WO2008018190A1 - Fat-derived neural crest cells - Google Patents

Fat-derived neural crest cells Download PDF

Info

Publication number
WO2008018190A1
WO2008018190A1 PCT/JP2007/051643 JP2007051643W WO2008018190A1 WO 2008018190 A1 WO2008018190 A1 WO 2008018190A1 JP 2007051643 W JP2007051643 W JP 2007051643W WO 2008018190 A1 WO2008018190 A1 WO 2008018190A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
neural crest
derived
stem cells
Prior art date
Application number
PCT/JP2007/051643
Other languages
French (fr)
Japanese (ja)
Inventor
Kotaro Yoshimura
Takashi Nagase
Daisuke Matsumoto
Tomokuni Shigeura
Original Assignee
Biomaster, Inc.
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 Biomaster, Inc. filed Critical Biomaster, Inc.
Publication of WO2008018190A1 publication Critical patent/WO2008018190A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1346Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells
    • C12N2506/1384Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells from adipose-derived stem cells [ADSC], from adipose stromal stem cells

Definitions

  • the present invention relates to the field of cell sorting. More particularly, the present invention relates to a fat-derived or fat-derived stem cell-derived neural crest cell, a method for producing the same, and a transplantation therapy using the same.
  • tissue stem cells that were previously thought to be absent have been discovered and identified from various tissues. Thus, disease treatment by regenerative medicine is attracting attention.
  • a fertilized egg is divided into three germ layers of endoderm, mesoderm and ectoderm after gastrulation, and cells derived from ectoderm mainly exist in the brain, and include neural stem cells and the like.
  • Cells derived from mesoderm are mainly present in the bone marrow, and include vascular stem cells, hematopoietic stem cells, mesenchymal stem cells, and the like.
  • Endoderm-derived cells are mainly present in the pharynx, lungs, liver, spleen, intestine, etc., and include spleen stem cells, liver stem cells, and the like.
  • Adipose-derived stem cells are originally subtypes of mesenchymal stem cells (MSCs) that have also isolated liposuction aspirate powers that can be separated into mesenchymal tissues such as bone, cartilage and adipose tissue.
  • MSCs mesenchymal stem cells
  • Non-Patent Document 1 At present, adipose-derived stem cells are considered as one of the most promising adult stem cells for regenerative medicine (Non-Patent Documents 2 to 3). This is because fat-derived stromal cells can be safely recovered by liposuction and good yields can be expected. Ruka.
  • Non-patent Documents 4 and 5 derivatives of the nervous system
  • mesoderm tissue eg, dermis and heart
  • Non-Patent Documents 6 to 8 stem cells with neural characteristics have been collected by the neurosphere method.
  • the neurosphere method was developed as a method for culturing isolated spheres of neural stem cells derived from embryos and human brain.
  • Non-patent Document 9 It has been reported that neural stem cells are also present in the central nervous system of adults, which has long been thought to regenerate the central nervous system of adult mammals.
  • the number is small and it requires a great deal of labor to isolate, and it is difficult to actually use it in regenerative medicine such as nervous system diseases. Therefore, obtaining pluripotent cells of the nervous system from the nervous system is not practical. Therefore, studies have been conducted on the division of the nervous system using fat.
  • Neural crest cells also called neural crest cells, have the ability to migrate and have multipotency into differentiated cells of the nervous system (eg dorsal root ganglion cells, autonomic ganglion cells, adrenal medulla chromaffinity) A cell, a Schwann cell, and a cell having a differentiation potential into a coat pigment cell). Therefore, neural crest cells can be said to be broad stem cells rather than differentiated cells. Nerve crest cells are identified as a group of cells (fourth germ layer) that originates from the limbal limbus (nerve crest) in the early fetal period and migrates widely in the embryo and divides into a very wide and specific repertoire. It was done. At present Neural crest cells have been used to treat facial malformations.
  • the eurosphere produced by the group of Kang, KS, etc. can be said to be an aggregate of differentiated cells, and this -eurosphere has multipotency to differentiate into cells of the nervous system. No cells are present, and even neural crest cells are expected to be completely absent from this -Eurosphere. Therefore, when adipose-derived stem cells are used, differentiation is remarkably induced and it is considered that induction into pluripotent cells of the nervous system, such as neural crest cells, does not occur.
  • Patent Documents 1 to 6 describe that adipose-derived stem cells appear to have the ability to be divided into nerves.
  • these documents do not substantially describe the induction of adipose-derived stem cells into pluripotent cells of the nervous system such as neural stem cells, neural progenitor cells, and neurospheres, and have the ability to differentiate directly into nerves. Whether or not it is being studied. Therefore, these documents do not describe anything that can be induced from fat to neural pluripotent cells such as neural stem cells, neural crest cells, etc. In other words, since it has been attempted to differentiate into differentiated cells), it is considered that it cannot be induced into pluripotent cells of the nervous system in this field.
  • adipose-derived stem cells are abundant in supply, if adipose-derived, in particular, adipose-derived stem cells can be used to produce pluripotent cells of the nervous system, It can be said that it will bring about a revolutionary change in the treatment of diseases and disorders of the trans-system, and the demand for such methods will increase.
  • Patent Document 1 Reprinted WO2003Z008592
  • Patent Document 2 Japanese Translation of Special Publication 2005-502352
  • Patent Document 3 Japanese Translation of Special Publication 2002-537849
  • Patent Document 4 US Patent Application Publication No. 2001Z0033834
  • Patent Document 5 Special Table 2003-523767
  • Patent Document 6 Special Table 2005-502712
  • Non-patent literature l Zuk PA, Zhu M, Mizuno H, et al. Tissue Eng. 2001; 7: 2 11-228
  • Non-Patent Document 2 Zuk PA, Zhu M, Ashjian P, et al. Mol. Biol. Cell 2002; 1 3: 4279-4295
  • Non-Patent Document 3 Yoshimura K, Shigeura T, Matsumoto D, et al. J. Cell. Phisiol. 2006; 208: 64-76
  • Non-Patent Document 4 Ashjian PH, Elbarbary AS, Edmonds B, et al. Plast. Recon nstr. Surg. 2003; 111: 1922-1931
  • Non-Patent Document 5 Kokai LE, Rubin JP, Marra KG. Plast. Reconstr. Surg. 20 05; 116: 1453-1460
  • Non-Patent Document 6 Toma JG, Akhavan M, Fernandes KJ, et al. Nat. Cell. Bio 1.2001; 3: 778-784
  • Non-Patent Document 7 Fernandes KJ, McKenzie IA, Mill P, et al. Nat. Cell. Biol. 2004; 6: 1082-1093
  • Non-Patent Document 8 Tomita Y, Matsumura K, Wakamatsu Y, et al. J. Cell. Biol. 2005; 170: 1135-1146
  • Non-Patent Document 9 Enoyuki Okano, Experimental Medicine, No. 20, No. 9, 2002; 1276-1279
  • Non-Patent Document 10 Kang SK, Putnam LA, Ylostalo J, et al. J. Cell. Sci. 200
  • Non-patent literature ll Kang SK, Putnam L, Dufour J, et al. Stem Cells 2004; 22: 1356-1372
  • Non-Patent Document 12 Bunnell BA, Ylostalo J, Kang SK.Biochem.Biophys.Res.Commun.2006; May 12; 343 (3): 762-71
  • Non-Patent Document 13 Safford KM, Hicok KC, Safford SD, et al. Biochem. Biophys. Res. Commun. 2002; Jun 7; 294 (2): 371-9
  • Non-Patent Document 14 Tholpady SS, Katz AJ, Ogle RC. Anat. Rec. A Discov. Mol. Cell. Evol. Biol. 2003; May; 272 (1): 398-402
  • Non-Patent Document 15 Kang SK, Lee DH, Bae YC, et al. Exp. Neurol. 2003; 0 ct; 183 (2): 355-66
  • Non-patent document 16 Hiroshi Mizuno J. Nippon Med. Sch. 2003; Oct; 70 (5): 428-31
  • Non-patent document 17 Saf ford KM, Safford SD, Gimble JM, et al. Exp. Neurol. 2004; Jun; 187 (2): 319-28
  • Non-Patent Document 18 Yang LY, Liu XM, Sun B, et al. Chin. Med. J. (Engl) .2 004; Mar; 117 (3): 425—9
  • Non-Patent Document 19 Safford KM, Rice HE. Curr. Drug. Targets. 2005; Feb 6 (1): 57-62
  • Non-Patent Document 20 Fujimura J, Ogawa R, Mizuno H, et al. Biochem. Biophys. Res. Commun. 2005; Jul 22; 333 (1): 116-21
  • Non-Patent Document 21 Strem BM, Hicok KC, Zhu M, et al. Keio J. Med. 2005
  • Non-Patent Document 22 Parker AM, Katz AJ. Expert. Opin. Biol. Ther. 2006 Jun; 6 (6): 567-78
  • an object of the present invention is to meet such a demand. Furthermore, an object of the present invention is to provide a surgical method capable of obtaining a desired treatment effect and a material or a medicine used therefor. An object of the present invention is to induce neural pluripotent cells using abundant fat as a raw material. More specifically, an object is to induce neural crest cells from fat.
  • the present invention provides the following.
  • Fat-derived neural crest cells Fat-derived neural crest cells.
  • the neural crest cell according to item 1 which is derived from an adipose-derived stem cell.
  • the neural crest cell according to item 1 wherein the neural crest cell expresses at least one neural stem cell marker.
  • neural stem cell marker is selected from the group consisting of Nestin, Musashi-1, CD133, notch1, Hesl, Mashl, Neurogenin, Pax6, CD15 and PDGFR.
  • Item 7 The neural crest cell according to item 6, wherein the neural stem cell marker includes Nestin and Musashi-1.
  • the neural crest cell according to item 1 wherein the neural crest cell expresses at least one neural crest cell marker.
  • the neural crest cell marker is selected from the group consisting of CRABP1, AP2, Slug, SoxlO, Snail, Twist, Pax3, Pax7, HNK1, p75NTR, TRP2, Wntl, PO, and tPA.
  • the neural crest cell according to 1.
  • the neural crest cell according to item 1 wherein the neural crest cell expresses Nestin and Musashi-1, and the expression level of Leptin is lower than the expression level of adipose-derived progenitor cells.
  • Item 2 The neural crest cell according to Item 1, wherein the fat is derived from a mammal.
  • Item 13 The neural crest cell according to Item 12, wherein the fat is derived from a human.
  • a method for preparing neural crest cells comprising the following steps:
  • Item 15 The method according to Item 14, wherein the anterior stem cell is an adipose-derived stem or stromal cell.
  • Item 15 The method according to Item 14, wherein the step of obtaining the stem cell comprises collagenase treatment and centrifugation treatment.
  • Item 15 The method according to Item 14, wherein the stem cell is selected from the group consisting of PLA cell and LAF cell force.
  • step B) the adipose-derived progenitor cells are seeded at a density of about 1 ⁇ 10 4 cells Zml to about 1 ⁇ 10 6 cells Zml.
  • Item 19 The method according to Item 18, wherein in the step B), the adipose-derived stem cells are seeded at a density of about 1 ⁇ 10 5 cells Zml.
  • Item 15 The method according to Item 14, wherein in the step B), the adipose-derived stem cells are cultured in a medium containing at least epidermal growth factor (EGF).
