymal stem cells have the ability to differentiate into one or more cells belonging to mesenchymal cells (osteocytes, cardiomyocytes, chondrocytes, tendon cells, adipocytes, etc.) and maintain the ability. A cell that can grow. The term mesenchymal stem cells used in the present invention means the same cells as stromal cells, and does not particularly distinguish between the two. Further, it may be simply referred to as a mesenchymal cell. The tissue containing mesenchymal stem cells, for example, adipose tissue, umbilical cord, bone marrow, cord blood, endometrium, placenta, amniotic membrane, chorion, decidua, dermis, skeletal muscle, periosteum, dental follicle, periodontal ligament, Examples include dental pulp and tooth germ. For example, adipose tissue-derived mesenchymal stem cells mean mesenchymal stem cells contained in adipose tissue, and may be referred to as adipose tissue-derived stromal cells. Of these, adipose tissue-derived mesenchymal stem cells, umbilical cord-derived mesenchymal stem cells, bone marrow-derived mesenchymal stem cells, placenta-derived mesenchymal cells, from the viewpoint of efficacy for treatment of neuropathic diseases, availability, etc. Stem cells and dental pulp-derived mesenchymal stem cells are preferable, adipose tissue-derived mesenchymal stem cells and umbilical cord-derived mesenchymal stem cells are more preferable, and umbilical cord-derived mesenchymal stem cells are most preferable.
 本発明における間葉系幹細胞は、処置される対象(被検体)と同種由来であってもよいし、異種由来であってもよい。本発明における間葉系幹細胞の種として、ヒト、ウマ、ウシ、ヒツジ、ブタ、イヌ、ネコ、ラビット、マウス、ラットが挙げられ、好ましくは処置される対象(被検体)と同種由来細胞である。本発明における間葉系幹細胞は、処置される対象(被検体)に由来、すなわち自家細胞であってもよいし、同種の別の対象に由来、すなわち他家細胞であってもよい。好ましくは他家細胞である。 The mesenchymal stem cells in the present invention may be derived from the same species as the subject (subject) to be treated or may be derived from different species. Examples of the mesenchymal stem cells of the present invention include humans, horses, cows, sheep, pigs, dogs, cats, rabbits, mice, and rats, preferably cells derived from the same species as the subject (subject) to be treated. .. The mesenchymal stem cells in the present invention may be derived from a subject (subject) to be treated, that is, autologous cells, or may be derived from another subject of the same species, that is, allogeneic cells. Allogeneic cells are preferred.
 間葉系幹細胞は同種異系の被験体に対しても拒絶反応を起こしにくいため、あらかじめ調製されたドナーの細胞を拡大培養して凍結保存したものを、本発明の疾患治療剤における間葉系幹細胞として使用することができる。そのため、自己の間葉系幹細胞を調製して用いる場合と比較して、商品化も容易であり、かつ安定して一定の効果を得られ易いという観点から、本発明における間葉系幹細胞は、同種異系であることがより好ましい。 Since mesenchymal stem cells are unlikely to cause rejection even in an allogeneic subject, cells of a previously prepared donor are expanded and cryopreserved, and the mesenchymal stem cells in the therapeutic agent for diseases of the present invention are used. It can be used as a stem cell. Therefore, compared to the case of preparing and using autologous mesenchymal stem cells, the mesenchymal stem cells of the present invention are easy to commercialize, and from the viewpoint that stable and constant effects are easily obtained, It is more preferable that they are allogeneic.
 本発明において間葉系幹細胞とは、間葉系幹細胞を含む任意の細胞集団を意味する。当該細胞集団は、少なくとも20%以上、好ましくは、30%、40%、50%、60%、70%、75%、80%、85%、90%、93%、96%、97%、98%又は99%が間葉系幹細胞である。 In the present invention, the mesenchymal stem cells mean any cell population containing mesenchymal stem cells. The cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98. % Or 99% are mesenchymal stem cells.
 本発明において臍帯とは、胎児と胎盤を結ぶ白い管状の組織であり、臍帯静脈、臍帯動脈、膠様組織(ウォートンジェリー;Wharton’s Jelly)、臍帯基質自体等から構成され、間葉系幹細胞を多く含む。臍帯は、本発明の疾患治療剤を使用する被験体(投与対象)と同種動物から入手されることが好ましく、本発明の疾患治療剤をヒトへ投与することを考慮すると、より好ましくは、ヒトの臍帯である。 In the present invention, the umbilical cord is a white tubular tissue that connects the fetus and the placenta, and is composed of umbilical vein, umbilical artery, glue-like tissue (Wharton's Jelly), umbilical cord matrix itself, and the like, and mesenchymal stem cells. Including a lot. The umbilical cord is preferably obtained from an animal of the same species as the subject (administration subject) using the disease therapeutic agent of the present invention, and more preferably human in consideration of administration of the disease therapeutic agent of the present invention to human. Umbilical cord.
 本発明において脂肪組織とは、脂肪細胞、及び微小血管細胞等を含む間質細胞を含有する組織を意味し、例えば、哺乳動物の皮下脂肪を外科的切除又は吸引して得られる組織である。脂肪組織は、皮下脂肪より入手され得る。後述する脂肪組織由来間葉系幹細胞の投与対象と同種動物から入手されることが好ましく、ヒトへ投与することを考慮すると、より好ましくは、ヒトの皮下脂肪である。皮下脂肪の供給個体は、生存していても死亡していてもよいが、本発明において用いる脂肪組織は、好ましくは、生存個体から採取された組織である。個体から採取する場合、脂肪吸引は、例えば、PAL(パワーアシスト)脂肪吸引、エルコーニアレーザー脂肪吸引、又は、ボディジェット脂肪吸引などが例示され、細胞の状態を維持するという観点から、超音波を用いないことが好ましい。 In the present invention, the adipose tissue means a tissue containing stromal cells including adipocytes and microvascular cells, and is, for example, a tissue obtained by surgically excising or aspirating subcutaneous fat of a mammal. Adipose tissue can be obtained from subcutaneous fat. It is preferably obtained from an animal of the same species as the subject to which the adipose tissue-derived mesenchymal stem cells described below are administered, and in view of administration to humans, human subcutaneous fat is more preferred. Although the subcutaneous fat supplying individual may be alive or dead, the adipose tissue used in the present invention is preferably a tissue collected from a living individual. When collected from an individual, examples of liposuction include PAL (power assist) liposuction, Eruconia laser liposuction, body jet liposuction, and the like, and ultrasonic waves are used from the viewpoint of maintaining the state of cells. Is preferably not used.
 本発明において骨髄とは、骨の内腔を満たしている柔組織のことをいい、造血器官である。骨髄中には骨髄液が存在し、その中に存在する細胞を骨髄細胞と呼ぶ。骨髄細胞には、赤血球、顆粒球、巨核球、リンパ球、脂肪細胞等の他、間葉系幹細胞、造血幹細胞、血管内皮前駆細胞等が含まれている。骨髄細胞は、例えば、ヒト腸骨、長管骨、又はその他の骨から採取することができる。 In the present invention, the bone marrow refers to a soft tissue that fills the inner cavity of a bone and is a hematopoietic organ. Bone marrow fluid is present in the bone marrow, and the cells present therein are called bone marrow cells. Bone marrow cells include erythrocytes, granulocytes, megakaryocytes, lymphocytes, adipocytes and the like, as well as mesenchymal stem cells, hematopoietic stem cells, vascular endothelial precursor cells and the like. Bone marrow cells can be obtained, for example, from human iliac bone, long bone bone, or other bone.
 本発明において、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞とは、それぞれ脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞を含む任意の細胞集団を意味する。当該細胞集団は、少なくとも20%以上、好ましくは、30%、40%、50%、60%、70%、75%、80%、85%、90%、93%、96%、97%、98%又は99%が、脂肪組織由来間葉系幹細胞、臍帯由来間葉系幹細胞、骨髄由来間葉系幹細胞といった各組織由来間葉系幹細胞である。 In the present invention, each tissue-derived mesenchymal stem cell such as adipose tissue-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, bone marrow-derived mesenchymal stem cell is adipose tissue-derived mesenchymal stem cell, umbilical cord-derived mesenchymal system, respectively. It means any cell population containing mesenchymal stem cells derived from each tissue such as stem cells and bone marrow-derived mesenchymal stem cells. The cell population is at least 20% or more, preferably 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 93%, 96%, 97%, 98. % Or 99% of each tissue-derived mesenchymal stem cell such as adipose tissue-derived mesenchymal stem cell, umbilical cord-derived mesenchymal stem cell, bone marrow-derived mesenchymal stem cell.
 本発明における間葉系幹細胞は、キヌレニン産生量、もしくはキヌレニン分泌量が高いことに加えて、例えば、成長特徴(例えば、継代から老化までの集団倍加能力、倍加時間)、核型分析(例えば、正常な核型、母体系統又は新生児系統)、フローサイトメトリー(例えば、FACS分析)による表面マーカー発現、免疫組織化学及び/又は免疫細胞化学(例えば、エピトープ検出)、遺伝子発現プロファイリング(例えば、遺伝子チップアレイ;逆転写PCR、リアルタイムPCR、従来型PCR等のポリメラーゼ連鎖反応)、miRNA発現プロファイリング、タンパク質アレイ、サイトカイン等のタンパク質分泌(例えば、血漿凝固解析、ELISA、サイトカインアレイ)、代謝産物(メタボローム解析)、本分野で知られている他の方法等によって、特徴付けられてもよい。 The mesenchymal stem cells of the present invention have a high kynurenine production amount or a high kynurenine secretion amount, and, for example, growth characteristics (for example, population doubling ability from passage to aging, doubling time), karyotype analysis (for example, , Normal karyotype, maternal or neonatal lineage, surface marker expression by flow cytometry (eg FACS analysis), immunohistochemistry and/or immunocytochemistry (eg epitope detection), gene expression profiling (eg genes Chip array; reverse transcription PCR, real-time PCR, polymerase chain reaction such as conventional PCR), miRNA expression profiling, protein array, protein secretion such as cytokine (eg, plasma coagulation analysis, ELISA, cytokine array), metabolite (metabolome analysis) ), by other methods known in the art, and the like.
(間葉系幹細胞の調製方法)
 キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞の調製方法は特に限定されないが、例えば以下のようにして調製することができる。すなわち、脂肪、臍帯、骨髄等の組織から、当業者に公知の方法に従って、間葉系幹細胞を分離、培養し、キヌレニン産生量、もしくはキヌレニン分泌量が高い細胞を、IDO発現を指標にセルソーター、磁気ビーズ等で分離することにより取得することができる。また、特定の培地を用いた培養により、間葉系幹細胞におけるキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を取得することもできる。この誘導によって得られる細胞集団において、細胞集団の50%以上がキヌレニン産生量、もしくはキヌレニン分泌量が高い細胞であることが好ましく、70%以上がキヌレニン産生量、もしくはキヌレニン分泌量が高い細胞であることがより好ましく、80%以上がキヌレニン産生量、もしくはキヌレニン分泌量が高い細胞であることがさらに好ましく、90%以上がキヌレニン産生量、もしくはキヌレニン分泌量が高い細胞であることが特に好ましく、実質的にキヌレニン産生量、もしくはキヌレニン分泌量が高い細胞の均一な細胞集団であることが最も好ましい。以下に、キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞の調製方法を具体的に説明する。
(Method for preparing mesenchymal stem cells)
The method for preparing mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount is not particularly limited, but can be prepared, for example, as follows. That is, from tissues such as fat, umbilical cord and bone marrow, according to a method known to those skilled in the art, mesenchymal stem cells are separated and cultured, cells with high kynurenine production amount or kynurenine secretion amount, cell sorter with IDO expression as an index, It can be obtained by separating with magnetic beads or the like. In addition, mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount in mesenchymal stem cells can be obtained by culturing using a specific medium. In the cell population obtained by this induction, it is preferable that 50% or more of the cell population is a cell having a high kynurenine production amount or a high kynurenine secretion amount, and 70% or more is a cell having a high kynurenine production amount or a high kynurenine secretion amount. More preferably, 80% or more is a cell having a high kynurenine production amount or a high kynurenine secretion amount, and more preferably 90% or more is a cell having a high kynurenine production amount or a high kynurenine secretion amount. Most preferably, it is a uniform cell population of cells having a high kynurenine production amount or a high kynurenine secretion amount. Hereinafter, a method for preparing mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount will be specifically described.
 間葉系幹細胞は、当業者に周知の方法により調製することができる。以下に、一つの例として、臍帯組織由来間葉系幹細胞及び脂肪組織由来間葉系幹細胞の調製方法を説明する。 The mesenchymal stem cells can be prepared by a method well known to those skilled in the art. The method for preparing umbilical cord tissue-derived mesenchymal stem cells and adipose tissue-derived mesenchymal stem cells will be described below as an example.
 臍帯は、経膣分娩および帝王切開にて娩出された胎盤および臍帯を含む産褥組織から適宜胎盤を取り除き回収することができる。回収した臍帯から臍帯血を除去した後、無菌または制菌処理を行っても良い。臍帯血の除去は、ヘパリン含有溶液などの抗凝固溶液ですすぐことによって行われる。無菌または制菌処理は、特に限定されるものではないが、ポピドンヨードの塗布、またはペニシリン、ストレプトマイシン、アムホテリシンB、ゲンタマイシン、およびナイスタチンなどの1種類以上の抗生剤および/または抗真菌剤を添加した培地またはバッファー中に浸漬してもよい。また、必要に応じて、赤血球を選択的に溶解する工程を含んでも良い。赤血球を選択的に溶解する方法として、例えば、塩化アンモニウムによる溶解による高張培地または低張培地中でのインキュベーションなど、当技術分野で周知の方法を使用することができる。 The umbilical cord can be collected by appropriately removing the placenta from the postpartum tissue including the placenta and umbilical cord delivered by vaginal delivery and cesarean section. Aseptic or bacteriostatic treatment may be performed after removing the cord blood from the collected umbilical cord. Removal of cord blood is accomplished by rinsing with an anticoagulant solution such as a heparin-containing solution. Aseptic or bacteriostatic treatment is not particularly limited, but is a medium supplemented with povidone-iodine or a medium supplemented with one or more antibiotics and/or antifungal agents such as penicillin, streptomycin, amphotericin B, gentamicin, and nystatin. Alternatively, it may be immersed in a buffer. In addition, a step of selectively lysing red blood cells may be included if necessary. As a method for selectively lysing red blood cells, a method well known in the art can be used, for example, incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride.
 本発明の臍帯由来細胞とは、臍帯を原材料として、調製された細胞集団を意味し、公知の製造方法によって得られれば良く、例えば、以下の工程(i)~(iii)を含む方法で製造することができる:
(i)臍帯を切断する工程;
(ii)(i)の工程で得られた臍帯を培養する工程;ならびに
(iii)継代する工程。
The umbilical cord-derived cell of the present invention means a cell population prepared using umbilical cord as a raw material, and may be obtained by a known production method, for example, a method including the following steps (i) to (iii) can do:
(I) cutting the umbilical cord;
(Ii) a step of culturing the umbilical cord obtained in the step (i); and (iii) a step of subculture.
 また、他の当該細胞の調製方法として、(i)臍帯を切断する工程の代わりに、(i')臍帯を酵素処理より組織を解離させる工程を含んでもよい。さらに、(i)臍帯を切断する工程に加えて、(i')臍帯を酵素処理より組織を解離させる工程を含んでもよい。 Further, as another method for preparing the cells, instead of (i) the step of cutting the umbilical cord, (i′) the step of dissociating the tissue by enzymatic treatment of the umbilical cord may be included. Further, in addition to (i) the step of cutting the umbilical cord, (i') the step of dissociating the tissue by enzymatic treatment of the umbilical cord may be included.
 本発明の(i)臍帯を切断する工程では、上述の方法で入手した臍帯を、羊膜、血管、血管周囲組織およびワルトンジェリーを含む状態にて機械力(細断力または剪断力)によって切断することによって行い得る。特に限定されないが、切断により得られた臍帯切片は、1から10mm、1から5mm、1から4mm、1から3mmまたは1から2mmの大きさが例示される。本発明の(i')臍帯を酵素処理より組織を解離させる工程では、上述の方法で入手した臍帯を、羊膜、血管、血管周囲組織およびワルトンジェリーを含む状態にて酵素処理にて、組織を解離させる工程にて行い得る。特に限定されないが、酵素処理には、コラゲナーゼ、ディスパーゼ及びヒアルロニダーゼなどの1種又は2種以上の酵素を用いた酵素処理が例示される。 In the step (i) of cutting the umbilical cord of the present invention, the umbilical cord obtained by the above method is cut by mechanical force (shredding force or shearing force) in a state including amniotic membrane, blood vessel, perivascular tissue and Walton jelly. It can be done by Although not particularly limited, examples of the umbilical cord section obtained by cutting have a size of 1 to 10 mm 3 , 1 to 5 mm 3 , 1 to 4 mm 3 , 1 to 3 mm 3 or 1 to 2 mm 3 . In the step (i') of dissociating the tissue from the umbilical cord by the enzyme treatment of the present invention, the umbilical cord obtained by the above-mentioned method is treated with the enzyme in a state containing amniotic membrane, blood vessel, perivascular tissue and Walton jelly to remove the tissue. It can be performed in the step of dissociating. The enzyme treatment is not particularly limited, but an enzyme treatment using one or more enzymes such as collagenase, dispase and hyaluronidase is exemplified.
 本発明の(ii)(i)の工程で得られた臍帯を培養する工程は、適切な細胞培地を使用して、(i)の工程で得られた臍帯を適切な細胞密度及び培養条件で培養する。 In the step of culturing the umbilical cord obtained in the step (ii) (i) of the present invention, the umbilical cord obtained in the step (i) is used at an appropriate cell density and culture conditions by using an appropriate cell culture medium. Incubate.