  • EGF epidermal growth factor
  • Item 15 The method according to Item 14, wherein in step B), the adipose-derived stem cells are cultured in a medium containing epidermal growth factor, basic fibroblast growth factor, nerve induction factor, penicillin, and streptomycin as auxiliary components. .
  • the nerve-inducing factor is piotin, L-carcin, corticosterone, ethanolamine, D (+)-galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retulacetate, selenium, 28.
  • the item according to item 21, comprising triodothyronine (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin (ushi), catalase, insulin, superoxide dismutase, and transferrin.
  • Item 15 The method according to Item 14, wherein in the step B), a medium used for culturing the stem cells contains glutamic acid and aspartic acid.
  • the medium used for culturing the stem cells is an eagle basic medium (BME), a minimum essential medium (MEM), a Dulbecco's modified Eagle medium (DMEM), a HAMF12 medium, or a mixture thereof.
  • BME eagle basic medium
  • MEM minimum essential medium
  • DMEM Dulbecco's modified Eagle medium
  • HAMF12 a HAMF12 medium
  • Item 15 The method according to Item 14, which is a medium.
  • Item 15 The method according to Item 14, wherein the medium used for culturing the stem cells in Step B) is Dulbecco's Modified Eagle Medium (DMEM) ZF12 (1: 1).
  • DMEM Dulbecco's Modified Eagle Medium
  • step B) includes replacing the medium with a new medium on days 2 to 6.
  • step B) includes replacing the medium with a new medium on the 4th to 5th days.
  • step B) includes subculture on the 6th to the 10th day.
  • Item 15 The method according to Item 14, wherein the fat is derived from a mammal.
  • the adipose-derived stem cells are human-derived
  • the adipose-derived stem cells are seeded at a density of about 1 ⁇ 10 5 cells Zml;
  • the adipose-derived stem cells are cultured in a medium containing epidermal growth factor, basic fibroblast growth factor, nerve induction factor, penicillin and streptomycin as auxiliary components, wherein the nerve induction factor is piotin, L— Carcin, corticosterone, ethanolamine, D (+) galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retino-acetate, selenium, tolydothyrone (T3), DL—a Tokov Contains erol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin, and
  • Item 15 The method according to Item 14, wherein the medium is Dulbecco's Modified Eagle Medium (DMEM) ZF12 (1: 1).
  • DMEM Dulbecco's Modified Eagle Medium
  • Item 15 The method according to Item 14, further comprising the step of confirming whether the stem cell expresses a neural stem cell marker.
  • the neural stem cell marker is selected from the group consisting of Nestin, Musashi-1, CD133, notch 1, Hesl, Mashl, Neurogenin, Pax6, CD15 and PDGFR The method according to 32.
  • Item 15 The method according to Item 14, further comprising the step of confirming whether the stem cells express a neural crest cell marker.
  • Item 15 The method according to Item 14, wherein the cell expresses at least one neural crest cell marker.
  • the neural crest cell marker is selected from the group consisting of CRABP1, AP2, Slug, SoxlO, Snail, Twist, Pax 3, Pax7, HNK1, p75NTR, TRP2, Wntl, PO, and tPA force. .
  • Item 15 The method according to Item 14, further comprising the step of confirming the level of expression of the adipocyte marker by the stem cell.
  • Item 15 The method according to Item 14, wherein the neural crest cell becomes a fat-derived neural crest cell precursor containing neural crest cells and adipose-derived stem cells by the step B).
  • the method according to item 14 further comprising the step of confirming whether the stem cell expresses a neural stem cell marker and a neural crest cell marker, and confirming the level of expression of the adipocyte marker by the stem cell.
  • Item 44 The method according to Item 41, wherein the neural stem cell marker is Nestin and Musashi-1, and the fat cell marker is Leptin.
  • a composition for cell transplantation for the treatment of a nervous system disease, disorder or condition comprising the cell according to item 1.
  • composition according to item 45 wherein the nervous system disease, disorder or condition is a disease, disorder or condition caused by a defect in differentiated cells of the nervous system.
  • composition according to item 45 wherein the fat is derived from an individual having an allogeneic relationship with an individual subject to the nervous system disease, disorder or condition.
  • composition according to item 45 wherein the fat is derived from an individual having a heterogeneous relationship with the subject of the nervous system disease, disorder or condition.
  • composition according to item 45 wherein the fat is derived from an individual having an allogeneic relationship with an individual subject to the nervous system disease, disorder or condition.
  • a method for cell transplantation for the treatment of a nervous system disease, disorder or condition comprising the step of administering the cell according to item 1.
  • Said nervous system disease, disorder or condition is a disease caused by a loss of differentiated cells of the nervous system, 51.
  • the method of item 50, wherein the method is a disorder or condition.
  • the nervous system disease, disorder or condition is a disease, disorder or condition caused by a defect in differentiated cells of the nervous system.
  • An adipose-derived neural crest cell mixture comprising neural crest cells and adipose-derived stem cells.
  • regenerative treatment can be performed using a neural crest cell derived from fat and a neural crest cell derived from fat precursor cell.
  • a neural crest cell can be further prepared by recovering a fat progenitor cell and subjecting the progenitor cell to conditions for isolating the neural crest cell.
  • the present invention also facilitates treatment of nervous system diseases, disorders or conditions by transplanting neural crest cells. These treatments provide a simple and efficient treatment method in regenerative medicine because few side effects are expected and their sources are abundant.
  • neurospheres containing neural crest cells could be provided at a remarkably high rate. Since fat can be used as a raw material, an effect that a large amount of neural crest cell mixture can be provided is also provided.
  • Fig. 1 shows culture in a neurosphere culture medium, Day 5 (Day 5: A), Day 6 (Day 6 (Day 2).
  • Figure 2 shows neural stem cell markers Nestin (A) and Musashi— 1 (B), and The results of quantitative real-time RT-PCR analysis of gene expression of the adipogenic marker Leptin (C) are shown (control: undifferentiated adipose-derived stem cells, sphere: Nyuro sphere). Atsey was conducted in triplicate. Standard error is indicated by an error bar.
  • FIG. 3 shows neural crest-like migration of GFP-transformed adipose-derived stem cells transplanted into mouse embryos cultured in vivo.
  • A A photograph of a mouse embryo that was cultured for 40 hours on day 8 embryos.
  • B, C Fluorescent photographs of embryos. GFP positive-Eurosphere cells were arranged in a row (arrow). This suggests that neurosphere cells are migrating along the second arch. The bar is 500 ⁇ m.
  • cell used herein is defined as the broadest meaning used in the field, and is a structural unit of a tissue of a multicellular organism and is enclosed in a membrane structure that isolates the living body from the outside world. Rarely, an organism with self-regenerative ability and genetic information and its expression mechanism. Any cell can be targeted in the method of the present invention.
  • the number of “cells” used in the present invention can be counted through an optical microscope. When counting through an optical microscope, count by counting the number of nuclei. The tissue is sliced into tissue slices and stained with hematoxylin-eosin (HE) to separate extracellular matrix (eg, elastin or collagen) and cell-derived nuclei.
  • HE hematoxylin-eosin
  • the cells used in the present specification may be naturally occurring cells or artificially modified cells (eg, fusion cells, genetically modified cells, etc.).
  • Examples of cell sources include: It can be a single cell culture, or a cell mixture such as cells from a normally grown transgenic animal embryo, blood, or body tissue (eg, adipose tissue), or a normally grown cell line However, it is not limited to them. Further, such a supply source can be used as a cell as it is.
  • the fat cells (fat eels or adipocytes) used in the present invention and their counterparts may be any organism (for example, larvae, jellyfishes) as long as the following organisms have adipocytes or their counterparts.
  • Cartilaginous fish, teleosts, amphibians, reptiles, birds, mammals, etc. Preferably, mammals (e.g. single pores, marsupials, rodents, wings, wings, carnivores, carnivores, long noses, odd-hoofed animals, even-hoofed animals, rodents, It may be derived from scales, sea cattle, cetaceans, primates, rodents, maggots, etc.). In one embodiment, cells derived from primates (eg, chimpanzee, dihonosa, human), particularly cells derived from humans, are used, but are not limited thereto.
  • stem cell refers to a precursor cell of a differentiated cell (pr ecursor or progenitor), which is monopotency, multipotency. Refers to a cell having totipotency.
  • stem cell can be used interchangeably with “progenitor cell”.
  • Stem cells can be differentiated in response to specific stimuli. Stem cells are usually able to regenerate the tissue when it is damaged.
  • Stem cells as used herein can be, but are not limited to, embryonic stem (ES) cells or tissue stem cells (including tissue stem cells, tissue-specific stem cells or somatic stem cells) or other progenitor cells. Not.
  • an artificially produced cell can also be a stem cell as long as it has the above-mentioned ability.
  • Embryonic stem cells are pluripotent stem cells derived from early embryos. Embryonic stem cells were first established in 1981 and have been applied since 1989 to the production of knockout mice. In 1998, human embryonic stem cells were established and are being used in regenerative medicine. Unlike embryonic stem cells, tissue stem cells are cells that have a relatively limited degree of separation, exist in the tissue, and have an undifferentiated intracellular structure. Tissue stem cells are poor in organelles with a high nuclear Z cytoplasm ratio. Tissue stem cells are generally individuals with pluripotency and slow cell cycle Maintains proliferative capacity over the life of the body. As used herein, stem cells can preferably be embryonic stem cells, although tissue stem cells can also be used depending on the circumstances.
  • stem cell may refer to a tissue containing at least a certain amount of stem cells or progenitor cells. Therefore, the stem cells can be stem cells (eg, adipose-derived stem cells used in the following examples) that have been treated with collagenase and collected adipose tissue strength, but are not limited thereto.
  • stem cells eg, adipose-derived stem cells used in the following examples
  • tissue stem cells When classified according to the site from which the cells are derived, tissue stem cells are classified into, for example, the skin system, digestive system, myeloid system, nervous system and the like.
  • Skin tissue stem cells include epidermal stem cells and hair follicle stem cells.
  • tissue stem cells of the extinct system include knee (common) stem cells and liver stem cells.
  • myeloid tissue stem cells include hematopoietic stem cells and mesenchymal stem cells.
  • Neural tissue stem cells include neural stem cells and retinal stem cells.
  • mesenchymal stem cell refers to a stem cell found in the mesenchyme.
  • the term “mesenchymal stem cell” may be abbreviated herein as “MSC”.
  • mesenchyme is defined by a population of free cells with stellate or irregular projections that fill the gaps between epithelial tissues, which are observed at various stages of development of multicellular animals, and the associated cytoplasm. This refers to the organization that is formed.
  • Mesenchymal stem cells have the ability to proliferate and differentiate into bone cells, chondrocytes, muscle cells, stromal cells, tendon cells, and adipocytes.
  • Mesenchymal stem cells are used to cultivate or proliferate bone marrow cells collected from patients, have chondrocytes! /, To differentiate into osteoblasts, and, for example, bones such as alveolar bone, arthropathy, It is used as a reconstruction material for cartilage or joints, and its demand is great. In addition, since mesenchymal stem cells can be separated into blood cells and lymphoid cells, the demand for them is increasing.
  • the term "adipose-derived stem cell” refers to a stem cell obtained by liposuction and also other progenitor cells (eg, peripheral blood or vascular stromal cells (vascular- stromal cell) (stem cell derived from preadipocyte). Adipose-derived stem cells are derived from adipose tissue or from any multipotent precursor cell) or monopotent precursor cell population obtained by a liposuction procedure. means.
  • Adipose-derived vascular stromal cells (adipose—derived interstitial cells, adipose—derived stromal cells), adipose-derived stem cells, adipose-derived progenitor cells, adipose stem cells , Endothelial progenitor cells, hematopoietic stem cells, and the like.
  • stem cell separation methods are known, and are described, for example, in Non-Patent Document 1, Patent Documents 1 and 2. The matters described in these documents are hereby incorporated by reference in particular in the present specification.
  • adipose-derived stem cells refers to all adipose tissue-derived stem cells including adipose tissue-derived stem cells obtained by these known separation methods.
  • progenitor cell includes unipotent undifferentiated cells as well as pluripotent undifferentiated cells.
  • stem cell includes progenitor cells.
  • Aspirated material collected by liposuction is separated into two layers in a suction bottle.
  • the upper layer of aspirate is composed of floating fat (lipoaspimte) and the lower layer is composed of liquid (liquid-aspirate or liposcution-aspirate fluid) forces.
  • PVA processed lipo aspirate cell
  • LAF cells liquid—aspirate
  • eel is a liposcution—aspirate fluid cell)), which is a progenitor cell derived from the liquid part of the aspirate.
  • Adipose-derived stem cells include PLA cells and LAF cells.
  • the term "somatic cell” refers to all cells that are cells other than germ cells, such as eggs and sperm, and that do not deliver the DNA to the next generation. Somatic cells usually have power or loss of limited pluripotency. Somatic cells as used herein may be naturally occurring or genetically modified as long as the intended treatment can be achieved.
  • neural stem cell refers to a cell that can differentiate into the nervous system and has both self-renewal ability and pluripotency. Neural stem cells It can be identified by confirming cell markers, self-renewal ability, and pluripotency of vesicles.
  • the "self-renewal ability" of a neural stem cell refers to the ability to repeat division and generate the same cell population as self).
  • the “self-renewal ability” of a neural stem cell can be confirmed by, for example, the neurosphere method.
  • the neurosphere method is the method originally reported by Reynolds et al., J Neurosci 1992; 12: 4565-4574 and Science 1992; 255: 17 07-1710.
  • the neurosphere method is one of the most frequently used methods for isolating neural stem cells from the embryonic or adult central nervous system.
  • neural stem cells are cultured in a floating state in the presence of epidermal growth factor (E GF) and basic fibroblast growth factor (basic-FGF, b-FGF) as growth factors.
  • E GF epidermal growth factor
  • basic-FGF basic fibroblast growth factor
  • Single cell force-forming the eurosphere, dissociating the formed eurosphere, seeding the dissociated cell in a new medium, and the ability of the dissociated cell to form the eurosphere again
  • a method including a step of confirming whether or not. In this method, a dissociated cell forms a neurosphere again when it forms a eurosphere.
  • pluripotency refers to the nervous system of nerve cells and glial cells (astrocytes, oligodendrocytes, etc.). The ability to divide into multiple cells.
  • the pluripotency of neural stem cells is, for example, that when neurospheres are placed under the condition of dividing them into cells of the nervous system, they enter the nervous system of neurons and glial cells (astrocytes, oligodendrocytes, etc.). If it is determined that the cell is pluripotent.
  • pluripotency refers to having the ability to differentiate into cells of multiple nervous systems, but is not limited thereto.
  • Examples of conditions for separating cells of the nervous system include, for example, a medium for separating the nervous system (for example, DMEMZF 12:49 ml in a nerve induction medium (50 ml), a nerve inducer (for example, B27 (GIBCO)) : Lml, which may contain antibiotics) Powers including, but not limited to, 1 week of culture (culture with half the medium every 2 days).
  • a glial induction medium 50 ml of medium, DMEMZF12: 45 ml, FBS: 5 ml, may contain antibiotics
  • the neurotrophic factor that can be used herein may be any as long as it has an action of assisting or promoting nerve induction.
  • the neuroinductive factor is piotin, L-carthine, corticosterone, ethanolamine, D (+)-galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retort acetate , Selenium, tolyothyrone (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin.
  • Examples of neural pluripotent cells include neural stem cells, neural crest cells, neural progenitor cells, and the like.
  • Neural stem cells can also be identified using only neural stem cell markers.
  • a "neural stem cell marker” refers to a neural stem cell that assists in identifying a neural stem cell by its localization or expression. .
  • its localization or expression for example, Nestin (NM006617 (t)), Musashi-1 (NM002442 (human)), CD133 (NM0006017 (human)), notchl (NM017617 (human))) Hes l ( NM005524 (human)) ⁇ Mashl (NM004316 (human)) ⁇ Neurogenin (N M006161 (human)), Pax6 (NM001604 (human)), CD15 (NM002033 (human)), PDG FR (NM002609 (human))
  • neural stem cell markers include, for example, Nestin (N M006617 (Hin)), Musashi-1 (NM002442 (Hin))) , CD133 (NM0006017 (human)), notch 1 (NM017617 (human)), Hesl (NM
  • neural stem cell markers can be performed using methods known in the art (eg, RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.). Here, whether it is expressed or not can be appropriately selected by RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.
  • “neural crest cell (NCC)” has at least one characteristic selected from the group consisting of a unique migratory ability and the expression of a cell marker of at least one neural crest cell. Cell.
  • neural crest cells are identified by confirming characteristics of both unique migration ability and expression of cell markers of at least one neural crest cell.
  • CRABP1 NM004378 (human)
  • a P2 NM002097 (chicken)
  • Slug NM003068 (chicken)
  • SoxlO NM006941 (chicken)
  • Snail NM005985)
  • Twist NM000474 (chicken)
  • Pax3 NM000438 (chicken)
  • Pax7 NM002584 (human)
  • HNKl NM004854 (human)
  • p75NTR NM0025 07 (human)
  • TRP2 NM006267 (human)
  • Wntl NM005430 (human)
  • PO NM002 723 (human)
  • tPA NM000930 (human)
  • neural stem cell markers are Nestin (NM006617 (Human)), Musashi-1 (NM002442 (Human)), CD133 (NM0006017 (Human)), Notch 1 (NM017617 (Human)), Hesl (NM005524 (Human)) ), Mashl (NM00 4316 (chicken)), Neurogenin (NM006161 (chicken)), Pax6 (NM001604 (chicken)), CD 15 (NM002033 (human)), PDGFR (NM002609 (human)), etc.
  • neural stem cell markers that can be used include Nestin and Musashi-1.
  • Neural crest cells originate from the edge of the neural tube (nerve crest) in the early fetal period and migrate widely in the embryo, and are divided into an extremely wide and specific repertoire (fourth germ layer). ) was identified. Neural crest cells can differentiate into, for example, dorsal root ganglion cells, autonomic ganglion cells, adrenal medulla chromaffin cells, Schwann cells, and outer pigment cells. Neural crest cells are also called neural crest cells. Neural crest cells have self-renewal and pluripotency and are very similar to neural stem cells.
  • the neural crest cell of the present invention may have a characteristic that expression of an adipocyte marker is lower than that of a fat-derived stem cell.
  • the adipocyte marker used may be any as long as it can determine adipocytes, but is preferably not limited to the force with which Leptin (NM000230 (human)) can be used. Accordingly, it is preferable that Leptin expression is lower than that of adipose-derived stem cells.
  • the neural crest cells of the present invention can be characterized by expressing Nestin and Musashi-1 and expressing Leptin, which is an adipocyte marker, lower than the expression level of adipose-derived stem cells.
  • a "neural crest cell marker” is a neural crest cell that has V, and its localization or expression assists in identifying the neural crest cell.
  • its localization or expression eg, CRABP1 (NM004378 (human)), AP2 (N M002097 (human)), Slug (NM003068 (human)), SoxlO (NM006941 (human)), Snail (NM005985 (Human)), Twist (NM000474 (Human)), Pax3 (NM000438 (Human)), Pa x7 (NM002584 (Human)), HNKl (NM004854 (Human)), p75NTR (NM002507 (Human)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002723 (human);), tPA (NM000930 (human)), which can be identified as neural crest cells.
  • CRABP1 NM004378 (human)
  • cell markers for crest cells include CRABP1 (NM004 378 (Hin)), AP2 (NM002097 (Hin)), Slug (NM003068 (Hin)), SoxlO (NM0 06941 (Hin)), Snail ( NM005985 (chicken)), Twist (NM000474 (chicken)), Pax3 (N M000438 (chicken)), Pax7 (NM002584 (chicken)), HNKl (NM004854 (chicken)), p75 NTR (NM002507 (human) )), TR P2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002723 (human)), tPA (NM000930 (human)) and the like can be mentioned, but not limited thereto.
  • CRABP1 NM004 378 (Hin)
  • AP2 NM002097 (Hin)
  • Slug NM003068 (Hin)
  • Determination by these neural crest cell marker, neural stem cell marker or adipocyte marker is a method known in the art (for example, RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.) Can be used. Here, whether it is expressed or not can be appropriately selected by RT-PCR, Northern blot, Western blot, immunostaining, FACS and the like.
  • Leptin NM000230 Han
  • embryo or “embryonic cell” refers to the state of an early era of development in a multicellular organism.
  • the “head region” of the embryo refers to the region that migrates to the hindbrain, in particular, the second arch, that is, around Lombomea 3 and 4.
  • the term “divided cell” refers to a cell with a special function and morphology (eg, muscle cell, nerve cell, etc.), and unlike stem cells, There is little or no performance.
  • Differentiated cells include, for example, epidermal cells, spleen Stromal cells, knee duct cells, hepatocytes, blood cells, cardiomyocytes, skeletal muscle cells, osteoblasts, skeletal myoblasts, neurons, vascular endothelial cells, pigment cells, smooth muscle cells, fat cells, bone cells, soft Examples include bone cells.
  • sperm cells may be in the form of a population or tissue! /.
  • the stem cell origin can be classified into ectoderm, mesoderm and endoderm.
  • Stem cells derived from ectoderm are mainly present in the brain and include neural stem cells.
  • Stem cells derived from mesoderm are mainly present in bone marrow, and include vascular stem cells and their differentiated cells, hematopoietic stem cells and their differentiated cells, and mesenchymal stem cells and their differentiation cells.
  • Endoderm-derived stem cells exist mainly in organs, and include hepatic stem cells and their differentiation cells, spleen stem cells and their differentiation cells.
  • somatic cells may be derived from any germ layer.
  • mesenchymal cells can be used as the somatic cells.
  • adipocyte refers to interstitial tissues, or forms adipose tissue as a population along a capillary run as one of the loose connective tissues.
  • a cell containing a large amount of fat include yellow fat cells and brown fat cells, and any of these can be used equivalently in this specification.
  • Intracellular fat can be easily detected by Sudan I or osmium tetroxide.
  • the term "desired site” refers to any site in a subject for which treatment is desired. In the present invention, it is understood that such a desired site can be selected from any organ or tissue in the subject.
  • tissue refers to a population of cells having substantially the same function and Z or morphology in a multicellular organism. “Organization” is usually
  • stem cell of the present invention is used.
  • tissue When regenerating a tissue by V, cell populations having two or more different origins can constitute one tissue.
  • tissue constitutes part of an organ. Animal tissues are classified into epithelial tissues, connective tissues, muscle tissues, and nerve tissues based on morphological, functional, or developmental basis. In plant tissue, meristem and permanent tissue depend on the stage of development of the cells that make up the tissue. Various classifications are made, such as dividing into single tissue and composite tissue according to the type of constituent cells. As used herein, any tissue can be intended as a subject to be treated.