 本発明の臍帯組織由来間葉系幹細胞は、浮遊培養製造法を用いて、製造することができる。浮遊培養製造法として、任意の方法にて形成したスフェア状の細胞塊を培養容器内で撹拌培養する方法、マイクロキャリア上に細胞を接着させてマイクロキャリアを撹拌することにより培養槽内で撹拌培養する方法などがある。なお、撹拌は容器内の撹拌翼をスターラーで回転させる方法、培養液と細胞の入ったバッグを振盪機に乗せてバッグごと揺らすことで培養液を懸濁する方法などがある。また、浮遊培養製造法で用いる培地は、間葉系幹細胞を培養できる培地であれば、特に限定されず、上記のような培地が例示される。マイクロキャリアとしては、浮遊培養で用いることができるものであれば、特に限定されないが、ポリエステル、ポリスチレン、ガラス、デキストラン、ゼラチン、コラーゲン等が例示される。入手可能なマイクロキャリアの具体例としては、GEヘルスケア社のCytodex1、Cytodex3、cytopore1、Cytopore2、Cultisphere、Cytoline 1、Cytoline 2や、Corning社のCellBIND、低濃度Synthemax II、高濃度Synthemax II、ポジティブチャージマイクロキャリア、コラーゲンコートマイクロキャリア、可溶性マイクロキャリア、Pall社のSoloHillマイクロキャリア等が挙げられる。マイクロキャリアの材質については好ましくは可溶性、生分解性といった性質を持ち合わせており、より好ましくはこれらに加えてゼノフリー、アニマルフリーであることが望ましい。 The umbilical cord tissue-derived mesenchymal stem cells of the present invention can be produced using a suspension culture production method. As a suspension culture production method, a method of stirring culture of sphere-shaped cell clusters formed by any method in a culture container, stirring culture in a culture tank by adhering cells on the microcarrier and stirring the microcarrier There are ways to do it. The stirring can be carried out by rotating a stirring blade in a container with a stirrer, placing a bag containing the culture solution and cells on a shaker, and shaking the bag to suspend the culture solution. Further, the medium used in the suspension culture production method is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, and the medium as described above is exemplified. The microcarrier is not particularly limited as long as it can be used in suspension culture, and examples thereof include polyester, polystyrene, glass, dextran, gelatin, collagen and the like. Specific examples of available microcarriers include GE Healthcare's Cytodex1, Cytodex3, cytopore1, Cytopore2, Cultisphere, Cytoline1, Cytoline2, CornBing's positive concentration, low concentration SynthemaxII and high concentration. Examples thereof include microcarriers, collagen-coated microcarriers, soluble microcarriers, and SoloHill microcarriers manufactured by Pall. The material of the microcarrier preferably has properties such as solubility and biodegradability, and more preferably xeno-free and animal-free in addition to these.
 脂肪組織由来間葉系幹細胞は、例えば米国特許第6,777,231号に記載の製造方法によって得られれば良く、例えば、以下の工程(i)~(iii)を含む方法で製造することができる:
(i) 脂肪組織を酵素による消化により細胞懸濁物を得る工程;
(ii) 細胞を沈降させ、細胞を適切な培地に再懸濁する工程;ならびに
(iii) 細胞を固体表面で培養し、固体表面への結合を示さない細胞を除去する工程。
The adipose tissue-derived mesenchymal stem cells may be obtained by, for example, the production method described in US Pat. No. 6,777,231, and may be produced by a method including the following steps (i) to (iii), for example. it can:
(I) a step of obtaining a cell suspension by digesting adipose tissue with an enzyme;
(Ii) sedimenting the cells and resuspending the cells in a suitable medium; and (iii) culturing the cells on a solid surface to remove cells that do not show binding to the solid surface.
 工程(i)において用いる脂肪組織は、洗浄されたものを用いることが好ましい。洗浄は、生理学的に適合する生理食塩水溶液(例えばリン酸緩衝食塩水(PBS))を用いて、激しく攪拌して沈降させることによって行い得る。これは、脂肪組織に含まれる夾雑物(デブリとも言い、例えば損傷組織、血液、赤血球など)を組織から除去するためである。したがって、洗浄及び沈降は一般に、上清からデブリが総体的に除去されるまで繰り返される。残存する細胞は、さまざまなサイズの塊として存在するので、細胞そのものの損傷を最小限に抑えながら解離させるため、洗浄後の細胞塊を、細胞間結合を弱めるか、又は破壊する酵素(例えば、コラゲナーゼ、ディスパーゼ又はトリプシンなど)で処理することが好ましい。このような酵素の量及び処理期間は、使用される条件に依存して変わるが、当技術分野で既知である。このような酵素処理に代えて、又は併用して、細胞塊から自然に滲出してくる細胞を用いる方法も好ましい(改良エクスプラント法等)。また、細胞塊を、機械的な攪拌、超音波エネルギー、熱エネルギーなどの他の処理法で分解することができるが、細胞の損傷を最小限に抑えるため、酵素処理のみで行うことが好ましい。酵素を用いた場合、細胞に対する有害な作用を最小限に抑えるために、適切な期間をおいた後に培地等を用いて酵素を失活させることが望ましい。 It is preferable to use washed adipose tissue for the step (i). Washing can be accomplished by using a physiologically compatible saline solution (eg, phosphate buffered saline (PBS)) and stirring vigorously to allow precipitation. This is because impurities (also called debris, for example, damaged tissue, blood, red blood cells) contained in adipose tissue are removed from the tissue. Therefore, washing and sedimentation are generally repeated until the debris have been totally removed from the supernatant. Remaining cells exist as clumps of various sizes, and in order to dissociate them while minimizing damage to the cells themselves, the washed cell clumps are weakened by an enzyme that weakens intercellular bonds or destroys them (for example, Treatment with collagenase, dispase or trypsin) is preferred. The amount of such enzymes and the duration of treatment vary depending on the conditions used, but are known in the art. A method of using cells that naturally exude from the cell mass instead of or in combination with such enzyme treatment is also preferable (improved explant method and the like). Further, the cell mass can be decomposed by other treatment methods such as mechanical stirring, ultrasonic energy, and heat energy, but it is preferable to perform only the enzyme treatment in order to minimize damage to the cells. When an enzyme is used, it is desirable to deactivate the enzyme using a medium or the like after an appropriate period of time in order to minimize harmful effects on cells.
 工程(i)により得られる細胞懸濁物は、凝集状の細胞のスラリー又は懸濁物、ならびに各種夾雑細胞、例えば赤血球、平滑筋細胞、内皮細胞、及び線維芽細胞を含む。従って、続いて凝集状態の細胞とこれらの夾雑細胞を分離、除去してもよいが、後述する工程(iii)での接着及び洗浄により、除去可能であることから、当該分離、除去は割愛してもよい。夾雑細胞を分離、除去する場合、細胞を上清と沈殿に強制的に分ける遠心分離によって達成しえる。得られた夾雑細胞を含む沈殿は、生理学的に適合する溶媒に懸濁させる。懸濁状の細胞には、赤血球を含む恐れがあるが、後述する個体表面への接着による選択により、赤血球は除外されるため、溶解する工程は必ずしも必要ではない。赤血球を選択的に溶解する方法として、例えば、塩化アンモニウムによる溶解による高張培地又は低張培地中でのインキュベーションなど、当技術分野で周知の方法を使用することができる。溶解後、例えば濾過、遠心沈降、又は密度分画によって溶解物を所望の細胞から分離してもよい。 The cell suspension obtained in step (i) contains a slurry or suspension of aggregated cells, and various types of contaminant cells such as erythrocytes, smooth muscle cells, endothelial cells, and fibroblasts. Therefore, the cells in an aggregated state and these contaminating cells may be subsequently separated and removed, but since they can be removed by adhesion and washing in step (iii) described later, the separation and removal are omitted. May be. The separation and removal of contaminating cells can be accomplished by centrifugation, which forces the cells into a supernatant and a precipitate. The resulting precipitate containing contaminating cells is suspended in a physiologically compatible solvent. The suspended cells may contain red blood cells, but the red blood cells are excluded by the selection by adhesion to the surface of the solid body, which will be described later, and thus the lysis step is not always necessary. As a method for selectively lysing red blood cells, methods well known in the art can be used, for example, incubation in a hypertonic medium or a hypotonic medium by lysis with ammonium chloride. After lysis, the lysate may be separated from the desired cells by, for example, filtration, centrifugation, or density fractionation.
 工程(ii)において、懸濁状の細胞において、間葉系幹細胞の純度を高めるために、1回もしくは連続して複数回洗浄し、遠心分離し、培地に再懸濁してもよい。この他にも、細胞を、細胞表面マーカープロファイルを基に、又は細胞のサイズ及び顆粒性を基に分離してもよい。 In step (ii), in order to increase the purity of mesenchymal stem cells in the cells in suspension, they may be washed once or continuously several times, centrifuged, and resuspended in the medium. Alternatively, cells may be separated based on cell surface marker profile or based on cell size and granularity.
 再懸濁において用いる培地は、間葉系幹細胞を培養できる培地であれば、特に限定されないが、このような培地は、基礎培地に、血清を添加する、及び/又は、アルブミン、トランスフェリン、脂肪酸、インスリン、亜セレン酸ナトリウム、コレステロール、コラーゲン前駆体、微量元素、2-メルカプトエタノール、3’-チオールグリセロール等の1つ以上の血清代替物を添加して作製してもよい。これらの培地には、必要に応じて、さらに脂質、アミノ酸、タンパク質、多糖、ビタミン、増殖因子、低分子化合物、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類等の物質を添加してもよい。 The medium used for resuspension is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, but such a medium includes serum added to a basal medium, and/or albumin, transferrin, fatty acid, It may be prepared by adding one or more serum substitutes such as insulin, sodium selenite, cholesterol, collagen precursor, trace elements, 2-mercaptoethanol, 3′-thiolglycerol. If necessary, lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular weight compounds, antibiotics, antioxidants, pyruvic acid, buffers, inorganic salts, and other substances are added to these media, if necessary. May be.
 続いて、工程(iii)では、工程(ii)で得られた細胞懸濁液中の細胞を分化させずに固体表面上で、上述の適切な細胞培地を使用して、適切な細胞密度及び培養条件で培養する。本発明において、「固体表面」とは、本発明における脂肪組織由来間葉系幹細胞の結合・接着を可能とする任意の材料を意味する。特定の態様では、このような材料は、その表面への哺乳類細胞の結合・接着を促すように処理されたプラスチック材料である。固体表面を有する培養容器の形状は特に限定されないが、シャーレやフラスコなどが好適に用いられる。非結合状態の細胞及び細胞の破片を除去するために、インキュベーション後に細胞を洗浄する。 Subsequently, in step (iii), on the solid surface without differentiating the cells in the cell suspension obtained in step (ii), using the appropriate cell culture medium described above, an appropriate cell density and Culture under culture conditions. In the present invention, the “solid surface” means any material capable of binding and adhering the adipose tissue-derived mesenchymal stem cells of the present invention. In a particular embodiment, such material is a plastic material that has been treated to promote the attachment and adhesion of mammalian cells to its surface. The shape of the culture vessel having a solid surface is not particularly limited, but a petri dish, a flask and the like are preferably used. The cells are washed after incubation to remove unbound cells and cell debris.
 本発明では、最終的に固体表面に結合・接着した状態で留まる細胞を、脂肪組織由来間葉系幹細胞の細胞集団として選択することができる。 In the present invention, cells that finally remain bound and adhered to a solid surface can be selected as a cell population of adipose tissue-derived mesenchymal stem cells.
 選択された細胞について、本発明における間葉系幹細胞であることを確認するために、表面抗原についてフローサイトメトリー等を用いて従来の方法で解析してもよい。さらに、各細胞系列に分化する能力について検査してもよく、このような分化は、従来の方法で行うことができる。 In order to confirm that the selected cells are the mesenchymal stem cells of the present invention, surface antigens may be analyzed by a conventional method using flow cytometry or the like. In addition, the ability to differentiate into each cell lineage may be tested and such differentiation may be performed by conventional methods.
 本発明の細胞の培養で用いる培地は、間葉系幹細胞を培養できる培地であれば、特に限定されないが、このような培地は、基礎培地に、血清を添加する、及び/又は、アルブミン、トランスフェリン、脂肪酸、インスリン、亜セレン酸ナトリウム、コレステロール、コラーゲン前駆体、微量元素、2-メルカプトエタノール、3’-チオールグリセロール等の1つ以上の血清代替物を添加して作製してもよい。これらの培地には、必要に応じて、さらに脂質、アミノ酸、タンパク質、多糖、ビタミン、増殖因子、低分子化合物、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類等の物質を添加してもよい。 The medium used for culturing the cells of the present invention is not particularly limited as long as it is a medium capable of culturing mesenchymal stem cells, but such a medium includes serum added to a basal medium and/or albumin and transferrin. , Serum fatty acid, insulin, sodium selenite, cholesterol, collagen precursors, trace elements, 2-mercaptoethanol, 3'-thiol glycerol, etc. If necessary, lipids, amino acids, proteins, polysaccharides, vitamins, growth factors, low molecular weight compounds, antibiotics, antioxidants, pyruvic acid, buffers, inorganic salts, and other substances are added to these media, if necessary. May be.
 上記基礎培地としては、例えば、IMDM培地、Medium 199培地、Eagle’s Minimum Essential Medium(EMEM)培地、MEM-α培地、Dulbecco’s modified Eagle’s Medium(DMEM)培地、Ham’s F12培地、RPMI 1640培地、Fischer’s培地、MCDB201培地及びこれらの混合培地等が挙げられる。 Examples of the basal medium include IMDM medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, MEM-α medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's medium, and Ham's medium. Examples include RPMI 1640 medium, Fischer's medium, MCDB201 medium and mixed medium thereof.
 上記血清としては、例えば、ヒト血清、ウシ胎児血清(FBS)、ウシ血清、仔ウシ血清、ヤギ血清、ウマ血清、ブタ血清、ヒツジ血清、ウサギ血清、ラット血清等が挙げられるがこれらに限定されない。血清を用いる場合、基礎培地に対して、5v/v%から15v/v%、好ましくは、10v/v%を添加してもよい。 Examples of the above-mentioned serum include, but are not limited to, human serum, fetal bovine serum (FBS), bovine serum, calf serum, goat serum, horse serum, pig serum, sheep serum, rabbit serum, rat serum and the like. .. When using serum, 5 v/v% to 15 v/v%, preferably 10 v/v% may be added to the basal medium.
 上記脂肪酸としては、リノール酸、オレイン酸、リノレイン酸、アラキドン酸、ミリスチン酸、パルミトイル酸、パルミチン酸、及びステアリン酸等が例示されるが、これらに限定されない。脂質は、フォスファチジルセリン、フォスファチジルエタノールアミン、フォスファチジルコリン等が例示されるが、これらに限定されない。アミノ酸は、例えば、L-アラニン、L-アルギニン、L-アスパラギン酸、L-アスパラギン、L-システイン、L-シスチン、L-グルタミン酸、L-グルタミン、L-グリシンなどを含むがこれらに限定されない。タンパク質は、例えば、エコチン、還元型グルタチオン、フィブロネクチン及びβ2-ミクログロブリン等が例示されるが、これらに限定されない。多糖は、グリコサミノグリカンが例示され、グリコサミノグリカンのうち特に、ヒアルロン酸、ヘパラン硫酸等が例示されるが、これらに限定されない。増殖因子は、例えば、血小板由来増殖因子(PDGF)、塩基性線維芽細胞成長因子(bFGF)、トランスフォーミング増殖因子ベータ(TGF-β)、肝細胞増殖因子(HGF)、上皮成長因子(EGF)、結合組織増殖因子(CTGF)、血管内皮細胞増殖因子(VEGF)等が例示されるが、これらに限定されない。本発明において得られる臍帯由来間葉系幹細胞を細胞移植に用いるという観点から、血清等の異種由来成分を含まない(ゼノフリー)培地を用いることが好ましい。このような培地は、例えば、PromoCell社、Lonza社、Biological Industries社、Veritas社、R&D Systems社、Corning社及びRohto社などから間葉系幹細胞(間質細胞)用として予め調製された培地として提供されている。 Examples of the above fatty acids include, but are not limited to, linoleic acid, oleic acid, linoleic acid, arachidonic acid, myristic acid, palmitic acid, palmitic acid, stearic acid and the like. Examples of the lipid include, but are not limited to, phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine. Amino acids include, but are not limited to, for example, L-alanine, L-arginine, L-aspartic acid, L-asparagine, L-cysteine, L-cystine, L-glutamic acid, L-glutamine, L-glycine and the like. Examples of the protein include, but are not limited to, ecotin, reduced glutathione, fibronectin, β2-microglobulin and the like. Glycosaminoglycans are exemplified as the polysaccharide, and hyaluronic acid, heparan sulfate, and the like are particularly exemplified among the glycosaminoglycans, but are not limited thereto. The growth factor is, for example, platelet-derived growth factor (PDGF), basic fibroblast growth factor (bFGF), transforming growth factor beta (TGF-β), hepatocyte growth factor (HGF), epidermal growth factor (EGF). , Connective tissue growth factor (CTGF), vascular endothelial cell growth factor (VEGF) and the like, but are not limited thereto. From the viewpoint of using the umbilical cord-derived mesenchymal stem cells obtained in the present invention for cell transplantation, it is preferable to use a (Zeno-free) medium containing no xeno-derived components such as serum. Such a medium is provided as a medium prepared in advance for mesenchymal stem cells (stromal cells) from PromoCell, Lonza, Biological Industries, Veritas, R&D Systems, Corning, and Rohto, for example. Has been done.
 本発明における間葉系幹細胞は、上述の通り調製することができるが、次の特性を持つ細胞として定義してもよい;
(1)標準培地での培養条件で、プラスチックに接着性を示す、
(2)表面抗原CD44、CD73、CD90が陽性であり、CD31、CD45が陰性であり、及び
(3)培養条件にて骨細胞、脂肪細胞、軟骨細胞に分化可能。
The mesenchymal stem cells of the present invention can be prepared as described above, but may be defined as cells having the following characteristics;
(1) Adhesiveness to plastic under culture conditions in standard medium,
(2) Surface antigens CD44, CD73, and CD90 are positive, CD31 and CD45 are negative, and (3) Differentiation into osteocytes, adipocytes, and chondrocytes is possible under culture conditions.
 上記工程によって得られた間葉系幹細胞から、IDO発現量、キヌレニン産生量、もしくはキヌレニン分泌量が高い細胞を、セルソーター、磁気ビーズ等を用いた免疫学的手法により選択的に分離することで、キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を取得することができる。またキヌレニン産生、もしくはキヌレニン分泌を誘導できる特定の培養方法、或いは特定の培地等による培養を行うことにより、間葉系幹細胞におけるキヌレニン産生、もしくはキヌレニン分泌を誘導し、効率的にキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を取得することもできる。一例として、セルソーターを用いた免疫学的手法による選択的分離の具体的方法、マイクロキャリア上に細胞を接着させてマイクロキャリアを撹拌することにより培養槽内で撹拌培養する方法を以下に説明する。 From the mesenchymal stem cells obtained by the above step, cells having a high IDO expression amount, kynurenine production amount, or kynurenine secretion amount are selectively separated by an immunological method using a cell sorter, magnetic beads, or the like, It is possible to obtain mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount. In addition, kynurenine production, or a specific culture method capable of inducing kynurenine secretion, or by culturing with a specific medium or the like, induces kynurenine production in mesenchymal stem cells, or kynurenine secretion, efficiently produces kynurenine, or It is also possible to obtain mesenchymal stem cells with high kynurenine secretion. As an example, a specific method of selective separation by an immunological method using a cell sorter, a method of adhering cells to a microcarrier and stirring the microcarrier to perform stirring culture in a culture tank will be described below.