  • an organ when an organ is a target, such an organ may be any organ, and the tissue or cell targeted by the present invention may be derived from any organ or organ of an organism.
  • the term “organ” refers to a structure in which a function of an individual organism is localized and operates in a specific part of the individual, and that part is morphologically independent. .
  • an organ In multicellular organisms (eg animals, plants), an organ consists of several tissues with a specific spatial arrangement, and each tissue also has a number of cellular forces. Such organs include those related to the vascular system.
  • organs targeted by the present invention include skin, blood vessels, cornea, kidney, heart, liver, umbilical cord, intestine, nerve, lung, placenta, spleen, brain, peripheral limbs, retina, and the like. Is not limited to them.
  • any organ may be a target, but preferably a mesenchymal tissue (for example, fat, bone, ligament, etc.) may be a target. It is not limited to that.
  • condition for inducing neural crest cells refers to time, medium, temperature, humidity, or a combination thereof, etc. that allow neural crest cells to be induced.
  • a condition seems to maintain adipose-derived stem cells or neural stem cells alone. It is understood that this can overlap with other conditions. Therefore, such conditions can be changed as appropriate. Also, once such favorable conditions have been established, such conditions can subsequently be used to induce similar neural crest cells.
  • sufficient conditions for inducing such neural crest cells can be used in vitro, in vivo, or ex vivo.
  • the conditions in the transplanted body part are applied as they are.
  • the stem cells and the differentiated cells may be immediately transplanted and mixed and cultured in an in-vitro environment in an in vivo environment. Autotransplantation can also be called ex vivo transplantation.
  • a “condition for inducing neural crest cells” the trunk cells that have been separated from the conventional tissue strength of the nervous system are used.
  • a condition known as a condition for dividing the cell into -eurosphere can be mentioned. It has been found for the first time in the present invention that such “conditions for separating the stem cells of the nervous system into the eurospheres” are effective as conditions for inducing adipose-derived stem cells into neural crest cells. Effect.
  • condition for separating the stem cells separated from the nervous system into -eurospheres are the conditions described in the examples (cultivation in "neurosphere culture medium”). In addition to the above, those having the following conditions can be mentioned.
  • epidermal growth factor e.g. epidermal growth factor, basic fibroblast growth factor, B27 supplement (GIBCO)>
  • BME Eagle Basal Medium
  • MEM Minimum Essential Medium
  • DMEM Dulbecco's Modified Eagle Medium
  • HAMF12 medium there is! /, or their mixed medium
  • the cell density condition is the condition of Kang KS et al.
  • DMEMZF12 (1: 1) is used, and it has been found that it is more advantageous to include glutamic acid, aspartic acid, and iron sulfate.
  • the present invention includes an excitatory amino acid, glutamate or aspartate, so the environment is completely different from the Neurobasal medium TM used to maintain differentiated neurons. It's different.
  • excitable amino acids is the Although it is thought to have an adverse effect such as wasting, on the other hand, it is considered to have a positive effect on the induction from fat into pluripotent cells of the nervous system (for example, neural crest cells).
  • in vivo refers to the interior of an organism. In a particular context, “in vivo” refers to the location (eg, desired site! / Where desired) where the tissue or organ of interest is to be placed.
  • in vitro refers to the removal or release of a portion of a living organism "ex vivo" (eg, in a test tube) for various research purposes. Let's see the state. A term that contrasts with in vivo.
  • ex vivo refers to the extraction of a target cell for gene transfer from a subject and the introduction of a therapeutic gene into the cell in vitro, and then again. When returning to the same subject, a series of operations is called ex vivo.
  • autograft tissue, cell, organ, etc.
  • tissue, cell, organ, etc. refers to a graft (tissue, cell, organ, etc.) from another genetically identical individual (eg, monozygotic twin) in a broad sense.
  • self expression is used interchangeably when derived from a subject.
  • the expression not derived from a subject has the same meaning as not being self (ie, non-self).
  • allogeneic grafts, tissues, cells, organs, etc.
  • allogeneic ones grafts, tissues, cells, organs, etc.
  • immune response in the transplanted individual (recipient).
  • examples of such include, but are not limited to, those derived from the parent (grafts, tissues, cells, organs, etc.).
  • heterologous refers to a transplant (graft, tissue, cell, organ, etc.) transplanted from a heterogeneous individual.
  • a human-derived one graft, tissue, cell, organ, etc.
  • tissue, cell, organ, etc. graft, tissue, cell, organ, etc.
  • the conditions for isolating such neural stem cells are independently about 1 X 10 4 cells. Zml to about 1 X 10 6 cells Zml. Density (eg, about 1 X 10 5 cells Zml), addition of growth factors (eg, EGF, bFGF), nerve inducers (eg, piotin, L-carcin, corticosterone, ethanolamine, D ( +) —Galactose, dartathione (reduced form), linolenoreic acid, linolenic acid, progesterone, retinoreacetate, selenium, triodothyronine (T3), DL—a—tocopherol, DL-a—tocopherol acetate, Addition of albumin (us), catalase, insulin, superoxide dismutase, transferrin), penicillin and streptomycin [], penicillin!
  • growth factors eg, EGF, bFGF
  • nerve inducers
  • the trophic factor may be any as long as it has the effect of assisting or promoting nerve induction
  • the nerve inducer may be piotin, L-carcin, corti Costerone, ethanolamine, D (+) galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retino-acetate, selenium, tolydothyrone (T3), DL—a Tocopheronole, DL- a Tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin
  • the nerve inducer may be B27 supplement (GIBCO).
  • the above conditions may be employed to maintain neural stem cells, neural crest cells, and the like, but are not limited thereto.
  • any culture medium containing a factor that promotes induction from adipose-derived stem cells to neural crest cells can be used.
  • Incubation in a medium supplemented with lOOUZml penicillin and 100 gZ ml streptomycin includes, but is not limited to, 37 ° C, oxygen 20%, carbon dioxide 5% and 80% or more. Can be used in humidity.
  • the term "factor that promotes induction into neural crest cells” or “nerve crest cell induction promoting factor” refers to a factor that promotes induction into neural crest cells (eg, As long as it is a chemical substance, temperature, etc.).
  • factors include various environmental factors. Examples of such factors include temperature, humidity, pH, salt concentration, nutrition, metal, gas, organic solvent, pressure, chemical Examples include, but are not limited to, substances (eg, steroids, antibiotics, etc.) or any combination thereof.
  • EGF epidermal growth factor
  • BMP-4 NM001202 (human)
  • Noelinl NM01 4279 (human)
  • condition for separating neuronal tissue from separated stem cells into neurospheres are effective as conditions for inducing adipose-derived stem cells to neural crest cells.
  • the “factor that promotes induction into neural crest cells” or “factor that promotes neural crest cell induction” is essentially based on “conditions for differentiating stem cells from the nervous system to differentiate into eurospheres”. Can be determined.
  • the term "adipose-derived neural crest cell mixture” refers to a mixture comprising the neural crest cells of the present invention and adipose stem cells.
  • the mixture of the present invention may be in any form as long as it contains neural crest cells and fat-derived hepatocytes.
  • the mixture of the present invention may further comprise neural stem cells.
  • the term "transplant” refers to the transfer of a cell, mixture, composition, medicament, etc. of the present invention into the body alone or in combination with other therapeutic agents.
  • the present invention relates to the following method and mode of introduction to the treatment site (for example, bone). And the amount introduced can be used: a method of directly injecting the pharmaceutical agent of the present invention into an injured site, suturing after insertion, and inserting.
  • the combination of adipose-derived stem cells of the invention and differentiated cells can be administered, for example, either simultaneously as a mixture, separately but simultaneously or concurrently; or sequentially.
  • Combination administration further includes the separate administration of one of the compounds or agents given first, followed by the second.
  • the term “self” or “self” refers to an individual, or an individual derived from the individual or a part thereof (eg, cell, thread and fabric, Organ). As used herein, the term “self” or “self” can broadly include grafts from other genetically identical individuals (eg, identical twins).
  • homologous refers to another individual force that is homogenous but genetically different (eg, cell, Tissue, organ, etc.). Because allogeneic individuals are genetically different, allogeneic individuals can elicit an immune response in the transplanted individual (recipient). Examples of such cells include, but are not limited to, parent-derived cells.
  • heterologous refers to something that is transplanted from a heterologous individual.
  • a transplant from a pig is a xenotransplant.
  • the term "recipient” refers to an individual that receives cells to be transplanted and the like, and is also referred to as "host”.
  • an individual that provides cells to be transplanted is called a “donor”.
  • the recipient and donor can be the same or different.
  • the cells used in the present invention may be derived from the same line (derived from the self (autologous)), from the same lineage (derived from another individual (other house)), or from a different species. Self-derived cells are preferred because of possible rejection, but may be from allogeneic if rejection is not a problem [0073]
  • "nervous disease, disorder or condition” refers to any disease, disorder or abnormal condition involving cells of the nervous system.
  • diagnosis, prevention, treatment or prognostic effective amount is medically effective in diagnosis, prevention, treatment (or therapy) or prognosis, respectively. This is the amount that can be recognized. Such an amount can be determined by a person skilled in the art using techniques well known in the art, taking into account various parameters.
  • the animal targeted by the present invention is an animal having adipocytes, any animal (for example, metal eels, shark eels, cartilaginous fish, teleosts, amphibians, reptiles, birds, mammals, etc.) ).
  • animals are mammals (eg, single pores, marsupials, rodents, wings, wings, carnivores, carnivores, long noses, odd hoofed animals, even hoofs, even Hoofs, rodents, scales, sea cattle, cetaceans, primates, rodents, maggots).
  • Exemplary subjects include, but are not limited to, animals such as rabbits, pigs, horses, chickens, cats and dogs. More preferably, a primate (for example, a chimpanzee, a second monkey, a human, etc.) is used. Most preferably, a human is used.
  • such medicament may further comprise a pharmaceutically acceptable carrier and the like.
  • a pharmaceutically acceptable carrier known in the art can be used.
  • Suitable formulation materials or pharmaceutically acceptable carriers include antioxidants, preservatives, colorants, flavors, diluents, emulsifiers, suspending agents, solvents, fillers, bulking agents, buffering agents. Including, but not limited to, delivery vehicles and Z or pharmaceutical adjuvants.
  • the medicament of the present invention is administered in the form of a composition comprising the cells of the present invention and other active ingredients together with at least one physiologically acceptable carrier, excipient or diluent.
  • a suitable vehicle may be an injection solution, a physiological solution, or an artificial cerebrospinal fluid that is supplemented with other substances commonly used in compositions for parenteral delivery. Is possible.
  • Acceptable carriers, excipients or stabilizers used herein are preferably non-toxic to the recipient, and preferably the dosage and concentration used.
  • Inactive preferably, for example, phosphate, citrate, or other organic acids; ascorbic acid, OC tocopherol; low molecular weight polypeptides; proteins (eg, serum albumin, gelatin or immunoglobulins) Hydrophilic polymers (eg polyvinylpyrrolidone); amino acids (eg glycine, glutamine, asparagine, arginine or lysine); monosaccharides, disaccharides and other carbohydrates (glucose, mannose or dextrin); chelating agents ( Eg, EDTA); sugar alcohols (eg, mantol or sorbitol); salt-forming counterions (eg, sodium); and Z or non-ionic surfactants (eg, Tween, pluronic or Polyethylene glycol (PEG)) I can get lost.