 上記調製した間葉系幹細胞をトリプシン・EDTA溶液等により処理して得られた細胞懸濁液を遠心(室温、400G、5分)して上清を除去する。細胞にStaining Buffer(1%BSA-PBS)を加え、1×10cells/500μLとなるように調製し、ピペッティングにより細胞懸濁液濃度を均一にした後、新しい1.5mLマイクロチューブに50μLずつ分注する。分注した細胞懸濁液に1次抗体(抗IDO抗体、抗キヌレニン抗体等)を5~20μg/mLの濃度で添加し懸濁した後に、遮光・冷蔵下で30分間~1時間反応させる。Staining Buffer 1mLで3回洗浄を行った後に、Staining Bufferを加え50μLとし、2次抗体を1~10μg/mLの濃度で添加し懸濁した後に、遮光・冷蔵下で30分間~1時間反応させる。Staining Buffer 1mLで3回洗浄を行った後に、PI Buffer(Staining buffer 14.4mLにPropidium iodide solution(SIGMA社製、P4864)28.8μLを添加して調製)300μLを加えてよく懸濁し、セルストレーナ付チューブに通し、fluorescence activated cell sorting(FACS)で分離を行うことができる。 The above-prepared mesenchymal stem cells are treated with a trypsin/EDTA solution or the like, and the cell suspension obtained is centrifuged (room temperature, 400 G, 5 minutes) to remove the supernatant. Staining Buffer (1% BSA-PBS) was added to the cells to prepare 1×10 6 cells/500 μL, the cell suspension concentration was made uniform by pipetting, and then 50 μL was added to a new 1.5 mL microtube. Dispense each. A primary antibody (anti-IDO antibody, anti-kynurenine antibody, etc.) is added to the dispensed cell suspension at a concentration of 5 to 20 μg/mL and suspended, and then reacted for 30 minutes to 1 hour under light-shielding and refrigeration. After washing 3 times with 1 mL of Staining Buffer, add 50 μL of Staining Buffer to make the volume 50 μL, and add the secondary antibody at a concentration of 1-10 μg/mL to suspend, then react for 30 minutes to 1 hour under light-shielding and refrigeration. .. After washing 3 times with 1 mL of Staining Buffer, 300 μL of PI Buffer (Staining buffer 14.4 mL with 28.8 μL of Propium iodide solution (manufactured by SIGMA, P4864) was added thereto) and well suspended, and the cell strainer was added. Separation can be performed by passing through an attached tube and using fluorescence activated cell sorting (FACS).
 上記調製した間葉系幹細胞を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加して、間葉系幹細胞用無血清培地等を用いて攪拌培養を行い、一定期間浮遊培養を行うことで、従来のフラスコ等での平面培養と比較して、IDO発現量、キヌレニン産生量、もしくはキヌレニン分泌量が高い細胞を得ることができる。 The cell suspension containing the prepared mesenchymal stem cells and a microcarrier are mixed, added to a culture tank, stirred culture is performed using a serum-free medium for mesenchymal stem cells, and suspension culture is performed for a certain period. By carrying out, it is possible to obtain cells having a higher IDO expression amount, kynurenine production amount, or kynurenine secretion amount, as compared with the conventional flat culture in a flask or the like.
(間葉系幹細胞の凍結保存)
 本発明における間葉系幹細胞は、免疫疾患治療効果、炎症性疾患治療効果を備えていれば、適宜、凍結保存及び融解を繰り返した細胞であってもよい。本発明において、凍結保存は、当業者に周知の凍結保存液へ間葉系幹細胞を懸濁し、冷却することによって行い得る。懸濁は、必要に応じて細胞をトリプシンなどの剥離剤によって剥離し、凍結保存容器に移し、適宜、処理した後、凍結保存液を加えることによって行い得る。
(Cryopreservation of mesenchymal stem cells)
The mesenchymal stem cells of the present invention may be cells that have been repeatedly cryopreserved and thawed as long as they have an immune disease therapeutic effect and an inflammatory disease therapeutic effect. In the present invention, cryopreservation can be performed by suspending mesenchymal stem cells in a cryopreservation solution well known to those skilled in the art and cooling. Suspension can be performed by detaching cells with a detaching agent such as trypsin if necessary, transferring to a cryopreservation container, appropriately treating and then adding a cryopreservation liquid.
 凍結保存液は、凍害防御剤として、DMSO(Dimethyl sulfoxide)を含有していてもよいが、DMSOは、細胞毒性に加えて、間葉系幹細胞を分化誘導する特性を有することから、DMSO含有量を減らすことが好ましい。DMSOの代替物として、グリセロール、プロピレングリコール又は多糖類が例示される。DMSOを用いる場合、5%~20%の濃度、好ましくは5%~10%の濃度、より好ましくは10%の濃度を含有する。この他にも、WO2007/058308に記載の添加剤を含んでもよい。このような凍結保存液として、例えば、バイオベルデ社、日本ジェネティクス株式会社、リプロセル社、ゼノアック社、コスモ・バイオ社、コージンバイオ株式会社、サーモフィッシャーサイエンティフィック社などから提供されている凍結保存液を用いてもよい。 The cryopreservation liquid may contain DMSO (Dimethyl sulfoxide) as a cryoprotective agent, but DMSO has the property of inducing differentiation of mesenchymal stem cells in addition to cytotoxicity, so the DMSO content is Is preferably reduced. As an alternative to DMSO, glycerol, propylene glycol or polysaccharides are exemplified. When DMSO is used, it contains a concentration of 5% to 20%, preferably a concentration of 5% to 10%, more preferably a concentration of 10%. In addition to these, the additives described in WO2007/058308 may be included. As such a cryopreservation liquid, for example, cryopreservation provided by BioVerde, Japan Genetics, Reprocell, Xenoac, Cosmo Bio, Kojin Bio, Thermo Fisher Scientific, etc. A liquid may be used.
 上述の懸濁した細胞を凍結保存する場合、-80℃~-100℃の間の温度(例えば、-80℃)で凍結することで良く、当該温度に達成しえる任意のフリーザーを用いて行い得る。特に限定されないが、急激な温度変化を回避するため、プログラムフリーザーを用いて、冷却速度を適宜制御してもよい。冷却速度は、凍結保存液の成分によって適宜選択しても良く、凍結保存液の製造者指示に従って行われ得る。 When the suspended cells described above are cryopreserved, they may be frozen at a temperature between −80° C. and −100° C. (for example, −80° C.), using any freezer capable of attaining the temperature. obtain. Although not particularly limited, the cooling rate may be appropriately controlled using a program freezer in order to avoid a rapid temperature change. The cooling rate may be appropriately selected depending on the components of the cryopreservation liquid, and may be performed according to the manufacturer's instruction of the cryopreservation liquid.
 保存期間は、上記条件で凍結保存した細胞が融解した後、凍結前と同等の性質を保持している限り、特に上限は限定されないが、例えば、1週間以上、2週間以上、3週間以上、4週間以上、2か月以上、3か月以上、4か月以上、5か月以上、6か月以上、1年以上、又はそれ以上が挙げられる。より低い温度で保存することで細胞障害を抑制することができるため、液体窒素上の気相(約-150℃以下から-180℃以上)へ移して保存してもよい。液体窒素上の気相で保存する場合、当業者に周知の保存容器を用いて行うことができる。特に限定されないが、例えば、2週間以上保存する場合、液体窒素上の気相で保存することが好ましい。 The storage period is not particularly limited as long as the cells that have been cryopreserved under the above conditions are thawed and retain the same properties as those before freezing, for example, 1 week or more, 2 weeks or more, 3 weeks or more, 4 weeks or more, 2 months or more, 3 months or more, 4 months or more, 5 months or more, 6 months or more, 1 year or more, or more. Since cell damage can be suppressed by storing it at a lower temperature, it may be stored in a gas phase on liquid nitrogen (from about −150° C. or lower to −180° C. or higher). When storing in the gas phase on liquid nitrogen, it can be performed using a storage container well known to those skilled in the art. Although not particularly limited, for example, when storing for 2 weeks or more, it is preferable to store in a gas phase on liquid nitrogen.
 融解した間葉系幹細胞は、次の凍結保存までに適宜、培養してもよい。間葉系幹細胞の培養は、上述した間葉系幹細胞を培養できる培地を用いて行われ、特に限定されないが、約30~40℃、好ましくは約37℃の培養温度で、CO含有空気の雰囲気下で行われてもよい。CO濃度は、約2~10%、好ましくは約5~10%である。培養において、培養容器に対して適切なコンフルエンシー(例えば、培養容器に対して、50%から80%を細胞が占有することが挙げられる)に達した後に、細胞をトリプシンなどの剥離剤によって剥離し、別途用意した培養容器に適切な細胞密度で播種して培養を継続してもよい。細胞を播種する際において、典型的な細胞密度として、100細胞/cm~100,000細胞/cm、500細胞/cm~50,000細胞/cm、1,000~10,000細胞/cm、2,000~10,000細胞/cmなどが例示される。特定の態様では、細胞密度は2,000~10,000細胞/cmである。適切なコンフルエンシーに達するまでの期間が、3日間から7日間となるように調整することが好ましい。培養中、必要に応じて、適宜、培地を交換してもよい。 The thawed mesenchymal stem cells may be appropriately cultured until the next cryopreservation. Culturing of mesenchymal stem cells is performed using a medium capable of culturing the above-mentioned mesenchymal stem cells, and is not particularly limited, but at a culture temperature of about 30 to 40° C., preferably about 37° C., CO 2 -containing air is used. It may be done in an atmosphere. The CO 2 concentration is about 2-10%, preferably about 5-10%. In culture, after reaching an appropriate confluency with respect to the culture container (for example, 50% to 80% of the cells are occupied in the culture container), the cells are detached with an exfoliating agent such as trypsin. Alternatively, the culture may be continued by seeding it in a separately prepared culture container at an appropriate cell density. When cells are seeded, typical cell densities are 100 cells/cm 2 to 100,000 cells/cm 2 , 500 cells/cm 2 to 50,000 cells/cm 2 , 1,000 to 10,000 cells. /Cm 2 , 2,000 to 10,000 cells/cm 2, and the like. In a particular embodiment, the cell density is 2,000-10,000 cells/cm 2 . It is preferable to adjust the time until reaching appropriate confluency to 3 to 7 days. During the culturing, the medium may be appropriately replaced as necessary.
 凍結保存した細胞の融解は、当業者に周知の方法によって行い得る。例えば、37℃の恒温槽内又は湯浴中にて静置又は振とうすることによって行う方法が例示される。 Thawing of cryopreserved cells can be performed by a method well known to those skilled in the art. For example, a method of carrying out by standing or shaking in a 37° C. constant temperature bath or in a hot water bath is exemplified.
 本発明の間葉系幹細胞は、いずれの状態の細胞であってもよいが、例えば培養中の細胞を剥離して回収された細胞でもよいし、凍結保存液中に凍結された状態の細胞でもよい。拡大培養して得られる同ロットの細胞を小分けして凍結保存したものを使用すると、安定して同様の作用効果が得られる点、取扱い性に優れる点等において好ましい。凍結保存状態の間葉系幹細胞は、使用直前に融解し、凍結保存液に懸濁したまま輸液もしくは培地等の溶液に直接混合してもよい。また、遠心分離等の方法により凍結保存液を除去してから輸液もしくは培地等の溶液に懸濁してもよい。ここで、本発明における「輸液」とは、ヒトの治療の際に用いられる溶液のことをいい、特に限定されないが、例えば、生理食塩水、日局生理食塩液、5%ブドウ糖液、日局ブドウ糖注射液、リンゲル液、日局リンゲル液、乳酸リンゲル液、酢酸リンゲル液、1号液(開始液)、2号液(脱水補給液)、3号液(維持液)、4号液(術後回復液)等が挙げられる。 The mesenchymal stem cells of the present invention may be cells in any state, for example, cells recovered by peeling cells in culture, or cells frozen in a cryopreservation solution Good. It is preferable to use cells of the same lot obtained by expansion culture that have been divided into small portions and frozen and stored, because the same effects and advantages can be stably obtained, and handling is excellent. The cryopreserved mesenchymal stem cells may be thawed immediately before use and may be directly mixed with a solution such as an infusion solution or a medium while being suspended in the cryopreservation solution. Alternatively, the cryopreservation solution may be removed by a method such as centrifugation and then suspended in an infusion solution or a solution such as a medium. Here, the “infusion” in the present invention refers to a solution used in the treatment of humans, and is not particularly limited, and examples thereof include physiological saline, Japanese Pharmacopoeia physiological saline, 5% glucose solution, and Japanese Pharmacopoeia. Glucose injection, Ringer's solution, Japanese Ringer's solution, Lactated Ringer's solution, Acetate Ringer's solution, 1st solution (starting solution), 2nd solution (dehydration replenishment solution), 3rd solution (maintenance solution), 4th solution (postoperative recovery solution) Etc.
[免疫疾患治療剤及び抗炎症剤]
 本発明の免疫疾患治療剤もしくは抗炎症剤は、上述した本発明の、キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を含有する。本発明の免疫疾患治療剤もしくは抗炎症剤によると、免疫疾患治療剤及び抗炎症剤を予防、抑制もしくは治療に利用することができる。本発明の免疫疾患治療剤及び抗炎症剤を含む間葉系幹細胞については、上記間葉系幹細胞の項の説明を適用できる。
[Immune disease therapeutic agent and anti-inflammatory agent]
The therapeutic agent for an immune disease or the anti-inflammatory agent of the present invention contains the mesenchymal stem cells of the present invention, which have a high kynurenine production amount or a high kynurenine secretion amount. According to the therapeutic agent for immune diseases or the anti-inflammatory agent of the present invention, the therapeutic agent for immune diseases and the anti-inflammatory agent can be used for prevention, suppression or treatment. Regarding the mesenchymal stem cells containing the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention, the above description of the mesenchymal stem cells can be applied.
 本発明の免疫疾患治療剤及び抗炎症剤は、本発明の効果を損なわない範囲であれば、上記間葉系幹細胞以外に、その用途や形態に応じて、常法に従い、薬学的に許容される担体や添加物を含有させてもよい。このような担体や添加物としては、例えば、等張化剤、増粘剤、糖類、糖アルコール類、防腐剤(保存剤)、殺菌剤又は抗菌剤、pH調節剤、安定化剤、キレート剤、油性基剤、ゲル基剤、界面活性剤、懸濁化剤、結合剤、賦形剤、滑沢剤、崩壊剤、発泡剤、流動化剤、分散剤、乳化剤、緩衝剤、溶解補助剤、抗酸化剤、甘味剤、酸味剤、着色剤、呈味剤、香料又は清涼化剤等が挙げられるが、これらに限定されない。代表的な成分として例えば次の担体、添加物等が挙げられる。 The therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention are pharmaceutically acceptable according to a conventional method according to their use and morphology, in addition to the above mesenchymal stem cells, as long as the effects of the present invention are not impaired. A carrier or an additive may be included. Examples of such carriers and additives include isotonic agents, thickeners, sugars, sugar alcohols, preservatives (preservatives), bactericides or antibacterial agents, pH regulators, stabilizers, chelating agents. , Oily base, gel base, surfactant, suspending agent, binder, excipient, lubricant, disintegrating agent, foaming agent, fluidizing agent, dispersing agent, emulsifying agent, buffering agent, solubilizing agent , Antioxidants, sweeteners, acidulants, coloring agents, flavoring agents, flavoring agents, and cooling agents, but are not limited thereto. Typical components include, for example, the following carriers and additives.
 担体としては、例えば、水、含水エタノール等の水性担体が;等張化剤(無機塩)としては、例えば、塩化ナトリウム、塩化カリウム、塩化カルシウム、塩化マグネシウム等が;多価アルコールとしては、例えば、グリセリン、プロピレングリコール、ポリエチレングリコール等が;増粘剤としては、例えば、カルボキシビニルポリマー、ヒドロキシエチルセルロース、ヒドロキシプロピルメチルセルロース、メチルセルロース、アルギン酸、ポリビニルアルコール(完全、又は部分ケン化物)、ポリビニルピロリドン、マクロゴール等が;糖類としては、例えば、シクロデキストリン、ブドウ糖等が;糖アルコール類としては、例えば、キシリトール、ソルビトール、マンニトール等(これらはd体、l体又はdl体のいずれでもよい)が;防腐剤、殺菌剤又は抗菌剤としては、例えば、ジブチルヒドロキシトルエン、ブチルヒドロキシアニソール、塩酸アルキルジアミノエチルグリシン、安息香酸ナトリウム、エタノール、塩化ベンザルコニウム、塩化ベンゼトニウム、グルコン酸クロルヘキシジン、クロロブタノール、ソルビン酸、ソルビン酸カリウム、トロメタモール、デヒドロ酢酸ナトリウム、パラオキシ安息香酸メチル、パラオキシ安息香酸エチル、パラオキシ安息香酸プロピル、パラオキシ安息香酸ブチル、硫酸オキシキノリン、フェネチルアルコール、ベンジルアルコール、ビグアニド化合物(具体的には、塩酸ポリヘキサニド(ポリヘキサメチレンビグアニド)等)、グローキル(ローディア社製商品名)等が;pH調節剤としては、例えば、塩酸、ホウ酸、アミノエチルスルホン酸、イプシロン-アミノカプロン酸、クエン酸、酢酸、水酸化ナトリウム、水酸化カリウム、水酸化カルシウム、水酸化マグネシウム、炭酸水素ナトリウム、炭酸ナトリウム、ホウ砂、トリエタノールアミン、モノエタノールアミン、ジイソプロパノールアミン、硫酸、硫酸マグネシウム、リン酸、ポリリン酸、プロピオン酸、シュウ酸、グルコン酸、フマル酸、乳酸、酒石酸、リンゴ酸、コハク酸、グルコノラクトン、酢酸アンモニウム等が;安定化剤としては、例えば、ジブチルヒドロキシトルエン、トロメタモール、ナトリウムホルムアルデヒドスルホキシレート(ロンガリット)、トコフェロール、ピロ亜硫酸ナトリウム、モノエタノールアミン、モノステアリン酸アルミニウム、モノステアリン酸グリセリン、亜硫酸水素ナトリウム、亜硫酸ナトリウム等が;油性基剤としては、例えば、オリーブ油、トウモロコシ油、大豆油、ゴマ油、綿実油等の植物油、中鎖脂肪酸トリグリセリド等が;水性基剤としては、例えば、マクロゴール400等が;ゲル基剤としては、例えば、カルボキシビニルポリマー、ガム質等が;界面活性剤としては、例えば、ポリソルベート80、硬化ヒマシ油、グリセリン脂肪酸エステル、セスキオレイン酸ソルビタン等が;懸濁化剤としては、例えば、サラシミツロウや各種界面活性剤、アラビアゴム、アラビアゴム末、キサンタンガム、大豆レシチン等が;結合剤としては、例えば、ヒドロキシエチルセルロース、ヒドロキシプロピルセルロース、ヒドロキシプロピルメチルセルロース、カルボキシメチルセルロースナトリウム、ポリビニルピロリドン、ポリビニルアルコール等が;賦形剤としては、例えば、ショ糖、乳糖、デンプン、コーンスターチ、結晶セルロース、軽質無水ケイ酸等が;滑沢剤としては、例えば、ショ糖脂肪酸エステル、ステアリン酸マグネシウム、タルク等が;崩壊剤としては、例えば、低置換度ヒドロキシプロピルセルロース、クロスポビドン、クロスカルメロースナトリウム等が;発泡剤としては、例えば、炭酸水素ナトリウム等が;流動化剤としては、例えば、メタケイ酸アルミン酸ナトリウム、軽質無水ケイ酸等が、それぞれ挙げられる。 As the carrier, for example, an aqueous carrier such as water or hydrous ethanol; as the tonicity agent (inorganic salt), for example, sodium chloride, potassium chloride, calcium chloride, magnesium chloride or the like; as the polyhydric alcohol, for example, , Glycerin, propylene glycol, polyethylene glycol, etc.; Examples of the thickener include carboxyvinyl polymer, hydroxyethyl cellulose, hydroxypropylmethyl cellulose, methyl cellulose, alginic acid, polyvinyl alcohol (completely or partially saponified product), polyvinylpyrrolidone, macrogol. Etc.; as sugars, for example, cyclodextrin, glucose, etc.; as sugar alcohols, for example, xylitol, sorbitol, mannitol, etc. (these may be d-form, l-form or dl-form); preservatives As the bactericide or antibacterial agent, for example, dibutylhydroxytoluene, butylhydroxyanisole, alkyldiaminoethylglycine hydrochloride, sodium benzoate, ethanol, benzalkonium chloride, benzethonium chloride, chlorhexidine gluconate, chlorobutanol, sorbic acid, sorbin Potassium acid, trometamol, sodium dehydroacetate, methyl paraoxybenzoate, ethyl paraoxybenzoate, propyl paraoxybenzoate, butyl paraoxybenzoate, oxyquinoline sulfate, phenethyl alcohol, benzyl alcohol, biguanide compound (specifically, polyhexanide hydrochloride ( Polyhexamethylene biguanide), etc.), Glowkill (trade name, manufactured by Rhodia), etc.; examples of the pH adjusting agent include hydrochloric acid, boric acid, aminoethylsulfonic acid, epsilon-aminocaproic acid, citric acid, acetic acid, sodium hydroxide. , Potassium hydroxide, calcium hydroxide, magnesium hydroxide, sodium hydrogen carbonate, sodium carbonate, borax, triethanolamine, monoethanolamine, diisopropanolamine, sulfuric acid, magnesium sulfate, phosphoric acid, polyphosphoric acid, propionic acid, shu Acid, gluconic acid, fumaric acid, lactic acid, tartaric acid, malic acid, succinic acid, gluconolactone, ammonium acetate and the like; as the stabilizer, for example, dibutyl hydroxytoluene, trometamol, sodium formaldehyde sulfoxylate (Rongalit), Tocopherol, sodium pyrosulfite, monoethanolamine, aluminum monostearate, glyceryl monostearate, Sodium bisulfite, sodium sulfite, etc.; as the oily base, for example, vegetable oil such as olive oil, corn oil, soybean oil, sesame oil, cottonseed oil, medium-chain fatty acid triglyceride, etc.; as the aqueous base, for example, Macrogol 400 Etc.; as the gel base, for example, carboxyvinyl polymer, gum, etc.; as the surfactant, for example, polysorbate 80, hydrogenated castor oil, glycerin fatty acid ester, sorbitan sesquioleate, etc.; suspending agent As, for example, beeswax wax and various surfactants, gum arabic, gum arabic powder, xanthan gum, soybean lecithin and the like; as the binder, for example, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, sodium carboxymethyl cellulose, polyvinyl Pyrrolidone, polyvinyl alcohol, etc.; Excipients, for example, sucrose, lactose, starch, corn starch, crystalline cellulose, light anhydrous silicic acid, etc.; Lubricants, for example, sucrose fatty acid ester, magnesium stearate , Talc and the like; disintegrators such as low-substituted hydroxypropylcellulose, crospovidone, croscarmellose sodium and the like; foaming agents such as sodium hydrogencarbonate and the like; fluidizing agents such as Examples thereof include sodium aluminometasilicate and light anhydrous silicic acid.