  • Inactive preferably, for example
  • Exemplary suitable carriers include neutral buffered saline or saline mixed with serum albumin.
  • the product is formulated as a lyophilizer using a suitable excipient (eg, sucrose).
  • suitable excipient eg, sucrose
  • Other standard carriers, diluents and excipients may be included as desired.
  • Other exemplary compositions include a Tris buffer having a pH of about 7.0 to 8.5 or an acetate buffer having a pH of about 4.0 to 5.5, which further includes sorbitol or a suitable replacement thereof. Can contain things.
  • a general method for preparing the pharmaceutical composition of the present invention is shown below. It should be noted that veterinary pharmaceutical compositions, quasi-drugs, marine pharmaceutical compositions, food compositions, baboon cosmetic compositions, and the like can also be produced by known preparation methods.
  • the cells of the present invention are blended with a pharmaceutically acceptable carrier as needed, and are administered parenterally, for example, as liquid preparations such as injections, suspensions, solutions and sprays. be able to.
  • pharmaceutically acceptable carriers include excipients, lubricants, binders, disintegrants, disintegration inhibitors, absorption enhancers, absorbents, wetting agents, solvents, solubilizers, suspending agents. , Isotonic agents, buffering agents, soothing agents and the like.
  • formulation additives such as preservatives, antioxidants, colorants, sweeteners and the like can be used as necessary.
  • substances other than the polynucleotide, polypeptide and the like of the present invention can be added to the composition of the present invention.
  • parenteral routes of administration include, but are not limited to, intravenous, intramuscular, subcutaneous, intradermal, mucosal, intrarectal, intravaginal, topical, and dermal.
  • the medicament used in the present invention does not contain pyrogens It can be in the form of a pharmaceutically acceptable aqueous solution. It is within the skill of the artisan to consider pH, isotonicity, stability, etc. for the preparation of such pharmaceutically acceptable compositions.
  • Preferable examples of the solvent in the liquid preparation include an injection solution, alcohol, propylene dallicol, macrogol, sesame oil, corn oil and the like.
  • solubilizer in the liquid preparation include polyethylene glycol, propylene glycol, D-mannitol, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, and ken Examples include, but are not limited to, sodium acid.
  • suspending agent in the liquid preparation include, for example, stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, salted benzethonium, glyceryl monostearate, etc.
  • Surfactants such as polyvinyl alcohol, polypyrrole pyrrolidone, sodium carboxymethylcellulose, methinoresenorelose, hydroxymethinoresenorelose, hydroxyethinoresenorelose, hydroxyethylcellulose, hydroxypropylcellulose, etc. Functional polymers, etc.
  • Preferable examples of the isotonic agent in the liquid preparation include, but are not limited to, sodium chloride salt, glycerin, D-manntol and the like.
  • buffering agent in the liquid preparation include, but are not limited to, phosphate, acetate, carbonate, citrate and the like.
  • the soothing agent in the liquid preparation include, but are not limited to, benzyl alcohol, benzalkonium chloride and pro-in hydrochloride.
  • Preferable examples of the preservative in the liquid preparation include, but are not limited to, paraoxybenzoates, chlorobutanol, benzyl alcohol, 2-phenylethyl alcohol, dehydroacetic acid, sorbic acid and the like. .
  • antioxidant in the liquid preparation include, but are not limited to, sulfite, ascorbic acid, a tocopherol and cysteine.
  • solutions and suspensions are sterilized and blood or other It is preferably isotonic with the solvent at the injection site for these purposes.
  • these are sterilized by filtration using a bacteria retention filter or the like, blending with a bactericide, or irradiation.
  • the pharmaceutical composition of the present invention may contain a coloring agent, a preservative, a fragrance, a flavoring agent, a sweetener and the like, and other agents.
  • the amount of the composition used in the treatment method of the present invention depends on the purpose of use, the target disease (type, severity, etc.), the patient's age, weight, sex, medical history, cell morphology or type, etc. In view of this, it can be easily determined by those skilled in the art.
  • the frequency with which the treatment method of the present invention is applied to a subject (or patient) also depends on the purpose of use, target disease (type, severity, etc.), patient age, weight, gender, medical history, treatment history, etc. In view of this, it can be easily determined by those skilled in the art. Examples of the frequency include administration once a few months every day (for example, once a week, once a month). It is preferable to administer once a week-once a month with the progress of the test.
  • the amount to be administered can be determined by estimating the amount required by the site to be treated.
  • the term "instruction” refers to a method for administering or diagnosing the medicament of the present invention, a person who administers the medicament of the present invention, or the like. It is written for a person, a person to be diagnosed, or a person to be diagnosed (eg, doctor, patient, etc.). This instruction describes a word indicating an appropriate method for administering the diagnostic agent or medicine of the present invention. These instructions are prepared according to the format prescribed by the national supervisory authority (for example, the Ministry of Health, Labor and Welfare in Japan and the Food and Drug Administration (FDA) in the United States). It will be clearly stated that it has been approved. Instructions are so-called package inserts, and are not normally limited to paper-delivered forces, such as electronic media (eg, home pages (websites) provided on the Internet, emails, etc. Etc.) can also be provided.
  • the end of treatment according to the method of the present invention may be determined by the results of standard laboratory tests using commercially available accessories or instrumentation or diseases associated with the intended treatment (eg, For example, it can be supported by the disappearance of clinical symptoms characteristic of bone disease, heart disease, nerve disease, etc., or recovery of the cosmetic state (eg, visual recovery, etc.). Treatment can be resumed by a recurrence of a disease (eg, neurological disease) associated with a lack of differentiated cells or a cosmetic condition injury.
  • a disease eg, neurological disease
  • the present invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more ingredients of a pharmaceutical composition.
  • a notice of the form prescribed by the government that regulates the manufacture, use or sale of a medicinal product or biological product may optionally be attached to such containers, and this notice may be made, used or administered for human administration. Represents approval by a government agency regarding sales.
  • the kit can include an infusion device.
  • Toxicity studies such as cells of the present invention can be performed using a model.
  • toxicity studies can be performed in appropriate animal models such as: (1) compounds are administered to mice (untreated control mice should also be used); (2) each One mouse in the treatment group also obtains blood samples periodically via the tail vein; and (3) The samples are analyzed for cells of the nervous system and the like. Comparison of the results for each dosing regimen with controls shows whether toxicity is present.
  • the neural crest cells of the present invention can be used for various purposes, for example, treatment of the nervous system.
  • forces including treatment of any disease, disorder or abnormal condition involving cells of the nervous system are not limited to these.
  • the present invention provides a method for preparing neural crest cells.
  • neural crest cells can be provided in a certain amount or more.
  • This method comprises the steps of A) obtaining stem cells from fat; and B) subjecting the stem cells to conditions for inducing neural crest cells.
  • stem cells derived from adipose are disclosed in International Publication No. OOZ53795, No. 03Z 022988, No. 01Z62901, Zuk, PA et al., Tissue Engineering, Vol. 7, 211-228, 2001, And Zuk, PA, et al., Molecular Biology of the Cell Vol., 13, 4279-4295, 2002, etc., or modifications thereof, and the fat portion of aspirate (lipoaspirate) by liposuction Force can also be separated. Specifically, for example, (1) suctioned fat is thoroughly washed with physiological saline using a 1-liter separatory funnel; (2) suction fat is in the upper layer, and physiological saline is in the lower layer.
  • the above sample can be filtered by suction using a 100 m mesh mesh; and (8) The resulting filtrate can be separated by centrifuging at 1200 g for 5 minutes.
  • adipose-derived stem cells can be isolated from, for example, a liquid part (liquid aspirate) of aspirate by liposuction as follows: (1) The liquid part of aspirate by liposuction is prepared. (2) The liquid portion is centrifuged to obtain a cell fraction; (3) The cell fraction is subjected to density gradient centrifugation, and the cell separation is performed based on specific gravity. And (4) the cells are recovered from a cell layer having a specific gravity lower than that of red blood cells.
  • the liquid portion of the aspirate can be prepared using saline or Ringer's infusion. Centrifugation can be performed at a speed of about 800 Xg or less, or about 400 Xg or more.
  • Density gradient centrifugation is performed at a rate of about 370 X g to l, 100 X g. Density gradient centrifugation is carried out using a solvent having a specific gravity (20 ° C.) of about 1.0 76 gZml to 1.07 gZml.
  • the solvent used in density gradient centrifugation can be Ficoll TM, Percoll TM or sucrose.
  • the specific gravity of the recovered cell layer can range from about 1.050 to 1.075.
  • the cell layer can be collected using a pipette.
  • the adipose-derived stem cells used in the present invention are selected from the group consisting of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106, CD 151, and SH3. Can express at least one (preferably two, three,... N) proteins. More preferably, the adipose-derived stem cells used in the present invention express all of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106, CD151, and SH3. It is possible.
  • CD antigens can be determined using a method known in the art (for example, an immunological technique using an antibody). Here, whether it is expressed or not can be appropriately selected by an immunological method or the like.
  • the adipose-derived stem cells used in the present invention are CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD135, and CD144. Does not express at least one (especially CD56).
  • a CD antigen is a marker for differentiated cells, and since it can be an indicator of stem cells that it is not expressed, it is not limited thereto.
  • the adipose-derived stem cells used in the present invention are CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD1 35 , And CD144 !, no deviations expressed! It may be advantageous to be a cell.
  • cells that express CD49d and do not express CD56 may be conveniently selected as adipose-derived stem cells.
  • the adipose-derived stem cells used in the present invention can be derived from aspirated fat.
  • aspirated fat Conventionally, sucked fat has been discarded, but in the present invention, neural stem cells (preferably neural crest cells) can be isolated from fat-derived stem cells.
  • neural stem cells preferably neural crest cells
  • aspiration fat may be, for example, the liquid part or the fat part of the aspirate from liposuction! /.
  • isolated ones may be used. Force-purified ones or completely purified ones may be used.
  • the adipose stem cells used in the present invention also have a portion of fat (for example, fat in the abdomen, chest, hips, thigh, upper arm, face, etc.) that is unnecessary for modern humans. Can be prepared from adipocytes.
  • the abdomen, buttocks, etc. are preferred. This is because the abdomen, buttocks, etc. are sites where fat tends to accumulate and are often desired to be removed.
  • a neural cell is prepared by subjecting a stem cell obtained from fat to a condition known as a condition for separating a stem cell separated from the tissue power of a conventional nervous system into -eurosphere.
  • Such conditions include, independently, a density of about 1 ⁇ 10 4 cells / ml to about 1 ⁇ 10 6 cells / ml (eg, about 1 ⁇ 10 5 cells / ml), a growth factor (eg, EGF) BFGF), nerve inducer (eg, piotin, L-carcin, corticosterone, ethanolamine, D (+)-galatose, glutathione (reduced form), linoleic acid, linolenic acid, progesterone, (Including tuluacetate, selenium, tolydothyrone (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin, catalase, insulin,
  • T3 a density of about 1
  • the power to use uncoated dishes can also be selected.
  • the neurotrophic factor that can be used herein may be any as long as it has an effect of assisting or promoting nerve induction.
  • the nerve-inducing factor is piotin, L-carthine, corticosterone, ethanolamine, D (+)-galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, Contains leturyl acetate, selenium, triodothyronine (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin.
  • the neural inducer can be B27 supplement (GIBCO).
  • neural stem cells eg, neural crest cells
  • neural stem cells eg, neural crest cells
  • any culture medium containing a factor that promotes induction from adipose-derived stem cells to neural crest cells can be used.
  • examples of such medium include DMEM / HAMF12 (1: 1) and human recombinant EGF (20 ng Zml, PeproTech, human thread and basic-FGF (20 ng / ml, Kaken Pharmaceutical, Japan), 2% B27 supplement.
  • EGF important for induction.
  • the method may further comprise the step of ascertaining whether the stem cell is expressing a neural crest cell marker, and optionally a neural stem cell marker.
  • the neural crest cells express at least one neural crest cell marker.
  • the neural crest cell may further express at least one neural stem cell marker.
  • cell markers of neural crest cells include, for example, CRABP1 (NM004378 (human)), AP2 (NM00 2097 (Human)), Slug (NM003068 (Human)), SoxlO (NM006941 (Human)), Snail (NM 005985 (Human)), Twist (NM000474 (Human)), Pax3 (NM000438 (Human)), Pax7 (N M002584 (chicken)), HNKl (NM004854 (chicken)), p75NTR (NM002507 (chicken)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002723 (human)) ), TPA (NM000930 (human)) and the like, but are not limited thereto.
  • CRABP1 NM004378 (human)
  • AP2 NM00 2097 (Human)
  • Slug NM003068 (Human)
  • SoxlO
  • neural stem cell markers include Nestin (NM006617 (human)), Musashi—1 (NM002 442 (blue)), CD133 (NM0006017 (blue)), notchl (NM017617 (blue)), Hesl (NM005524 ( ⁇ )), Mashl (NM004316 ( ⁇ )), Neurogenin (NM006161 ( ⁇ )), Pax6 (NM001604 ( ⁇ )), CD15 (NM002033 ( ⁇ )), PDGFR (NM0026 09 (human)) Although it is mentioned, it is not limited to these.
  • neural stem cell markers include Nestin and Musashi-l.
  • the method may further comprise the step of confirming the level of expression of the adipocyte marker by the stem cell.
  • the adipocyte marker used in the present method may be any as long as it can determine adipocytes, but preferably Leptin (NM000230 (human)) can be used. It is not limited to. Thus, confirmation of the level of adipocyte markers can be confirmed by a decrease in Leptin (NM000230 (human)).
  • the stem cells that have also obtained fat power become fat-derived neural crest cell precursors including neural crest cells and adipose-derived stem cells by the step of subjecting them to conditions for inducing neural crest cells. Can do.
  • the method wherein the stem cell expresses a neural crest cell marker or neural stem cell marker! may further comprise the step of confirming whether or not the stem cell is confirmed, and the step of confirming the level of expression of the fat cell marker by the stem cell.
  • cell markers of neural crest cells include, for example, CRABP1 (NM004378 (Hin)), AP2 (NM002097 (Hin)), Slug (NM003068 (Hin)), SoxlO (NM0069 41 (Hin) )), Snail (NM005985 (Hui)), Twist (NM000474 (H2)), Pax3 (NM00 0438 (H2)), Pax7 (NM002584 (H2)), HNKl (NM004854 (H2)), p75NTR (NM002507 (human)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), P0 (NM002723 (human)), tPA (NM000930 (human)), and the like.
  • CRABP1 NM004378 (Hin)
  • AP2 NM002097 (Hin)
  • Slug NM003068 (Hin)
  • SoxlO NM0069 41 (Hin)
  • neural stem cell markers include Nestin (NM006617 (tl)), Mu sashi-1 (NM002442 (chicken)), CD133 (NM0006017 (chicken)), notchl (NM017 617 (chicken)), Hesl (NM005524 ( ⁇ )), Mashl (NM004316 (()), Neurogenin (NM006161 ( ⁇ )), Pax6 (NM001604 ( ⁇ )), CD15 (NM002033 ( ⁇ )), PDGFR (NM002609 (human)) But is not limited to these!
  • neural stem cell markers include Nestin and Musashi-l.
  • the neural crest cells of the present invention may be characterized by expressing Nestin and Musashi-1 and expressing Leptin, which is an adipocyte marker, lower than the expression level of adipose-derived stem cells.
  • the determination using these neural crest cell marker, neural stem cell marker, or adipocyte marker is performed using a method known in the art (eg, RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.). Can be done. Here, whether it is expressed or not can be appropriately selected by RT-PCR, Northern blot, Western blot, immunostaining, FACS and the like.
  • a method known in the art eg, RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.
  • the method may further comprise the step of determining whether the stem cell has the ability to migrate. This migratory ability can be confirmed by transplanting into the head region of the embryonic cell and then observing that it migrates to the frontal nasal protuberance, maxillary protuberance, and arch mesenchyme.
  • a neural crest cell derived from fat is provided.
  • Such neural crest cells can be obtained by the above-described method of the present invention.
  • the fat can be a fat-derived stem cell, but it will be understood that the adipose tissue itself may be used.
  • the neural crest cells of the present invention may also have migration ability. This migration ability can be confirmed by transplanting the cells of interest into the head region of the embryo and then observing that they migrate to the frontal nasal protuberance, maxillary protuberance, and arch mesenchyme.
  • the neural crest cells of the present invention also have neural crest cell markers (for example, CRABP1 (NM004 378 (Hin)), AP2 (NM002097 (Hin)), Slug (NM003068 (Hin)), SoxlO (NM0).
  • CRABP1 NM004 378 (Hin)
  • AP2 NM002097 (Hin)
  • Slug NM003068 (Hin)
  • SoxlO NM0
  • the neural crest cells of the present invention further comprise neural stem cell markers (for example, Nestin (NM0066 17 (Hin)), Musashi-1 (NM002442 (Hin)), CD133 (NM0006017 (Hin)), not chl (NM017617 (Human)), Hesl (NM005524 (Human)), Mashl (NM004316 (Human)), Neurogenin (NM006161 (Human)), Pax6 (NM001604 (Human)), CD15 (NM00 2033 (Human)) PDGFR (NM002609 (human)) can be expressed, preferably the neural crest cells of the present invention can be identified by identifying cells that express Nestin and Musashi-1.
  • neural stem cell markers for example, Nestin (NM0066 17 (Hin)), Musashi-1 (NM002442 (Hin)), CD133 (NM0006017 (Hin)), not chl (NM017617 (Human)), Hesl (NM005524 (Human)), Mashl (
  • the present invention it is confirmed whether or not it has at least one characteristic selected from the group consisting of unique migration ability and expression of a cell marker of at least one neural crest cell. It can be identified as a neural crest cell derived from fat. Preferably, neural crest cells of the present invention are identified by confirming both the unique migration ability and the expression of cell markers of at least one neural crest cell. This unique migratory ability can be confirmed by transplanting into the embryonic head region and then observing that it migrates to the frontal nasal protuberance, maxillary protuberance, and meridian mesenchyme.
  • Leptin NM000230 (human) expression is lower than that of a fat-derived stem cell.
  • the cell of the present invention is characterized in that it expresses Nestin and Musashi-1, and the expression level of Leptin is lower than that of an adipose-derived stem cell.
  • the present invention provides a fat-derived neural crest cell mixture comprising neural crest cells and adipose-derived stem cells.
  • the neural crest cells contained in the fat-derived neural crest cell mixture of the present invention are in any form as described in the “adipose-derived neural crest cells” and “method for preparing neural crest cells” in the present specification. possible.
  • the adipose-derived stem cells contained in the adipose-derived neural crest cell mixture of the present invention can be in any form as described in “Adipose-derived stem cells” in the present specification.
  • the mixture of the present invention Any form is acceptable as long as it includes vesicles and adipose-derived hepatocytes.
  • the mixture of the present invention may also contain neural stem cells.
  • the present invention provides a composition for cell transplantation for the treatment of nervous system diseases, disorders or conditions, comprising adipose-derived or adipose-derived stem cell-derived neural crest cell.
  • the nervous system disease, disorder or condition can be, but is not limited to, a disease, disorder or condition resulting from, for example, a loss of differentiated cells of the nervous system.
  • the composition can be used for any purpose as long as it is intended to treat or prevent a nervous system disease, disorder or condition.
  • the neural crest cells in the composition used may be in any form as described herein in “Fat-derived neural crest cells” and “Methods for preparing neural crest cells”. .
  • the neural crest cells can be independently xenogeneic, allogeneic or syngeneic with respect to the host to be transplanted. Preferably, they are allogeneic or syngeneic, more preferably syngeneic, but not limited thereto. Without being bound by theory, it is possible to suppress immune rejection if it is syngeneic. However, if rejection is predicted, a step of avoiding rejection may be further included. Procedures to avoid rejection are well known in the art, for example, from the New Surgery System, Section 12, Heart / Lung Transplantation from technical and ethical arrangements to implementation (Revised 3rd Edition), See Nakayama Shoten.
  • Immunosuppressants that prevent rejection include, for example, “Cyclosporine” (Sunday Miyun Z Neoral), “Tacrolimus” (Prograf), “Azathioprine” (Imlan), “Steroid Hormone” (predonin, methylpredonin) ), And “T-cell antibodies” ( ⁇ KT3, ATG, etc.), and the method used in many centers around the world as a preventive immunosuppressive therapy is a combination of three drugs “cyclosporine, azathioprine and steroid hormones”. is there.
  • the immunosuppressive agent is desirably administered at the same time as the agent of the present invention, but it is not absolutely necessary. Therefore, as long as the immunosuppressive effect is achieved, the immunosuppressive agent can be administered before or after the regeneration therapy / treatment of the present invention.
  • Such a composition may be provided as a medicament.
  • Such medicines are used for nervous system diseases, disorders It is used to treat or prevent a harm or condition (eg, a disease, disorder or condition resulting from a loss of neuronal dividing cells).
  • the medicament of the present invention may contain a pharmaceutically acceptable carrier in addition to such a composition.
  • a carrier e.g, a pharmaceutically acceptable carrier
  • any carrier described herein can be selected and used by one of ordinary skill in the art.
  • the invention provides a method for cell transplantation for the treatment of a nervous system disease, disorder or condition (eg, a disease, disorder or condition resulting from a loss of differentiated cells of the nervous system). To do.
  • This method includes the step of administering a neural crest cell derived from a fat or a fat-derived stem cell.
  • the neural crest cells used for transplantation can be in any form as described herein in “Adipose-derived neural crest cells” and “Methods for preparing neural crest cells”.
  • the neural crest cells can be administered by any method known in the art.
  • the neural crest cells can be injected using a syringe, a catheter, a tube, and the like, but are not limited thereto.
  • exemplary dosage forms include, but are not limited to, local injection (subcutaneous injection, intramuscular or intramuscular injection), intravenous injection, intraarterial injection, or tissue administration.
  • the present invention provides the use of neural crest cells for the preparation of a medicament for treating or preventing a nervous system disease, disorder or condition.
  • the neural crest cells used for the preparation of the medicament are in any form as described herein in “Adipose-derived neural crest cells” and “Method of preparing neural crest cells”. obtain [0131]
  • the present invention will be described based on examples. However, the following examples are provided for illustrative purposes only. Accordingly, the scope of the present invention is limited only by the appended claims, which are not limited to the above-described embodiment and the following examples.