 本発明の免疫疾患治療剤もしくは抗炎症剤は、目的に応じて種々の形態、例えば、固形剤、半固形剤、液剤等の様々な剤形で提供することができる。例えば、固形剤(錠剤、粉末、散剤、顆粒剤、カプセル剤等)、半固形剤[軟膏剤(硬軟膏剤、軟軟膏剤等)、クリーム剤等]、液剤[ローション剤、エキス剤、懸濁剤、乳剤、シロップ剤、注射剤(輸液剤、埋め込み注射剤、持続性注射、用時調製型の注射剤を含む)、透析用剤、エアゾール剤、軟カプセル剤、ドリンク剤等]、貼付剤、パップ剤等の形態で利用できる。また、本発明の免疫疾患治療剤もしくは抗炎症剤は、油性又は水性のビヒクル中の溶液又は乳液等の形態でも利用できる。さらに、本発明の免疫疾患治療剤もしくは抗炎症剤は噴霧により、患部に適用することもでき、本発明の免疫疾患治療剤もしくは抗炎症剤は噴霧した後に患部でゲル化もしくはシート化される形態でも利用できる。本発明の免疫疾患治療剤もしくは抗炎症剤は上記間葉系幹細胞をシート状または立体構造体とした後に、患部に適用することもできる。 The therapeutic agent for immune diseases or the anti-inflammatory agent of the present invention can be provided in various forms depending on the purpose, for example, various dosage forms such as solid dosage forms, semi-solid dosage forms and liquid dosage forms. For example, solid agents (tablets, powders, powders, granules, capsules, etc.), semi-solid agents [ointments (hard ointments, ointments, etc.), creams], liquids [lotions, extracts, suspensions] Suspensions, emulsions, syrups, injections (including infusions, embedded injections, continuous injections, ready-to-use injections), dialysis agents, aerosols, soft capsules, drinks, etc.], patches It can be used in the form of agents, poultices and the like. The therapeutic agent for immune diseases or anti-inflammatory agent of the present invention can also be used in the form of a solution or emulsion in an oily or aqueous vehicle. Further, the therapeutic agent or anti-inflammatory agent for immune diseases of the present invention can be applied to the affected area by spraying, and the therapeutic agent or anti-inflammatory agent for immune diseases of the present invention is gelated or formed into a sheet at the affected area after spraying. But it can be used. The immune disease therapeutic agent or anti-inflammatory agent of the present invention can be applied to the affected area after the mesenchymal stem cells are formed into a sheet or a three-dimensional structure.
 本発明の免疫疾患治療剤もしくは抗炎症剤は、生理食塩水、日局生理食塩液、5%ブドウ糖液、日局ブドウ糖注射液、リンゲル液、日局リンゲル液、乳酸リンゲル液、酢酸リンゲル液、重炭酸リンゲル液、1号液(開始液)、2号液(脱水補給液)、3号液(維持液)、4号液(術後回復液)等の輸液、又は、DMEM等の細胞培養培地を用いて、懸濁もしくは希釈して用いることができ、好ましくは生理食塩液、5%ブドウ糖液、1号液(開始液)で、より好ましくは5%ブドウ糖液、1号液(開始液)で懸濁もしくは希釈して用いることができる。 The immunological disease therapeutic agent or anti-inflammatory agent of the present invention is physiological saline, Japanese saline solution, 5% glucose solution, Japanese glucose injection solution, Ringer's solution, Japanese Ringer's solution, lactated Ringer's solution, acetate Ringer's solution, bicarbonate Ringer's solution, Solution 1 (starting solution), solution 2 (dehydration replenisher), solution 3 (maintenance solution), solution 4 (postoperative recovery solution), or a cell culture medium such as DMEM, It can be used by suspending or diluting, and is preferably suspended in physiological saline, 5% glucose solution, No. 1 solution (starting solution), more preferably in 5% glucose solution, No. 1 solution (starting solution), or It can be diluted and used.
 本発明の免疫疾患治療剤及び抗炎症剤が液剤である場合、免疫疾患治療剤及び抗炎症剤のpHは、医薬上、薬理学的に(製薬上)又は生理学的に許容される範囲内であれば特に限定されるものではないが、一例として、2.5~9.0、好ましくは3.0~8.5、より好ましくは3.5~8.0となる範囲が挙げられる。 When the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention are liquids, the pH of the therapeutic agent for immune diseases and the anti-inflammatory agent is within a pharmaceutically, pharmacologically (pharmaceutically) or physiologically acceptable range. It is not particularly limited as long as it is present, but an example is a range of 2.5 to 9.0, preferably 3.0 to 8.5, and more preferably 3.5 to 8.0.
 本発明の免疫疾患治療剤及び抗炎症剤が液剤である場合、免疫疾患治療剤及び抗炎症剤の浸透圧については、生体に許容される範囲内であれば、特に制限されない。本発明の組成物の浸透圧比の一例として、好ましくは0.7~5.0、より好ましくは0.8~3.0、さらに好ましくは0.9~1.4となる範囲が挙げられる。浸透圧の調整は無機塩、多価アルコール、糖アルコール、糖類等を用いて、当該技術分野で既知の方法で行うことができる。浸透圧比は、第十五改正日本薬局方に基づき286mOsm(0.9w/v%塩化ナトリウム水溶液)の浸透圧に対する試料の浸透圧の比とし、浸透圧は日本薬局方記載の浸透圧測定法(氷点降下法)を参考にして測定する。なお、浸透圧比測定用標準液(0.9w/v%塩化ナトリウム水溶液)は、塩化ナトリウム(日本薬局方標準試薬)を500~650℃で40~50分間乾燥した後、デシケーター(シリカゲル)中で放冷し、その0.900gを正確に量り、精製水に溶かし正確に100mLとして調製するか、市販の浸透圧比測定用標準液(0.9w/v%塩化ナトリウム水溶液)を用いる。 When the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention are liquids, the osmotic pressure of the therapeutic agent for immune diseases and the anti-inflammatory agent is not particularly limited as long as it is within the range that is acceptable for the living body. An example of the osmotic pressure ratio of the composition of the present invention is a range of preferably 0.7 to 5.0, more preferably 0.8 to 3.0, and further preferably 0.9 to 1.4. The osmotic pressure can be adjusted by a method known in the art using an inorganic salt, a polyhydric alcohol, a sugar alcohol, a saccharide or the like. The osmotic pressure ratio is the ratio of the osmotic pressure of the sample to the osmotic pressure of 286 mOsm (0.9 w/v% sodium chloride aqueous solution) based on the 15th revised Japanese Pharmacopoeia, and the osmotic pressure is the osmotic pressure measurement method described in the Japanese Pharmacopoeia ( Freezing point depression method) The standard solution for measuring the osmotic pressure ratio (0.9 w/v% sodium chloride aqueous solution) was dried in sodium chloride (Japanese Pharmacopoeia standard reagent) at 500 to 650° C. for 40 to 50 minutes, and then in a desiccator (silica gel). Allow to cool, measure exactly 0.900 g of it, dissolve in purified water to prepare exactly 100 mL, or use a commercially available standard solution for osmotic pressure ratio measurement (0.9 w/v% sodium chloride aqueous solution).
 本発明の免疫疾患治療剤及び抗炎症剤の対象への投与経路は、経口投与、皮下投与、筋肉内投与、静脈内投与、動脈内投与、臍帯静脈内投与、脳室内投与、髄腔内投与、腹腔内投与、舌下投与、経直腸投与、経腟投与、眼内投与、経鼻投与、吸入、経皮投与、インプラント、臓器表面への噴霧及びシート等の貼付による直接投与等が挙げられるが、本発明の免疫疾患治療剤及び抗炎症剤の有効性の観点から、好ましくは動脈内投与、静脈内投与及び脳室内投与であり、対象者の負担の軽減の観点から、より好ましくは静脈内投与、筋肉内投与、鼻腔内投与が好ましい。 The route of administration of the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention to the subject is oral administration, subcutaneous administration, intramuscular administration, intravenous administration, intraarterial administration, umbilical cord intravenous administration, intracerebroventricular administration, intrathecal administration. , Intraperitoneal administration, sublingual administration, transrectal administration, transvaginal administration, intraocular administration, nasal administration, inhalation, transdermal administration, implant, direct administration by spraying on the surface of an organ and sticking a sheet, etc. However, from the viewpoint of the efficacy of the therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention, it is preferably intraarterial administration, intravenous administration and intraventricular administration, and more preferably intravenous from the viewpoint of reducing the burden on the subject. Inner administration, intramuscular administration, intranasal administration are preferred.
 本発明の免疫疾患治療剤及び抗炎症剤において、その用量(投与量)は、患者の状態(体重、年齢、症状、体調等)、及び本発明の免疫疾患治療剤及び抗炎症剤の剤形等によって異なりうるが、十分な免疫疾患治療剤及び抗炎症剤の治療効果を奏する観点からは、その量は多い方が好ましい傾向にあり、一方、副作用の発現を抑制する観点からはその量は少ない方が好ましい傾向にある。通常、成人に投与する場合には、細胞数として、1×10~1×1012個/回、好ましくは1×10~1×1011個/回、より好ましくは1×10~1×1010個/回、さらに好ましくは5×10~1×10個/回である。また、患者の体重あたりの投与量としては、1×10~5×1010個/kg、好ましくは1×10~5×10個/kg、より好ましくは1×10~5×10個/kg、さらに好ましくは1×10~5×10個/kgである。新生児に投与する場合には、細胞数として、1×10~1×1011個/回、好ましくは1×10~1×1010個/回、より好ましくは1×10~1×10個/回、さらに好ましくは5×10~5×10個/回である。また、患者の体重あたりの投与量としては、1×10~5×1010個/kg、好ましくは1×10~5×10個/kg、より好ましくは1×10~5×10個/kg、さらに好ましくは1×10~5×10個/kgである。なお、本用量を1回量として、複数回投与してもよく、本用量を複数回に分けて投与してもよい。 In the therapeutic agent for immune diseases and anti-inflammatory agent of the present invention, the dose (dosage) depends on the patient's condition (weight, age, symptoms, physical condition, etc.), and the dosage form of the therapeutic agent for anti-inflammatory diseases and anti-inflammatory agent of the present invention. Although it may vary depending on the like, from the viewpoint of exhibiting sufficient therapeutic effect of the therapeutic agent for immune diseases and anti-inflammatory agent, the larger amount tends to be preferable, while the amount from the viewpoint of suppressing the occurrence of side effects is A smaller amount tends to be preferable. Usually, when administered to an adult, the number of cells is 1×10 3 to 1×10 12 cells/time, preferably 1×10 4 to 1×10 11 cells/time, more preferably 1×10 5 to It is 1×10 10 pieces/time, more preferably 5×10 6 to 1×10 9 pieces/time. The dose per body weight of the patient is 1×10 to 5×10 10 cells/kg, preferably 1×10 2 to 5×10 9 cells/kg, and more preferably 1×10 3 to 5×10 5. 8 pieces/kg, more preferably 1×10 4 to 5×10 7 pieces/kg. When administered to a newborn, the number of cells is 1×10 3 to 1×10 11 cells/time, preferably 1×10 4 to 1×10 10 cells/time, more preferably 1×10 5 to 1×. The number is 10 9 pieces/time, more preferably 5×10 5 to 5×10 8 pieces/time. The dose per body weight of the patient is 1×10 to 5×10 10 cells/kg, preferably 1×10 2 to 5×10 9 cells/kg, and more preferably 1×10 3 to 5×10 5. 8 pieces/kg, more preferably 1×10 4 to 5×10 7 pieces/kg. It should be noted that this dose may be a single dose and may be administered in multiple doses, or the dose may be administered in multiple doses.
 本発明の免疫疾患治療剤及び抗炎症剤は、一又は二以上の他の薬剤と共に投与してもよい。他の薬剤としては、免疫疾患治療剤もしくは抗炎症剤として用いることができる任意の薬剤が挙げられ、たとえば、アブシキシマブ、アダリムマブ、アレムツズマブ、バシリキシマブ、ババシズマブ、セツキシマブ、ダクリズマブ、エファリズマブ、ゲムツズマブ、イブリツモマブ、インフリキシマブ、メポリズマブ、OKT3、オマルズマブ、パリビズマブ、ペキセリズマブ、リツキシマブ、トシツモマブ、トラスツズマブ等のモノクローナル抗体、アレファセプト、デニロイキンジフチトクス、エタネルセプト等の融合蛋白、アナキンラ等の可溶性サイトカインレセプター、IFN-α、IFN-β、IFN-γ、IL-2、IL-11、G-CSF、GM-CSF等のサイトカイン、マレイン酸アザタジン、マレイン酸ブロムフェニラミン、マレイン酸クロルフェニラミン、フマル酸クレマスチン、塩酸シプロヘプタジン、マレイン酸dクロルフェニラミン、塩酸ジフェンヒドラミン、塩酸ジフェニルピラリン、塩酸ヒドロキシジン、塩酸メトジラジン、塩酸プロメタジン、酒石酸トリメプラジン、クエン酸ロエイペレンアミン、塩酸トリペレンアミン、塩酸トリプロリジン、アクリバスチン、セチリジン、デスロラタジン、エバスチン、フェキソフェナジン、レボセチリジン、ロラタジン、ミゾラスチン等のH1ブロッカー、シメチジン等のH2ブロッカー、ジプロピオン酸ベクロメタゾン、ブデソニド、フルニソリド、フルチカゾン、トリアムシノロンアセトニド、デキサメタゾン、メチルプレドニゾロン等のコルチコステロイド、クロモリン、ネドクロミル等の肥満細胞安定薬、アルブテロール等のβ作動薬、ダウノマイシン、エトポシド、6-メルカプトプリン等の細胞毒性薬、抗ヒスタミン薬、NSAID、エピネフリン、グルカゴン、アスピリンが挙げられる。また、本発明の免疫疾患治療剤もしくは抗炎症剤の投与とともに、低温療法を合わせて行う事もできる。 The therapeutic agent for immune diseases and the anti-inflammatory agent of the present invention may be administered together with one or more other drugs. The other drug includes any drug that can be used as a therapeutic agent for immune disorders or an anti-inflammatory agent, and examples thereof include abciximab, adalimumab, alemtuzumab, basiliximab, bavacizumab, cetuximab, daclizumab, efalizumab, gemtuzumab, infliximab, infliximab, infliximab, and infliximab. Monoclonal antibodies such as mepolizumab, OKT3, omaluzumab, palivizumab, pexelizumab, rituximab, tositumomab, trastuzumab, alefacept, fusion protein such as denileukin diftitox, etanercept, soluble cytokine receptors such as anakinra, IFN-IF, IFN-β, IFN-IF. -Γ, IL-2, IL-11, G-CSF, GM-CSF, and other cytokines, azatadine maleate, brompheniramine maleate, chlorpheniramine maleate, clemastine fumarate, cyproheptadine hydrochloride, d chlorphene maleate Lamin, diphenhydramine hydrochloride, diphenylpyraline hydrochloride, hydroxyzine hydrochloride, metzirazine hydrochloride, promethazine hydrochloride, trimeprazine tartrate, loeiperenamine citrate, triperenamine hydrochloride, triprrolidine hydrochloride, acrivastine, cetirizine, desloratadine, ebastine, fexofenadine , H1 blockers such as levocetirizine, loratadine and mizolastine, H2 blockers such as cimetidine, beclomethasone dipropionate, budesonide, flunisolide, fluticasone, corticosteroids such as triamcinolone acetonide, dexamethasone, methylprednisolone, obesity cell stable such as cromolyn, nedocromil, etc. Drugs, β-agonists such as albuterol, cytotoxic drugs such as daunomycin, etoposide, 6-mercaptopurine, antihistamines, NSAIDs, epinephrine, glucagon, and aspirin. In addition, cryotherapy can be combined with administration of the therapeutic agent for immune diseases or anti-inflammatory agent of the present invention.
 本発明の間葉系幹細胞は様々な自己免疫疾患及び炎症性疾患に用いることができるが、具体的疾患としては、GVHD、軟骨分解、関節リウマチ、全身性エリテマトーデス、乾癬性関節炎、脊椎関節炎、変形性関節症、痛風、乾癬、多発性硬化症、筋萎縮性側索硬化症、アルツハイマー病、パーキンソン病、うっ血性心不全、脳卒中、大動脈弁狭窄症、腎不全、ネフローゼ症候群、尿毒症、狼瘡、膵炎、アレルギー、線維症、貧血、アテローム性動脈硬化症、再狭窄、化学療法/放射線関連合併症、I型糖尿病、II型糖尿病、肝不全、自己免疫性肝炎、C型肝炎、原発性胆汁性肝硬変、原発性硬化性胆管炎、劇症肝炎、セリアック病、非特異性大腸炎、腸リンパ管拡張症、蛋白喪失性腸症、クローン病、アレルギー性結膜炎、糖尿病性網膜症、シェーグレン症候群、アトピー性疾患、ブドウ膜炎、アレルギー性鼻炎食物性アレルギー、アナフィラキシー、自己免疫疾患、薬物過敏症、肥満細胞症、喘息、石綿症、珪肺、慢性閉塞性肺疾患、慢性肉芽腫性炎症、嚢胞性線維症、組織球症、サルコイドーシス、糸球体腎炎、脈管炎、皮膚炎、HIV関連悪液質、大脳マラリア、強直性脊椎炎、らい病、COPD、肺線維症、線維筋痛、食道癌等の癌、胃食道逆流症、バレット食道、再生不良性貧血、移植片対宿主病、鎌状赤血球症、CVID、高IgM症候群、IgA欠損症、一過性低ガンマグロブリン血症、X連鎖無ガンマグロブリン血症、慢性皮膚粘膜カンジダ症、ディ・ジョージ症候群、X連鎖リンパ球増殖性症候群、毛細血管拡張性運動失調症、軟骨毛髪形成不全症、複合免疫不全、高IgE症候群、MHC欠損症、重症複合免疫不全症、ヴィスコットーオールドリッチ症候群、チェディアックー東症候群、慢性肉芽腫性疾患、白血球接着欠乏症、IFN-γレセプター欠損症、インターロイキン(IL)-12欠損症、IL-12レセプターβ1欠損症、ZAP―70欠損症、血管性浮腫等が挙げられる。 The mesenchymal stem cells of the present invention can be used for various autoimmune diseases and inflammatory diseases. Specific diseases include GVHD, cartilage degradation, rheumatoid arthritis, systemic lupus erythematosus, psoriatic arthritis, spondyloarthritis, and deformity. Osteoarthritis, gout, psoriasis, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, congestive heart failure, stroke, aortic stenosis, renal failure, nephrotic syndrome, uremia, lupus, pancreatitis , Allergy, fibrosis, anemia, atherosclerosis, restenosis, chemotherapy/radiation related complications, type I diabetes, type II diabetes, liver failure, autoimmune hepatitis, hepatitis C, primary biliary cirrhosis , Primary sclerosing cholangitis, fulminant hepatitis, celiac disease, nonspecific colitis, intestinal lymphangiectasia, protein-losing enteropathy, Crohn's disease, allergic conjunctivitis, diabetic retinopathy, Sjogren's syndrome, atopic Disease, uveitis, allergic rhinitis, food allergy, anaphylaxis, autoimmune disease, drug hypersensitivity, mastocytosis, asthma, asbestosis, silicosis, chronic obstructive pulmonary disease, chronic granulomatous inflammation, cystic fibrosis , Histiocytosis, sarcoidosis, glomerulonephritis, vasculitis, dermatitis, HIV-related cachexia, cerebral malaria, ankylosing spondylitis, leprosy, COPD, pulmonary fibrosis, fibromyalgia, esophageal cancer, etc. , Gastroesophageal reflux disease, Barrett's esophagus, aplastic anemia, graft-versus-host disease, sickle cell disease, CVID, high IgM syndrome, IgA deficiency, transient hypogammaglobulinemia, X-linked agammaglobulinemia Disease, chronic cutaneous mucosal candidiasis, Di George syndrome, X-linked lymphoproliferative syndrome, ataxia-telangiectasia, cartilage dysplasia, combined immunodeficiency, high IgE syndrome, MHC deficiency, severe combined immunity Deficiency, Viscott-Aldrich syndrome, Chediak-East syndrome, chronic granulomatous disease, leukocyte adhesion deficiency, IFN-γ receptor deficiency, interleukin (IL)-12 deficiency, IL-12 receptor β1 deficiency, Examples include ZAP-70 deficiency and angioedema.