  • adipose-derived stem cells were prepared from sucked fat from humans who gave consent to this experiment. Specifically, the aspirated fat was thoroughly washed with physiological saline using a 1-liter separatory funnel. After confirming that the aspirated fat was sufficiently separated in the upper layer and the physiological saline was sufficiently separated in the lower layer, the lower layer was discarded, and this was repeated until the physiological saline became almost transparent with the naked eye. In this example, it was performed five times.
  • the aspirated fat was added in an amount equal to 10 ml of 0.075% collagenase ZPBS, and incubated at 37 ° C for 30 minutes with good agitation. To this sample, the same amount of 10% serum-added DMEM was added and centrifuged at 1200 ⁇ g for 10 minutes.
  • the precipitated cells (mostly red blood cells) were transferred to several 50 ml polypropylene tubes and centrifuged (400 X g, 5 minutes).
  • Ficoll registered trademark
  • the cell solution was separated into 4 layers. From the top (A layer) Cell-free layer (Transparent), (B layer) Mononuclear cell layer (light red), (C layer) Ficoll layer (transparent), (D layer) Red blood cell layer (dark red) Adherent cell groups including vesicles were included in B and C layers. After the A layer was aspirated, the B layer and about 3 ml of the C layer were collected as a cell suspension and transferred to a 50 ml tube.
  • Rough cell specific gravity is determined by mixing density gradient centrifuge separation media such as Percoll TM , Readygrad TM, etc. in saline-sodium solution or sucrose solution, and collecting collected cells and density-one marker beads ( Add density marker beads) to the mixture, centrifuge, and check by checking the force of cells in which of the 5-10 layers divided by the beads (the layer containing the cells indicates the specific gravity of the cells) It is possible.
  • density gradient centrifuge separation media such as Percoll TM , Readygrad TM, etc. in saline-sodium solution or sucrose solution
  • Example 2 The stem cells collected in Example 2 were characterized using FACS according to the following procedure:
  • SM staining medium
  • a gZml labeled antibody (phycoerythrin (PE), allophycocyanin (APC) and Z or fluorescein isothiocyanate (FITC) was used for labeling) was added.
  • PE phytoerythrin
  • APC allophycocyanin
  • FITC fluorescein isothiocyanate
  • FACS Vantage (Becton Dickinson) was used. Using the antibody label as an indicator, the expression of various CD proteins in the isolated stem cells was analyzed. As a result, as shown in Table 4, stem cells derived from the liquid portion of the aspirate from fat suction were found to express CD90 and CD49d.
  • the isolated stem cells were subcultured twice in DMEM medium! Passaging was done at 80% confluence. Use the same procedure as above to incubate cells after the second passage. Analysis was performed. The results are shown in Table 4 below.
  • stem cells prepared by liquid partial force of aspirate by liposuction contain mesenchymal stem cells, but differ from the adipose-derived stem cell group prepared by conventional methods, and CD31, 34 positive cells are Included. Therefore, it can be understood that the stem cells prepared by the method of the present invention are a group of cells that can easily differentiate into vascular endothelium (angiogenesis) with high efficiency. Furthermore, since the CD expression used as an index in the present specification has been confirmed after being subcultured twice, the stem cell of the present invention shows its phenotype after about two subcultures. It is understood that there is little change.
  • stem cells were collected from the liquid portion of the aspirate obtained by multiple subject force liposuction and characterized. The results are shown below.
  • Numbers indicate the percentage of stem cells expressing each protein in the cell population.
  • the collected stem cells are most of the population of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106 Positive for CD151 and SH3. Therefore, the adipose-derived stem cells of the present invention are selected from the group consisting of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106, CD151, and SH3 force. Both are cells that express one protein.
  • One feature of the adipose-derived stem cells used in the present invention is that they are stem cells that express CD106. Also, for CD31, CD45, CD117, and CD146, some of the stem cell populations were positive and some were negative.
  • the stem cell population was negative for CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD135, and CD144. Therefore, the adipose-derived stem cells of the present invention do not express at least one of CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD135, and CD144 It is a cell.
  • this stem cell population was cultured in a differentiation-inducing medium, expression of organ-specific proteins such as bone, cartilage, and fat was observed in 2 to 3 weeks.
  • This stem cell population unlike human fibroblast-derived cultured fibroblasts, did not express CD56, which is expressed in many fibroblast cells.
  • CD105 expressed by this population of stem cells was not normally seen in fibroblasts.
  • CD49d expressed by this stem cell population was not normally seen in bone marrow-derived mesenchymal stem cells.
  • CD31, CD34, CD36, CD45, CD106, and CD117 tended to disappear when the culture period was prolonged. Therefore, when subculture is continued, CD106 expression observed before subculture may not be observed.
  • Example 5 Isolation of human adipose-derived stem cells and cell culture of neurospheres
  • Adipose-derived stem cells were also isolated from human aspirated fat as reported by Yoshimura K et al., J Cell Physiol 2006; 208: 64-76.
  • suctioned fat was digested on a shaker for 30 minutes at 37 ° C with 0.075% collagenase in PBS. Mature adipocytes and connective yarns and weaves were removed by centrifugation. Blood cells again The fat-derived stem cell pellet was obtained by treatment with an erythrocyte lysis buffer.
  • the fluid partial force of liposuction aspirate can also be achieved by treating fat-derived stem cells with erythrocyte lysis buffer and density gradient centrifugation using FicolKGE Healthcare Bio-sciences, Piscataway, NJ). Isolated. It was confirmed that the obtained cells were adipose-derived stem cells by subjecting the cells to a method for inducing differentiation into fat, cartilage, and bone.
  • a method slightly modified from the method described in Kanemura Y et al., J Neurosci Res 2002; 69: 869-879 was used.
  • Freshly isolated adipose-derived stem cells are plated in 10 cm uncoated dishes at a density of 2 x 10 7 cells (approximately 1 x 10 5 cells Zml) and in a 5% CO environment under humidity. Underneath at 37 ° C, Neuros Hue
  • Neurosphere culture medium consists of human recombinant EGF (20ng / ml, PeproTech), human thread and basic-FGF (20ngZ ml, Kaken Pharmaceutic al, Japan), 2% B27 supplement (GIBCO), lOOUZml penicillin and 100 ⁇ gZml DMEMZHAMF12 (1: 1) as a basal medium supplemented with streptomycin. Half of the medium was replaced with fresh medium on days 4-5 and passage was performed on day 8.
  • adipose-derived stem cells When adipose-derived stem cells are cultured in a neurosphere culture medium (without serum, and containing EGF and basic FGF), the floating adipose-derived stem cells form small clumps on the third day of culture of the newly prepared adipose-derived stem cells.
  • aggregation of o neuro Sufuea like cells were observed in bright et force on day 5 (see FIG. 1A.) 0 the number and size of the Sufuea, due within 2 days (See Figures 1B and 1C.) Passaged on the 8th day. During this passage, the spheres were dissociated and resuspended in fresh media. On day 7 after passage, a new globular cell mass formed (data not shown.) O This suggests the neurosphere's ability to self-replicate.
  • Adipose-derived stem cells were treated with M199 medium and 10% fetal bovine serum. : Pre-cultured in a standard medium containing FBS). Next, total mRNA was extracted from Neurospora derived from the first passage of adipose-derived stem cells using an RNeasy-mini kit (Qiagen, Hilden Germany). This preculture was necessary to reduce blood cell contamination. Control mRNA was also extracted from undissolved adipose-derived stem cells cultured in M199 and 10% FBS.
  • RT-PCR was used to examine the expression of the neural stem cell markers Nestin and Musashi-1 genes and the adipogenic marker Leptin.
  • the Nestin and Musashi-1 gene expression was cultured in neurosphere culture media! /, Well upregulated in neurospheres compared to control adipose-derived stem cells (see Figure 2A and B). O This result indicates that neurospheres have characteristics of neural progenitor cells Is suggested. In contrast, Leptin expression was dramatically reduced in neurospheres (see Figure 2C). 0 This result indicates that neurospheres have reduced adipogenic capacity.
  • adipose-derived stem cell force-eurosphere was obtained.
  • This-you The cell growth of the rothsphere is remarkably rapid and can be achieved with a variety of other organs (eg, dermis and heart)-faster than the eurosphere.
  • This remarkably fast cell growth suggests the availability of adipose-derived stem cells as a source of neural progenitor cells in regenerative medicine.
  • the cells constituting the neurosphere expressed Nestin and Musaxhi-l, which are the genes of neural stem cells. This is thought to reflect the tendency of neurospheres to differentiate into neural progenitor cells. This finding is further supported by reduced expression of Leptin, a marker for adipogenesis and maturation.
  • Eurospheres derived from human adipose-derived stem cells were isolated from the Sendai virus vector (as described in Li HO et al., J Virol 2000; 74: 6 564-6565 and Inoue M et al., J Virol 2003; 77: 3238-3246).
  • DNAVEC corp. Tsukuba, Japan was used to transfect with green fluorescent protein (GFP).
  • GFP green fluorescent protein
  • the original vector SeVZ lacks the F gene encoding the fusion protein required for entry of the ribonucleotide complex into infected cells, so this vector is neither infectious nor pathogenic.
  • the modified SevZ A F vector has additional mutations that reduce cytotoxicity.
  • a modified vector was used. Neurospheres were incubated for 1 hour in a medium containing modified SeVZ (multiplicity of infection of 250) carrying the GFP gene, and then rinsed with phosphate buffered saline.
  • the eurospheres induced by the present invention expressed the neural stem cell marker genes Ne stin and Musashi-1 (see Example 6), and had the same migration ability as neural crest cells. Therefore, it can be said that this -Eurosphere contains neural crest cells.
  • the human adipose-derived stem cells obtained by this example were able to produce -eurosphere in about one week. It was observed to form. Furthermore, it was confirmed that the neurosphere formed from the human adipose-derived stem cells of the present invention is a cell mixture containing neural crest cells, adipose-derived stem cells, and neural stem cells. The cell mixture of the present invention was also able to form eurospheres in about one week even after repeated passages.
  • the cell mixture of the present invention proliferates at a remarkably fast rate. It was confirmed that it has the ability to form a and includes neural crest cells.
  • the neural crest cells derived from fat or adipose-derived stem cells of the present invention are considered to be applicable to regenerative medicine for treating nervous system diseases, disorders or conditions.
  • Example 8 Isolation of mouse adipose-derived stem cells and cell culture of neurospheres
  • Mouse adipose-derived stem cells are isolated using the same method as in Example 5 except that mice are used instead of humans as specimens.
  • the mouse adipose-derived stem cells are characterized by the same method as in Example 3.
  • mouse adipose-derived stem cells are cultured in a neurosphere culture medium. Half of the medium is replaced with fresh medium on days 4-5 and passage is performed on day 8.
  • Adipose-derived stem cells were treated with neurosphere culture medium (serum-free, EGF and basic
  • Example 9 Quantitative real-time RT—PCR (reverse transcriptase polymerase chain reaction)
  • PCR reverse transcriptase polymerase chain reaction
  • Example 10 Mouse whole embryo culture and mouse-eurosphere-like cell transplantation
  • Example 8 Mouse whole embryo culture and mouse-eurosphere-like cell transplantation
  • Mouse GFP positive cells transplanted into mouse embryos are clearly observed in some of the transplanted embryos and appear to survive.
  • This GFP positive cell can be confirmed to be incorporated into the craniofacial region of the embryo and the heart and trunk.