 本発明は、キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、免疫疾患の治療方法、IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、免疫疾患の治療方法、JAK/STAT経路が、定常状態で活性化している間葉系幹細胞を用いることを特徴とする、免疫疾患の治療方法も含む。また、本発明は、キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、炎症性疾患の治療方法、IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、炎症性疾患の治療方法、JAK/STAT経路が、定常状態で活性化している間葉系幹細胞を用いることを特徴とする、炎症性疾患の治療方法も含む。さらに、本発明は、キヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、GVHDの治療方法、IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高い間葉系幹細胞を用いることを特徴とする、GVHDの治療方法、JAK/STAT経路が、定常状態で活性化している間葉系幹細胞を用いることを特徴とする、GVHDの治療方法も含む。なお、各文言の説明については、上述の間葉系幹細胞、免疫疾患治療剤、抗炎症剤、GVHD治療剤における説明を適用できる。 The present invention is characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, a method for treating an immune disease, kynurenine synthesized from tryptophan by IDO induced intracellularly by IFNγ treatment. A method for treating an immune disease, characterized by using mesenchymal stem cells having high production or kynurenine secretion, characterized by using mesenchymal stem cells in which JAK/STAT pathway is activated in a steady state Also included are methods of treating immune disorders. Further, the present invention is characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, a method for treating an inflammatory disease, and IDO induced intracellularly by IFNγ treatment to synthesize from tryptophan. Method for treating inflammatory diseases, characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, and using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state Also included is a method for treating an inflammatory disease, which is characterized in that Furthermore, the present invention is characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, a method for treating GVHD, and IDO induced intracellularly by IFNγ treatment to synthesize from tryptophan. A method for treating GVHD, characterized by using mesenchymal stem cells having a high kynurenine production amount or a high kynurenine secretion amount, characterized by using mesenchymal stem cells in which the JAK/STAT pathway is activated in a steady state, The method of treating GVHD is also included. In addition, regarding the description of each wording, the description regarding the mesenchymal stem cells, the therapeutic agent for immune diseases, the anti-inflammatory agent, and the therapeutic agent for GVHD can be applied.
 以下に、実施例及び試験例を挙げて本発明を詳細に説明するが、本発明はこれらの実施例等によって限定されるものではない。 Hereinafter, the present invention will be described in detail with reference to Examples and Test Examples, but the present invention is not limited to these Examples and the like.
[臍帯由来間葉系幹細胞(UCMSC-EXP、UCMSC-CP)の調製及び培養]
 臍帯由来細胞は、Cytotherapy, 18, 229-241, 2016に記載の方法で採取した。簡潔には、東京大学医科学研究所の倫理委員会の承認を得た上で、提供者の同意を得て採取された臍帯を1から2mmの断片に細断し、培養皿上へ播種し、セルアミーゴ(株式会社 椿本チエイン)を被せ、10% fetal bovine serum(FBS)と抗生物質を添加したα-minimal essential medium(MEM-α)中で培養する、「改良エクスプラント法」により、臍帯由来間葉系幹細胞(以下「UCMSC-EXP」という)を得た。
[Preparation and culture of umbilical cord-derived mesenchymal stem cells (UCMSC-EXP, UCMSC-CP)]
Umbilical cord-derived cells were collected by the method described in Cytotherapy, 18, 229-241, 2016. Briefly, with the approval of the Ethics Committee of the Institute of Medical Science, the University of Tokyo, the umbilical cord collected with the consent of the donor was shredded into 1 to 2 mm 3 pieces and seeded on a culture dish. Then, cell amigo (Tsubakimoto Chain Co., Ltd.) is covered, and the cells are cultured in α-minimal essential medium (MEM-α) supplemented with 10% fetal bovine serum (FBS) and antibiotics by the “improved explant method”. Umbilical cord-derived mesenchymal stem cells (hereinafter referred to as "UCMSC-EXP") were obtained.
 同様に、臍帯由来細胞は、Cytotherapy, 18, 229-241, 2016に記載の方法で採取して、東京大学医科学研究所の倫理委員会の承認を得た上で、提供者の同意を得て採取された臍帯を1から2mmの大きさに切断し、コラゲナーゼで処理した後に、遠心分離(400×gで5分間)して得られた細胞を間葉系幹細胞用無血清培地(Rohto社製)で、培養フラスコ中、付着培養する、「コラゲナーゼ法」により、臍帯由来間葉系幹細胞(以下「UCMSC-CP」という)を得た。 Similarly, umbilical cord-derived cells were collected by the method described in Cytotherapy, 18, 229-241, 2016, and after obtaining the approval of the Ethics Committee of the Institute of Medical Science, University of Tokyo, with the consent of the donor. The collected umbilical cord was cut into a size of 1 to 2 mm 3 , treated with collagenase, and then centrifuged (400×g for 5 minutes) to obtain cells, which were then used as serum-free medium for mesenchymal stem cells (Rohto Umbilical cord-derived mesenchymal stem cells (hereinafter referred to as "UCMSC-CP") were obtained by "collagenase method" in which the cells were adherently cultured in a culture flask.
 なお、継代数については、臍帯組織から前述の改良エクスプラント法もしくはコラゲナーゼ法により得られた細胞を第一継代細胞(P1)とし、継代を行う事により継代数が進み、第二継代細胞(P2)、第三継代細胞(P3)のように記載する。 Regarding the passage number, the cells obtained from the umbilical cord tissue by the above-mentioned improved explant method or collagenase method were used as the first passage cells (P1). Cell (P2), third passage cell (P3), etc.
 得られたUCMSC-EXP及びUCMSC-CPを、細胞剥離液(TrypLE Select (1X))を用いて剥離し、遠沈管に移し、400×gで5分間、遠心分離して細胞の沈殿を得た。上清を除去した後、細胞凍結保存液(STEM-CELLBANKER(ゼノアック社))を適量加え懸濁した。当該細胞懸濁溶液を、クライオチューブに分注し、フリーザー内で-80℃にて保存した。その後、液体窒素上の気相に移し、保存を継続した。 The obtained UCMSC-EXP and UCMSC-CP were detached using a cell detachment solution (TrypLE Select (1X)), transferred to a centrifuge tube, and centrifuged at 400×g for 5 minutes to obtain a cell precipitate. .. After removing the supernatant, an appropriate amount of cell cryopreservation solution (STEM-CELLBANKER (Zenoac)) was added and suspended. The cell suspension solution was dispensed into a cryotube and stored at -80°C in a freezer. Then, it moved to the vapor phase on liquid nitrogen, and continued preservation.
[平面培養細胞(ADH)と浮遊培養細胞(SUS)の細胞内キヌレニン量の測定]
 前述の、改良エクスプラント法により得られた臍帯組織由来凍結細胞であるUCMSC-EXPを起眠し、細胞を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加して、間葉系幹細胞用無血清培地(Rohto社)を用いて攪拌培養を行い、8日間浮遊培養細胞(以下、「EXP-SUS」と言う)を得た。同様に、前述のUCMSC-EXPを起眠し、細胞を細胞培養フラスコに播種して間葉系幹細胞用無血清培地(Rohto社)を用いて平面培養行い、8日間培養し、平面培養細胞(以下、「EXP-ADH」と言う)を得た。上記方法により8日間の培養期間により得られた平面培養細胞(EXP-ADH)、または浮遊培養細胞(EXP-SUS)の細胞内容物を抽出し、キャピラリー電気泳動法及びLC-MSにより細胞内に蓄積した代謝物の網羅的解析(メタボローム解析)を行った。その結果、EXP-ADHよりもEXP-SUSにおいて、細胞内キヌレニン蓄積量が高くなっていることがわかった(図1)。また、培養上清中のキヌレニン量をELISA法(Immundiagnostik GmbH、品番K7728)により定量した。こちらの定量法でも、EXP-ADHよりもEXP-SUSにおいて、8日間培養後の細胞内キヌレニン蓄積量が高いという結果が得られた(図2)。これにより、定常状態において浮遊培養により細胞内キヌレニン産生が亢進していることが明らかになった。
[Measurement of intracellular kynurenine amount in flat culture cells (ADH) and suspension culture cells (SUS)]
UMCSC-EXP, which is the umbilical cord tissue-derived frozen cells obtained by the improved explant method described above, were put to sleep, and the cell suspension containing the cells and the microcarriers were mixed, added to the culture tank, and the mesenchyme was added. Stirring culture was performed using a serum-free medium for line stem cells (Rohto) to obtain suspension-cultured cells (hereinafter referred to as “EXP-SUS”) for 8 days. Similarly, the above-described UCMSC-EXP was put to sleep, cells were seeded in a cell culture flask, subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto), and cultured for 8 days to obtain flat culture cells ( Hereinafter referred to as "EXP-ADH"). The cell contents of flat culture cells (EXP-ADH) or suspension culture cells (EXP-SUS) obtained by a culture period of 8 days by the above method were extracted, and the cells were intracellularized by capillary electrophoresis and LC-MS. A comprehensive analysis (metabolome analysis) of accumulated metabolites was performed. As a result, it was found that the intracellular kynurenine accumulation amount was higher in EXP-SUS than in EXP-ADH (FIG. 1). In addition, the amount of kynurenine in the culture supernatant was quantified by an ELISA method (Immundiagnostic GmbH, product number K7728). This quantification method also resulted in a higher intracellular kynurenine accumulation amount after 8 days of culture in EXP-SUS than in EXP-ADH (FIG. 2). This revealed that intracellular kynurenine production was enhanced by suspension culture in the steady state.
[浮遊培養細胞(SUS)と平面培養細胞(ADH)のキヌレニン産生及びIDO活性の測定1]
 前述の、コラゲナーゼ法により得られた臍帯組織由来凍結細胞であるUCMSC-CPを起眠し、細胞を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加して、間葉系幹細胞用無血清培地(Rohto社)を用いて攪拌培養を行い、4日間浮遊培養細胞(以下、「CP-SUS」と言う)を得た。同様に、前述のUCMSC-CPを起眠し、細胞を細胞培養フラスコに播種して間葉系幹細胞用無血清培地(Rohto社)を用いて平面培養行い、4日間培養し、平面培養細胞(以下、「CP-ADH」と言う)を得た。最終濃度が100ng/mL及び200ng/mLとなる量のIFNγ(品番:AF-300-02、PeproTeck, Inc.社製)を添加し、48時間後の細胞上清中のキヌレニン量を、Ehrlich試薬を使用した比色法により測定した(IFNγの濃度100ng/mL群及び200ng/mL群)。また、この反応がIDOを介したものである事を確認する目的で、IFNγ(100ng/mL)を添加し、同時にIDO阻害剤(30nM、Epacadostat、CAS No:1204669-58-8)を添加した群(阻害剤群)も設定した。なお、比較対象としてIFNγを添加しないコントロール群についても培養上清中のキヌレニンの定量を行った。また、各間葉系幹細胞の細胞活性をWST-8(Cell Counting Kit-8、同仁化学研究所)にて測定し、キヌレニンの測定値を補正した。結果を図3に示す。IDO阻害剤によりキヌレニン産生が抑えられたことから、各試料のキヌレニン産生能を指標としてIDO活性を評価することができる。
[Measurement of kynurenine production and IDO activity of suspension culture cells (SUS) and flat culture cells (ADH) 1]
The above-mentioned frozen cells of umbilical cord tissue-derived UCMSC-CP obtained by the collagenase method were put to sleep, and the cell suspension containing the cells and the microcarriers were mixed and added to a culture tank to give mesenchymal stem cells. Stirring culture was performed using a serum-free medium (Rohto) for 4 days to obtain suspension-cultured cells (hereinafter referred to as "CP-SUS"). Similarly, the aforementioned UCMSC-CP was put to sleep, the cells were seeded in a cell culture flask, and the cells were subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto), and cultured for 4 days to obtain flat culture cells ( Hereinafter, referred to as "CP-ADH"). IFNγ (product number: AF-300-02, manufactured by PeproTeck, Inc.) was added in an amount to give final concentrations of 100 ng/mL and 200 ng/mL, and the amount of kynurenine in the cell supernatant after 48 hours was determined using the Ehrlich reagent. (IFNγ concentration of 100 ng/mL group and 200 ng/mL group). In addition, in order to confirm that this reaction was mediated by IDO, IFNγ (100 ng/mL) was added, and at the same time, an IDO inhibitor (30 nM, Epacadostat, CAS No: 1204669-58-8) was added. A group (inhibitor group) was also set up. As a control, kynurenine in the culture supernatant was also quantified for a control group to which IFNγ was not added. In addition, the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories) to correct the measured value of kynurenine. Results are shown in FIG. Since the kynurenine production was suppressed by the IDO inhibitor, the IDO activity can be evaluated using the kynurenine production ability of each sample as an index.
 CP-SUSでは、CP-ADHに比べ、いずれの条件でもIFNγ刺激に応答したキヌレニンの培養上清中への放出量が多いことが明らかとなった。これにより、CP-SUSの方がCP-ADHよりもIFNγ刺激によるIDOの誘導及び活性化、それに伴うキヌレニンの細胞内での産生及び培養上清中への分泌能が高いことが分かった(図3)。 It was revealed that CP-SUS released more kynurenine in response to IFNγ stimulation into the culture supernatant than CP-ADH under all conditions. From this, it was found that CP-SUS was higher than CP-ADH in the induction and activation of IDO by IFNγ stimulation, the accompanying production of kynurenine in the cell, and the higher secretion ability into the culture supernatant (Fig. 3).
[浮遊培養細胞(SUS)と平面培養細胞(ADH)のキヌレニン産生及びIDO活性の測定2]
 前述の臍帯組織由来凍結細胞(UCMSC-EXP)を起眠し、細胞を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加して、間葉系幹細胞用無血清培地(Rohto社)を用いて攪拌培養を行い、8日間浮遊培養細胞(以下、「EXP-SUS」と言う)を得た。同様に、前述の臍帯組織由来凍結細胞(UCMSC-EXP)を起眠し、細胞を細胞培養フラスコにて播種して間葉系幹細胞用無血清培地(Rohto社)を用いて平面培養行い、4日間培養後継代を行いさらに4日間培養し、平面培養細胞(以下、「EXP-ADH」と言う)を得た。最終濃度が50ng/mL及び100ng/mLとなる量のIFNγ(品番:AF-300-02、PeproTeck, Inc.社製)を添加し、64時間後の細胞上清中のキヌレニン量を、Ehrlich試薬を使用した比色法により測定した(IFNγの濃度50ng/mL群及び100ng/mL群)。なお、比較対象としてIFNγを添加しないコントロール群についても培養上清中のキヌレニンの定量を行った。また、各間葉系幹細胞の細胞活性をWST-8(Cell Counting Kit-8、同仁化学研究所)にて測定し、キヌレニンの測定値を補正し、各試料のキヌレニン産生能を指標としてIDO活性を評価した。EXP-ADHのコントロール群のIDO/WST8値を1として、IDO活性比率を算出した(図4)。
[Measurement of kynurenine production and IDO activity of suspension culture cells (SUS) and flat culture cells (ADH) 2]
The above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-EXP) were put to sleep, the cell suspension containing the cells and the microcarriers were mixed, added to a culture tank, and serum-free medium for mesenchymal stem cells (Rohto ) Was used to carry out agitation culture to obtain suspension-cultured cells for 8 days (hereinafter referred to as "EXP-SUS"). Similarly, the above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-EXP) were put to sleep, and the cells were seeded in a cell culture flask and subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto). After culturing for one day, the cells were subcultured and further cultivated for 4 days to obtain flat-cultured cells (hereinafter referred to as "EXP-ADH"). IFNγ (product number: AF-300-02, manufactured by PeproTeck, Inc.) was added in an amount to give final concentrations of 50 ng/mL and 100 ng/mL, and the amount of kynurenine in the cell supernatant after 64 hours was determined by the Ehrlich reagent. (IFNγ concentration of 50 ng/mL group and 100 ng/mL group). As a control, kynurenine in the culture supernatant was also quantified for a control group to which IFNγ was not added. In addition, the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity. Was evaluated. The IDO activity ratio was calculated by setting the IDO/WST8 value of the EXP-ADH control group to 1 (FIG. 4).
 EXP-SUSでは、EXP-ADHに比べいずれの条件でもIDO活性が高いことが明らかとなった。これにより、EXP-SUSの方がEXP-ADHよりもIFNγ刺激によるIDOの誘導及び活性化、それに伴うキヌレニンの細胞内での産生及び培養上清中への分泌能が高いことが分かった。 It was revealed that EXP-SUS has higher IDO activity under any condition than EXP-ADH. From this, it was found that EXP-SUS was higher than EXP-ADH in the induction and activation of IDO by IFNγ stimulation, the accompanying production of kynurenine in the cell, and the higher secretion ability into the culture supernatant.
[浮遊培養細胞(SUS)と平面培養細胞(ADH)のキヌレニン産生及びIDO活性の測定3]
 前述の臍帯組織由来凍結細胞(UCMSC-CP)を起眠し、細胞を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加して、間葉系幹細胞用無血清培地(Rohto社)を用いて攪拌培養を行い、8日間浮遊培養細胞(以下、「CP-SUS」と言う)を得た。同様に、UCMSC-CPを起眠し、細胞を細胞培養フラスコにて播種して間葉系幹細胞用無血清培地(Rohto社)を用いて平面培養行い、8日間培養し、平面培養細胞(以下、「CP-ADH」と言う)を得た。最終濃度が50ng/mL及び100ng/mLとなる量のIFNγ(品番:AF-300-02、PeproTeck, Inc.社製)を添加し、64時間後の細胞上清中のキヌレニン量を、Ehrlich試薬を使用した比色法により測定した(IFNγの濃度50ng/mL群及び100ng/mL群)。なお、比較対象としてIFNγを添加しないコントロール群についてもIDO活性の測定を行った。また、各間葉系幹細胞の細胞活性をWST-8(Cell Counting Kit-8、同仁化学研究所)にて測定し、キヌレニンの測定値を補正し、各試料のキヌレニン産生能を指標としてIDO活性を評価した。CP-ADHのコントロール群のIDO/WST8値を1として、IDO活性比率を算出した(図5)。
[Measurement of kynurenine production and IDO activity of suspension culture cells (SUS) and flat culture cells (ADH) 3]
The above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-CP) were put to sleep, the cell suspension containing the cells and the microcarriers were mixed, added to a culture tank, and serum-free medium for mesenchymal stem cells (Rohto) ) Was used to carry out agitation culture to obtain suspension-cultured cells for 8 days (hereinafter referred to as "CP-SUS"). Similarly, UCMSC-CP was put to sleep, the cells were seeded in a cell culture flask, and the cells were subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto), and cultured for 8 days. , "CP-ADH") was obtained. IFNγ (product number: AF-300-02, manufactured by PeproTeck, Inc.) was added in an amount to give final concentrations of 50 ng/mL and 100 ng/mL, and the amount of kynurenine in the cell supernatant after 64 hours was determined by the Ehrlich reagent. (IFNγ concentration of 50 ng/mL group and 100 ng/mL group). The IDO activity was also measured for a control group to which IFNγ was not added as a comparison target. In addition, the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity. Was evaluated. The IDO activity ratio was calculated by setting the IDO/WST8 value of the CP-ADH control group to 1 (FIG. 5).
 CP-SUSでは、CP-ADHに比べいずれの条件でもIDO活性が高いことが明らかとなった。これにより、CP-SUSの方がCP-ADHよりもIFNγ刺激によるIDOの誘導及び活性化、それに伴うキヌレニンの細胞内での産生及び培養上清中への分泌能が高いことが分かった。 It was revealed that CP-SUS has higher IDO activity under all conditions than CP-ADH. From this, it was found that CP-SUS was higher than CP-ADH in the induction and activation of IDO by IFNγ stimulation, the accompanying production of kynurenine in cells, and the secretion ability into the culture supernatant.
[浮遊培養細胞(SUS)と平面培養細胞(ADH)のキヌレニン産生及びIDO活性の測定4]
 前述の臍帯組織由来凍結細胞(UCMSC-CP)を起眠し、細胞を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加して、間葉系幹細胞用無血清培地(Rohto社)を用いて攪拌培養を行い、8日間浮遊培養細胞(以下、「CP-SUS」と言う)を得た。同様に、前述の臍帯組織由来凍結細胞(UCMSC-CP)を起眠し、細胞を細胞培養フラスコにて播種して間葉系幹細胞用無血清培地(Rohto社)を用いて平面培養行い、8日間培養し、平面培養細胞(以下、「CP-ADH」と言う)を得た。最終濃度が50ng/mL及び100ng/mLとなる量のIFNγ(品番:AF-300-02、PeproTeck, Inc.社製)を添加し、90時間後の細胞上清中のキヌレニン量を、Ehrlich試薬を使用した比色法により測定した(IFNγの濃度50ng/mL群及び100ng/mL群)。なお、比較対象としてIFNγを添加しないコントロール群についてもIDO活性の測定を行った。また、各間葉系幹細胞の細胞活性をWST-8(Cell Counting Kit-8、同仁化学研究所)にて測定し、キヌレニンの測定値を補正し、各試料のキヌレニン産生能を指標としてIDO活性を評価した。CP-ADHのコントロール群のIDO/WST8値を1として、IDO活性比率を算出した(図6)。
[Measurement of kynurenine production and IDO activity of suspension culture cells (SUS) and flat culture cells (ADH) 4]
The above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-CP) were put to sleep, the cell suspension containing the cells and the microcarriers were mixed, added to a culture tank, and serum-free medium for mesenchymal stem cells (Rohto) ) Was used to carry out agitation culture to obtain suspension-cultured cells for 8 days (hereinafter referred to as "CP-SUS"). Similarly, the above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-CP) were put to sleep, and the cells were seeded in a cell culture flask and subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto). The cells were cultured for one day to obtain flat culture cells (hereinafter referred to as "CP-ADH"). IFNγ (product number: AF-300-02, manufactured by PeproTeck, Inc.) was added in an amount to give final concentrations of 50 ng/mL and 100 ng/mL, and the amount of kynurenine in the cell supernatant 90 hours later was determined by the Ehrlich reagent. (IFNγ concentration of 50 ng/mL group and 100 ng/mL group). The IDO activity was also measured for a control group to which IFNγ was not added as a comparison target. In addition, the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity. Was evaluated. The IDO activity ratio was calculated by setting the IDO/WST8 value of the CP-ADH control group to 1 (FIG. 6).
 CP-SUSでは、CP-ADHに比べいずれの条件でもIDO活性が高いことが明らかとなった。これにより、CP-SUSの方がCP-ADHよりもIFNγ刺激によるIDOの誘導及び活性化、それに伴うキヌレニンの細胞内での産生及び培養上清中への分泌能が高いことが分かった。 It was revealed that CP-SUS has higher IDO activity under all conditions than CP-ADH. From this, it was found that CP-SUS was higher than CP-ADH in the induction and activation of IDO by IFNγ stimulation, the accompanying production of kynurenine in cells, and the secretion ability into the culture supernatant.
[浮遊培養細胞(SUS)と平面培養細胞(ADH)のキヌレニン産生及びIDO活性の測定5]
 前述の臍帯組織由来凍結細胞(UCMSC-EXP)を起眠し、細胞を含んだ細胞懸濁液とマイクロキャリアを混合し、培養槽に添加して、間葉系幹細胞用無血清培地(Rohto社)を用いて攪拌培養を行い、8日間浮遊培養細胞(以下、「EXP-SUS」と言う)を得た。同様に、前述の臍帯組織由来凍結細胞(UCMSC-EXP)を起眠し、細胞を細胞培養フラスコに播種して間葉系幹細胞用無血清培地(Rohto社)を用いて平面培養行い、8日間培養し、平面培養細胞(以下、「EXP-ADH」と言う)を得た。最終濃度が50ng/mL及び100ng/mLとなる量のIFNγ(品番:AF-300-02、PeproTeck, Inc.社製)を添加し、90時間後の細胞上清中のキヌレニン量を、Ehrlich試薬を使用した比色法により測定した(50ng/mL群及び100ng/mL群)。なお、比較対象としてIFNγを添加しないコントロール群についてもIDO活性の測定を行った。また、各間葉系幹細胞の細胞活性をWST-8(Cell Counting Kit-8、同仁化学研究所)にて測定し、キヌレニンの測定値を補正し、各試料のキヌレニン産生能を指標としてIDO活性を評価した。EXP-ADHのコントロール群のIDO/WST8値を1として、IDO活性比率を算出した(図7)。
[Measurement of kynurenine production and IDO activity of suspension culture cells (SUS) and flat culture cells (ADH) 5]
The above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-EXP) were put to sleep, the cell suspension containing the cells and the microcarriers were mixed, added to a culture tank, and serum-free medium for mesenchymal stem cells (Rohto ) Was used to carry out agitation culture to obtain suspension-cultured cells for 8 days (hereinafter referred to as "EXP-SUS"). Similarly, the above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-EXP) were put to sleep, and the cells were seeded in a cell culture flask and subjected to flat culture using a serum-free medium for mesenchymal stem cells (Rohto) for 8 days. The cells were cultured to obtain flat culture cells (hereinafter referred to as "EXP-ADH"). IFNγ (product number: AF-300-02, manufactured by PeproTeck, Inc.) was added in an amount to give final concentrations of 50 ng/mL and 100 ng/mL, and the amount of kynurenine in the cell supernatant 90 hours later was determined by the Ehrlich reagent. Was measured by a colorimetric method using (50 ng/mL group and 100 ng/mL group). The IDO activity was also measured for a control group to which IFNγ was not added as a comparison target. In addition, the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity. Was evaluated. The IDO activity ratio was calculated by setting the IDO/WST8 value of the EXP-ADH control group to 1 (FIG. 7).
 EXP-SUSでは、EXP-ADHに比べいずれの条件でもIDO活性が高いことが明らかとなった。これにより、EXP-SUSの方がEXP-ADHよりもIFNγ刺激によるIDOの誘導及び活性化、それに伴うキヌレニンの細胞内での産生及び培養上清中への分泌能が高いことが分かった。 It was revealed that EXP-SUS has higher IDO activity under any condition than EXP-ADH. From this, it was found that EXP-SUS was higher than EXP-ADH in the induction and activation of IDO by IFNγ stimulation, the accompanying production of kynurenine in the cell, and the higher secretion ability into the culture supernatant.
[臍帯組織由来間葉系幹細胞及びその他の細胞のキヌレニン産生及びIDO活性の測定]
 前述の臍帯組織由来凍結細胞(UCMSC-CP)を起眠し、細胞を細胞培養フラスコに播種して間葉系幹細胞用無血清培地(Rohto社)もしくは、10%血清含有MEMα培地を用いて平面培養を行い、8日間培養し、それぞれ平面培養細胞(以下、それぞれ「UCMSC-SF」、「UCMSC-MEM」と言う)を得た。また、MRC―5細胞(ATCC)、ヒト新生児由来皮膚線維芽細胞(HDF、ScienCell Research Laboratories社)を解凍し、10%血清含有MEMα培地を用いて3日間平面培養を行い、それぞれ平面培養細胞を得た。さらに、ヒト臍帯静脈内皮細胞(HUVEC、PromoCell社)を解凍し、Endotherlial cell growth medium(PromoCell社製、品番C-22210)を用いて3日間平面培養を行い、平面培養細胞を得た。UCMSC-SF、UCMSC-MEM、MRC―5、HDF及びHUVECをそれぞれ、30,000cells/cmで4日間培養して、培養上清を回収した。培養上清中のキヌレニン量を、Ehrlich試薬を使用した比色法により測定した。また、各間葉系幹細胞の細胞活性をWST-8(Cell Counting Kit-8、同仁化学研究所)にて測定し、キヌレニンの測定値を補正し、各試料のキヌレニン産生能を指標としてIDO活性を評価した(図8)。
[Measurement of kynurenine production and IDO activity of umbilical cord tissue-derived mesenchymal stem cells and other cells]
The above-mentioned umbilical cord tissue-derived frozen cells (UCMSC-CP) were put to sleep, and the cells were seeded in a cell culture flask and then plated using a serum-free medium for mesenchymal stem cells (Rohto) or a MEMα medium containing 10% serum. The cells were cultured and cultured for 8 days to obtain flat-cultured cells (hereinafter referred to as "UCMSC-SF" and "UCMSC-MEM", respectively). In addition, MRC-5 cells (ATCC) and human newborn-derived skin fibroblasts (HDF, ScienceCell Research Laboratories) were thawed and subjected to flat culture for 3 days using MEMα medium containing 10% serum. Obtained. Further, human umbilical vein endothelial cells (HUVEC, PromoCell) were thawed and subjected to planar culture for 3 days using Endotherial cell growth medium (PromoCell, product number C-22210) to obtain planar cultured cells. UCMSC-SF, UCMSC-MEM, MRC-5, HDF and HUVEC were each cultured at 30,000 cells/cm 2 for 4 days, and the culture supernatant was recovered. The amount of kynurenine in the culture supernatant was measured by a colorimetric method using Ehrlich reagent. In addition, the cell activity of each mesenchymal stem cell was measured by WST-8 (Cell Counting Kit-8, Dojindo Laboratories), the kynurenine production value was corrected, and the kynurenine-producing ability of each sample was used as an index for the IDO activity. Was evaluated (FIG. 8).
 UCMSC-SFでは、UCMSC-MEMに比べIDO活性が高いことが明らかとなった。また、UCMSC-SF及びUCMSC-MEMでは、MRC―5、HDF及びHUVECに比べIDO活性が高いことが明らかとなった。これにより、10%血清含有MEMαで培養した臍帯組織由来間葉系幹細胞に比べて、間葉系幹細胞用無血清培地で培養した臍帯組織由来間葉系幹細胞は、細胞内でのキヌレニン産生及び培養上清中への分泌能が高いことが分かった。また、臍帯組織由来間葉系幹細胞は、MRC―5、HDF及びHUVECに比べて、細胞内でのキヌレニン産生及び培養上清中への分泌能が高いことが分かった。 UCMSC-SF was found to have higher IDO activity than UCMSC-MEM. Further, it was revealed that UCMSC-SF and UCMSC-MEM have higher IDO activity than MRC-5, HDF and HUVEC. As a result, umbilical cord tissue-derived mesenchymal stem cells cultured in a serum-free medium for mesenchymal stem cells produced kynurenine in cells and cultured compared to umbilical cord tissue-derived mesenchymal stem cells cultured in MEMα containing 10% serum. It was found that the secretory ability into the supernatant was high. Further, it was found that umbilical cord tissue-derived mesenchymal stem cells have higher intracellular kynurenine production and higher secretory ability into the culture supernatant than MRC-5, HDF and HUVEC.
[平面培養細胞(ADH)と浮遊培養細胞(SUS)のマイクロアレイ解析による比較]
 上記方法により8日間の培養期間により得られた平面培養細胞(EXP-ADH)、または浮遊培養細胞(EXP-SUS)より、常法にしたがいtotal RNAを抽出し、cDNAを合成した。アジレント社のSurePrint G3 Human GE マイクロアレイ 8x60K Ver. 3.0により発現遺伝子を網羅的に解析した。その結果、Kyoto Encyclopedia of Genes and Genomes (KEGG)に登録されているJAK―STATシグナル経路に関連する遺伝子155個の内、33個が定常状態においてEXP-ADHと比較しEXP-SUSにおいて、1.5倍以上発現が高くなっていた。EXP-ADHにおける発現量対するEXP-SUSにおける発現量の比(発現量比)が大きい順に並べると、CSF3、CSF2、LIF、IL6、IL6R、STAT4、CNTFR、GHR、CBLB、IL15、IL6ST、IL10、CLCF1、CCND2、IL11RA、IL15RA、PIM1、SOCS1、STAT2、IL19、IL7、IL24、IL22RA1、SOCS3、JAK2、IFNAR1、IFNAR2、IFNGR2、PRL、LIFR、SPRY2、AKT3、IL4Rの順となっていた。図9に上位9個の遺伝子の発現量比を示した。
[Comparison of flat culture cells (ADH) and suspension culture cells (SUS) by microarray analysis]
Total RNA was extracted from the flat-cultured cells (EXP-ADH) or the suspension-cultured cells (EXP-SUS) obtained by the above method for a culture period of 8 days according to a conventional method to synthesize cDNA. Agilent SurePrint G3 Human GE Microarray 8x60K Ver. The expressed genes were comprehensively analyzed by 3.0. As a result, 33 out of 155 genes related to the JAK-STAT signal pathway registered in Kyoto Encyclopedia of Genes and Genomes (KEGG) were compared to EXP-ADH in the steady state, and in EXP-SUS, 1. The expression was higher than 5-fold. When arranged in descending order of the ratio of the expression level in EXP-ADH to the expression level in EXP-SUS (expression ratio), CSF3, CSF2, LIF, IL6, IL6R, STAT4, CNTFR, GHR, CBLB, IL15, IL6ST, IL10, The order was CLCF1, CCND2, IL11RA, IL15RA, PIM1, SOCS1, STAT2, IL19, IL7, IL24, IL22RA1, SOCS3, JAK2, IFNAR1, IFNAR2, IFNGR2, PRL, LIFR, SPRY2, AKT3, IL4R. FIG. 9 shows the expression level ratios of the top 9 genes.
[IFNγ刺激UC-MSCにおけるIDO発現の定量解析]
 上記方法により、7,14もしくは21日間培養して得られた平面培養細胞(EXP-ADH)、または浮遊培養細胞(EXP-SUS)の凍結ストックを解凍し、細胞培養プレートに播種してIFNγ(品番300-02、メーカーPEPROTECH)を添加し、2日間培養した。その後NucleoSpin RNA(品番740955.50、メーカーMACHERY-NAGEL)を使用してtotal RNAを抽出し、PrimeScript(登録商標)RT reagent Kit(PrimeScript(登録商標)RT reagent Kit 品番RR037B、メーカーTakara)によりcDNAを合成し、SYBR(登録商標)Premix Ex Taq(商標)II(品番RR081A、メーカーTakara)を使用しPikoReal96(メーカーThermo)でリアルタイムPCRを行い、PikoReal Software2.0(メーカーThermo)で解析した。各培養日数におけるADHのIDO発現量を1とした場合の、SUSのIDO発現量を求めた(表1)。使用したプライマー配列は以下のとおりである。
[Quantitative analysis of IDO expression in IFNγ-stimulated UC-MSC]
According to the above method, a frozen stock of flat culture cells (EXP-ADH) or suspension culture cells (EXP-SUS) obtained by culturing for 7, 14 or 21 days was thawed, seeded on a cell culture plate, and IFNγ ( No. 300-02, manufacturer PEPROTECH) was added, and the cells were cultured for 2 days. Then, total RNA was extracted using NucleoSpin RNA (product number 740955.50, manufacturer MACHERY-NAGEL), and PrimeScript (registered trademark) RT reagent kit (Primescript (registered trademark) RT reagent kit RR037B, manufactured by manufacturer RR0ar). After synthesizing, real-time PCR was performed using SYBR (registered trademark) Premix Ex Taq (trademark) II (part number RR081A, manufacturer Takara) with PikoReal96 (manufacturer Thermo), and analyzed with PikoReal Software 2.0 (manufacturer Thermo). The IDO expression level of SUS was calculated when the IDO expression level of ADH in each culture period was set to 1 (Table 1). The primer sequences used are as follows.
 IDO:GGG ACA CTT TGC TAA AGG CG(F;配列番号1)及びGTC TGA TAG CTG GGG GTT GC(R;配列番号2)
 GAPDH:AGC CTC AAG ATC ATC AGC AAT G(F;配列番号3)及びATG GAC TGT GGT CAT GAG TCC TT(R;配列番号4)
IDO: GGG ACA CTT TGC TAA AGG CG (F; SEQ ID NO: 1) and GTC TGA TAG CTG GGG GTT GC (R; SEQ ID NO: 2)
GAPDH: AGC CTC AAG ATC ATC AGC AAT G (F; SEQ ID NO: 3) and ATG GAC TGT GGT CAT GAG TCC TT (R; SEQ ID NO: 4)
 いずれの培養日数の細胞においても、IFNγ刺激によるIDO発現はEXP-SUSがEXP-ADHを上回った。 EXP-SUS was higher than EXP-ADH in the expression of IDO by IFNγ stimulation in cells of any culture days.
Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001
[GVHDモデルマウスにおけるUC-MSC投与効果の評価]
 NOGマウスにヒトPBMCを移植し(5×10cells/body)、1週間後から週に1回、合計4回浮遊培養細胞(EXP-SUS)を尾静脈より投与した。なお、高用量群(EXP-SUS high)及び低用量群(EXP-SUS low)は、一回の投与でそれぞれ1×10cells/kg、2×10cells/kgの細胞を投与した。合計6週間の経過観察の結果、EXP-SUS投与群においてコントロール群と比較し、GVHDによる体重減少の抑制(図10)及びGVHDによる生存率の低下の抑制が見られた(図11)。このことから、EXP-SUSは、GVHDに対して有効であることがわかった。
[Evaluation of UC-MSC administration effect in GVHD model mouse]
Human PBMCs were transplanted into NOG mice (5×10 6 cells/body), and 1 week later, once a week, a total of 4 suspension culture cells (EXP-SUS) were administered through the tail vein. The high-dose group (EXP-SUS high) and the low-dose group (EXP-SUS low) each received 1×10 7 cells/kg and 2×10 6 cells/kg of the single administration. As a result of the follow-up observation for a total of 6 weeks, suppression of weight loss due to GVHD (FIG. 10) and suppression of decrease in survival rate due to GVHD were observed in the EXP-SUS administration group as compared with the control group (FIG. 11). From this, it was found that EXP-SUS was effective against GVHD.
[UC-MSCにおける抗炎症効果の検討]
 ヒト由来単球細胞であるTHP1細胞を3×10 cells/wellで12ウェルプレートに播種し、Phorborl 12-Myristate 13-Acetate(PMA、品番162-23591、富士フイルム和光純薬株式会社)を100nM添加して3日間培養した。上記の方法により、8日間培養して得られた平面培養細胞(EXP-ADH)、または浮遊培養細胞(EXP-SUS)の凍結ストックを解凍し、セルカルチャーインサートに40,000cells/wellで播種し、PMA刺激THP1細胞を播種したウェルに設置し、Lipopolysaccharide(LPS、品番14874-24、InvivoGen)を1μg/mL添加した。2日後にTHP1細胞からtotal RNAを抽出し、リアルタイムPCR法によりCCL2遺伝子発現量を解析した(図12)。使用したプライマー配列は以下のとおりである。
[Study of anti-inflammatory effect in UC-MSC]
Human-derived monocytic THP1 cells were seeded in a 12-well plate at 3×10 4 cells/well, and Phorbol 12-Myristate 13-Acetate (PMA, product number 162-23591, FUJIFILM Wako Pure Chemical Industries, Ltd.) at 100 nM. The cells were added and cultured for 3 days. By the above-mentioned method, a frozen stock of flat culture cells (EXP-ADH) or suspension culture cells (EXP-SUS) obtained by culturing for 8 days was thawed and seeded at 40,000 cells/well on a cell culture insert. , PMA-stimulated THP1 cells were placed in the seeded well, and 1 μg/mL of Lipopolysaccharide (LPS, product number 14874-24, InvivoGen) was added. Two days later, total RNA was extracted from THP1 cells, and the CCL2 gene expression level was analyzed by the real-time PCR method (FIG. 12). The primer sequences used are as follows.
 CCL2:AAGAAGCTGTGATCTTCAAGAC(F;配列番号5)及びCCATGGAATCCTGAACCCA(R;配列番号6) CCL2: AAGAAGCTGTGATCTTCAAGAC (F; SEQ ID NO: 5) and CCATGGAATCCTGAACCCA (R; SEQ ID NO: 6)
 LPS刺激無し(未処理群)と比較し、LPS刺激によりCCL2mRNA発現が上昇する(LPS群)が、EXP-SUSおよびEXP-ADHとの共培養によりその発現は抑制された。更に、EXP-SUSでより抑制能が大きいという結果が得られた(図12)。このことから、EXP-SUSおよびEXP-ADHは炎症に対して有効であり、特にEXP-SUSは優れた抗炎症効果を有することがわかった。 Compared to the case without LPS stimulation (untreated group), LPS stimulation increased CCL2 mRNA expression (LPS group), but its expression was suppressed by co-culture with EXP-SUS and EXP-ADH. Furthermore, the result that EXP-SUS had a larger inhibitory ability was obtained (FIG. 12). From this, it was found that EXP-SUS and EXP-ADH are effective against inflammation, and especially EXP-SUS has an excellent anti-inflammatory effect.
 本発明によると、免疫疾患治療剤及び抗炎症剤を提供することができる。 According to the present invention, a therapeutic agent for immune diseases and an anti-inflammatory agent can be provided.

Claims (7)

  1.  キヌレニン産生量、もしくはキヌレニン分泌量が高いことを特徴とする、間葉系幹細胞。 A mesenchymal stem cell characterized by high kynurenine production or high kynurenine secretion.
  2.  IFNγ処理により細胞内で誘導されるIDOにより、トリプトファンから合成されるキヌレニン産生量、もしくはキヌレニン分泌量が高いことを特徴とする、間葉系幹細胞。 A mesenchymal stem cell characterized by having a high amount of kynurenine synthesized from tryptophan or a high amount of kynurenine secreted by IDO induced intracellularly by IFNγ treatment.
  3.  JAK/STAT経路が、定常状態で活性化していることを特徴とする、間葉系幹細胞。 A mesenchymal stem cell characterized in that the JAK/STAT pathway is activated in a steady state.
  4.  臍帯組織由来である、請求項1から3のいずれか1項に記載の間葉系幹細胞。 The mesenchymal stem cell according to any one of claims 1 to 3, which is derived from umbilical cord tissue.
  5.  請求項1から4のいずれか1項に記載の間葉系幹細胞を含有する免疫疾患治療剤。 An agent for treating an immune disease, which comprises the mesenchymal stem cells according to any one of claims 1 to 4.
  6.  請求項1から4のいずれか1項に記載の間葉系幹細胞を含有する抗炎症剤。 An anti-inflammatory agent containing the mesenchymal stem cells according to any one of claims 1 to 4.
  7.  請求項1から4のいずれか1項に記載の間葉系幹細胞を含有するGVHD治療剤。 A therapeutic agent for GVHD, comprising mesenchymal stem cells according to any one of claims 1 to 4.
PCT/JP2019/046098 2018-12-14 2019-11-26 Mesenchymal stem cell, therapeutic agent for immune disease, and anti-inflammatory agent WO2020121800A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2020559070A JP7464956B2 (en) 2018-12-14 2019-11-26 Mesenchymal stem cells, immune disease treatments and anti-inflammatory agents
CN201980081889.1A CN113164770A (en) 2018-12-14 2019-11-26 Mesenchymal stem cell, therapeutic agent for immune disease, and anti-inflammatory agent

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2018234113 2018-12-14
JP2018-234113 2018-12-14

Publications (1)

Publication Number Publication Date
WO2020121800A1 true WO2020121800A1 (en) 2020-06-18

Family

ID=71076383

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2019/046098 WO2020121800A1 (en) 2018-12-14 2019-11-26 Mesenchymal stem cell, therapeutic agent for immune disease, and anti-inflammatory agent

Country Status (3)

Country Link
JP (1) JP7464956B2 (en)
CN (1) CN113164770A (en)
WO (1) WO2020121800A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112748080A (en) * 2020-12-25 2021-05-04 北京泽辉辰星生物科技有限公司 Method for detecting activity of immunoregulation stem cell IDO1 meeting product release requirements

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114209796A (en) * 2021-12-31 2022-03-22 海南康盾生物制药有限公司 Cell therapy medicine for nephropathy and preparation method and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018074758A2 (en) * 2016-10-17 2018-04-26 사회복지법인 삼성생명공익재단 Method for sorting highly effective stem cells for treating immune disorder
JP2018527403A (en) * 2015-09-15 2018-09-20 カンステム バイオテック カンパニー リミテッド Composition for preventing or treating inflammatory diseases comprising stem cells overexpressing SOD3 as an active ingredient
JP2018536403A (en) * 2015-11-09 2018-12-13 サムソン ライフ パブリック ウェルフェア ファウンデーション Stem cells with improved immunosuppressive ability with suppressed SOCS and use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2018527403A (en) * 2015-09-15 2018-09-20 カンステム バイオテック カンパニー リミテッド Composition for preventing or treating inflammatory diseases comprising stem cells overexpressing SOD3 as an active ingredient
JP2018536403A (en) * 2015-11-09 2018-12-13 サムソン ライフ パブリック ウェルフェア ファウンデーション Stem cells with improved immunosuppressive ability with suppressed SOCS and use thereof
WO2018074758A2 (en) * 2016-10-17 2018-04-26 사회복지법인 삼성생명공익재단 Method for sorting highly effective stem cells for treating immune disorder

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ATSUKO TAKAHASHI, ET AL: "Establishment of immunosuppressive effect evaluation method for umbilical cord-derived mesenchymal cell products", JAPANESE JOURNAL OF TRANSFUSION AND CELL THERAPY, vol. 65, no. 2, 23 April 2019 (2019-04-23), pages 386, XP055720209 *
HADIZADEHMOGHADDAM SEPEHR; ADVE RAVIRAJ S.: "Simultaneous execution of multiple radar tasks using OFDMA", 2017 IEEE RADAR CONFERENCE (RADARCONF), IEEE, 8 May 2017 (2017-05-08), pages 0622 - 0626, XP033104091, DOI: 10.1109/RADAR.2017.7944278 *
TOJO, ARINOBU: "Development of novel immunotherapy and regenerative medicine using formulations of umbilical cord blood and umbilical cord- derived mesenchymal stem cells", STUDY ON IMMUNOREGULATORY EFFECTS OF UMBILICAL CORD-DERIVED MESENCHYMAL STEM CELLS, REPORT ON THE RESULTS OF CONSIGNMENT WORK 2014, vol. 26, 2015, pages 32 - 39 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112748080A (en) * 2020-12-25 2021-05-04 北京泽辉辰星生物科技有限公司 Method for detecting activity of immunoregulation stem cell IDO1 meeting product release requirements

Also Published As

Publication number Publication date
CN113164770A (en) 2021-07-23
JP7464956B2 (en) 2024-04-10
JPWO2020121800A1 (en) 2021-11-04

Similar Documents

Publication Publication Date Title
US11723930B2 (en) Treatment of inflammatory diseases using placental stem cells
JP2022046511A (en) Immunomodulation using placental stem cell
JP7082372B2 (en) Pulmonary fibrosis treatment agent, PTPRR expression promoter and pulmonary fibrosis treatment kit
WO2018159432A1 (en) Mesenchymal stem cells and pharmaceutical composition
WO2020121800A1 (en) Mesenchymal stem cell, therapeutic agent for immune disease, and anti-inflammatory agent
WO2018116732A1 (en) Therapeutic agent for non-alcoholic steatohepatitis, and kit for treatment of non-alcoholic steatohepatitis
JP2019156739A (en) Mesenchymal stem cell, disease therapeutic agent, and microparticle
JP2023024665A (en) Mesenchymal stem cells and therapeutic agent for liver disease
JP2023103416A (en) Therapeutic agent for dilated cardiomyopathy
WO2018123627A1 (en) Cell pharmaceutical composition, disease treatment kit, and cell suspension solution
AU2014203165B2 (en) Treatment of inflammatory diseases using placental stem cells
JPWO2018123628A1 (en) Cell pharmaceutical composition, disease treatment kit and cell suspension solution
AU2017248436A1 (en) Immunomodulation using placental stem cells
AU2016256718A1 (en) Treatment of inflammatory diseases using placental 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: 19894772

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020559070

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19894772

Country of ref document: EP

Kind code of ref document: A1