  • the transplanted cells are arranged in a row along the second arch, which can be confirmed to be very similar to the turn of the neural crest cells that migrate in the second arch. This result suggests an interesting possibility that the cells of murine adipose-derived stem cells exhibit neuronal crest-like properties in mouse embryos.
  • the mouse adipose-derived stem cells obtained in this example form the spheres in about one week.
  • the neurosphere formed with the mouse adipose-derived stem cell force of the present invention is a cell mixture containing neural crest cells, adipose-derived stem cells and neural stem cells.
  • the cell mixture of the present invention can also form a microsphere in about 1 week even after repeated passages.
  • the cell mixture of the present invention has the ability to proliferate at a remarkably fast rate to form a rosphae and contains neural crest cells.
  • the neural crest cells in which the mouse adipose-derived stem cell force of the present invention is induced can also be used for transplantation into mouse embryos of the same species.
  • DMEMZHAMF12 (1: 1) as the medium for neurosphere culture
  • BME MEM DMEM or HAMF12 medium to examine the effect of the medium on cell culture of rosophane.
  • the other materials and methods are the same as in Example 5.
  • Adipose-derived stem cells were treated with neurosphere culture medium (serum-free, EGF and basic
  • Example 7 (Mouse whole embryo culture and transplantation of eurosphere-like cells) [0183] The same method as in Example 7 is used, except that the eurosphere cultured in this example is used.
  • GFP positive cells transplanted into mouse embryos are clearly observed in some of the transplanted embryos and appear to survive. It can be confirmed that the GFP positive cells are incorporated into the craniofacial region of the embryo and the heart and trunk. In particular, the transplanted cells are arranged in a row along the second arch, which can be confirmed to be very similar to the pattern of neural crest cells that migrate in the second arch. This result suggests that the neurosphere cells induced by adipose-derived stem cell force have a neural crest cell-like property, which is interesting and possible.
  • Example 5 the human adipose-derived stem cells isolated in the above are passaged 5 times in DMEM medium to prepare and culture eurosphere.
  • Example 7 The same method as in Example 7 is used except that the spheres of this example are used.
  • Mouse GFP positive cells transplanted into mouse embryos are clearly observed in some of the transplanted embryos and appear to survive.
  • This GFP positive cell can be confirmed to be incorporated into the craniofacial region of the embryo and the heart and trunk.
  • the transplanted cells are arranged in a row along the second arch, which can be confirmed to be very similar to the turn of the neural crest cells that migrate in the second arch. This result suggests an interesting possibility that the cells of murine adipose-derived stem cells exhibit neuronal crest-like properties in mouse embryos.
  • adipose-derived stem cells can be used even if they are cultured and expanded and maintained after acquisition. Furthermore, it is confirmed that neurospheres can be prepared from adipose-derived stem cells cultured and proliferated, and neural crest cells can be prepared.
  • CRABP1 (NM004378 (human)), AP2 (NM002097 (human)), Slug (NM003068 (human)), SoxlO (NM006941 (human)), Snail (NM005985 (human)), Twist (NM0004 74) (Human)), Pax3 (NM000438 (human)), Pax7 (NM002584 (human)), HNKl (NMO 04854 (human)), p75NTR (NM002507 (human)), TRP2 (NM006267 (human)), Wntl (NM005430 ( (Human)), PO (NM002723 (Human)), tPA (NM000930 (Human)) gene expression is significantly up-regulated in neurospheres compared to control adipose-derived stem cells not cultured in neurosphere culture medium Make sure. This result indicates that neurospheres express neural crest cell markers.
  • neurospheres express neural crest cell markers by specific staining methods, immunohistochemical methods, in situ hybridization methods, and Western blotting methods for each marker.
  • the present invention proves that fat-derived or fat-derived stem cell-derived neural crest cells that can be obtained by a simple method can be applied to regenerative medicine. Therefore, the industrial use of the present invention is found in the pharmaceutical industry.

Abstract

The invention provides neural crest cells applicable to regenerative medicine in a treatment of a nervous system disease, disorder or state by a simple method. In particular, the invention provides fat-derived neural crest cells. The invention also provides neural crest cells induced from fat-derived stem cells. The invention also provides a method for preparing the neural crest cells. The method includes the steps of: A) obtaining progenitor cells from fat; and B) subjecting the stem cells to conditions to induce neural crest cells. The invention also provides a composition and a mixture for cell transplantation containing the neural crest cells, a method for cell transplantation and use of the neural crest cells for preparing a pharmaceutical.

Description

明 細 書  Specification
脂肪由来の神経堤細胞  Fat-derived neural crest cells
技術分野  Technical field
[0001] 本発明は、細胞分ィ匕の分野に関する。より詳細には、本発明は、脂肪由来または 脂肪由来幹細胞由来の神経堤細胞、その生成方法およびそれを利用した移植療法 に関する。  [0001] The present invention relates to the field of cell sorting. More particularly, the present invention relates to a fat-derived or fat-derived stem cell-derived neural crest cell, a method for producing the same, and a transplantation therapy using the same.
背景技術  Background art
[0002] 幹細胞の利用を中心とした、再生医療は、ここ数年で、力なりの発展を遂げてきた。  [0002] Regenerative medicine, centered on the use of stem cells, has made significant progress in recent years.
従来であれば、存在しないと考えられていた、種々の組織幹細胞が種々の組織から 発見され、同定されてきた。このように、再生医療による疾患治療が注目を浴びてい る。  Various tissue stem cells that were previously thought to be absent have been discovered and identified from various tissues. Thus, disease treatment by regenerative medicine is attracting attention.
[0003] しかし、再生医療は、臓器ないし組織機能不全を呈する多くの患者に対して日常的 に適応するまでには至っていない。今日まで、臓器移植のほか、医療機器での補助 システムの利用がごく限られたこのような患者に適応されて 、るにすぎな!/、。これらの 治療法には、ドナー不足、拒絶、感染、耐用年数などの問題がある。特に、ドナー不 足は深刻な問題である。  [0003] However, regenerative medicine has not yet been routinely applied to many patients with organ or tissue dysfunction. To date, it has only been applied to such patients with limited use of assistive systems in medical devices as well as organ transplants! /. These therapies have problems such as donor shortage, rejection, infection, and lifetime. In particular, lack of donors is a serious problem.
[0004] 受精卵は、原腸陥入の後、内胚葉、中胚葉および外胚葉の 3つの胚葉に分かれ、 外胚葉由来の細胞は、主に脳に存在し、神経幹細胞などが含まれる。中胚葉由来の 細胞は、主に骨髄に存在し、血管幹細胞、造血幹細胞および間葉系幹細胞などが 含まれる。内胚葉由来の細胞は、主に咽頭、肺、肝臓、脾臓、腸などに存在し、脾臓 幹細胞、肝臓幹細胞などが含まれる。  [0004] A fertilized egg is divided into three germ layers of endoderm, mesoderm and ectoderm after gastrulation, and cells derived from ectoderm mainly exist in the brain, and include neural stem cells and the like. Cells derived from mesoderm are mainly present in the bone marrow, and include vascular stem cells, hematopoietic stem cells, mesenchymal stem cells, and the like. Endoderm-derived cells are mainly present in the pharynx, lungs, liver, spleen, intestine, etc., and include spleen stem cells, liver stem cells, and the like.
[0005] 脂肪にも幹細胞があることが分力つてきた。脂肪由来幹細胞は、もともと、骨、軟骨 および脂肪組織のような間葉系組織に分ィ匕する脂肪吸引物 (liposuction aspirate )力も単離した間葉系幹細胞(mesenchymal stem cell : MSC)のサブタイプとし て報告されている(非特許文献 1)。現在、脂肪由来幹細胞は、再生医療のための最 も有望な成体幹細胞の 1つとして考えられている(非特許文献 2〜3)。なぜなら、脂 肪由来間質細胞は、脂肪吸引により安全に回収でき、かつ、良好な収量を期待でき るカゝらである。 [0005] The presence of stem cells in fat has been a major factor. Adipose-derived stem cells are originally subtypes of mesenchymal stem cells (MSCs) that have also isolated liposuction aspirate powers that can be separated into mesenchymal tissues such as bone, cartilage and adipose tissue. (Non-Patent Document 1). At present, adipose-derived stem cells are considered as one of the most promising adult stem cells for regenerative medicine (Non-Patent Documents 2 to 3). This is because fat-derived stromal cells can be safely recovered by liposuction and good yields can be expected. Ruka.
[0006] 幹細胞研究が進むにつれて、胚葉の境界を越えた細胞分化の新規な概念を見出 されてきた。 MSCおよび脂肪由来幹細胞は、本来、中胚葉系である力 神経系(つ まり、外胚葉)誘導体に分化し得る(非特許文献 4および 5)。最近、神経系の特徴を 有する幹細胞が、中胚葉組織 (例えば、真皮および心臓)から単離され得ることが報 告されている(非特許文献 6〜8)。これらの報告では、神経系の特徴を有する幹細胞 は、ニューロスフェア法により回収されている。ニューロスフェア法は、胚およびヒト脳 由来の神経幹細胞の単離スフ アの培養方法として開発されたものである。  [0006] As stem cell research has progressed, a new concept of cell differentiation across the germ layer boundary has been discovered. MSCs and adipose-derived stem cells can be differentiated into derivatives of the nervous system (ie, ectoderm), which is essentially a mesoderm system (Non-patent Documents 4 and 5). Recently, it has been reported that stem cells having nervous system characteristics can be isolated from mesoderm tissue (eg, dermis and heart) (Non-Patent Documents 6 to 8). In these reports, stem cells with neural characteristics have been collected by the neurosphere method. The neurosphere method was developed as a method for culturing isolated spheres of neural stem cells derived from embryos and human brain.
[0007] 成体哺乳動物の中枢神経系は再生しないと長い間考えられてきた力 成体の中枢 神経系にも神経幹細胞が存在することが報告された (非特許文献 9)。しかし、その数 は少なぐまた、単離するにも非常に多くの労力を必要とし、神経系疾患などの再生 医療において実際に用いることは困難である。したがって、神経系から神経系の多能 性細胞を得ることは実用的でない。そこで、脂肪を用いた神経系の分ィ匕について検 討がなされている。  [0007] It has been reported that neural stem cells are also present in the central nervous system of adults, which has long been thought to regenerate the central nervous system of adult mammals (Non-patent Document 9). However, the number is small and it requires a great deal of labor to isolate, and it is difficult to actually use it in regenerative medicine such as nervous system diseases. Therefore, obtaining pluripotent cells of the nervous system from the nervous system is not practical. Therefore, studies have been conducted on the division of the nervous system using fat.
[0008] Rang, KSらのグループは、霊長類由来の脂肪組織間質細胞 (非特許文献 10およ び 12では、ァカケサルが使用されている。非特許文献 11では、使用した動物は、「 非ヒト」霊長類であるとしか記載がな 、ようである。 )を神経細胞の維持のために開発 された Neurobasal medium™培地中で培養することによって-ユーロスフェアを形 成させる試みを行った (非特許文献 10〜 12)。しかし、 Rang KSらの文献では、作 製された-ユーロスフェアは、 NeuN、 MAP2、 NF160等を発現していることから、神 経細胞 (すなわち、分化細胞)に分化したものの凝集体が形成されていると思われる  [0008] A group of Rang, KS, et al. Has found that adipose tissue stromal cells derived from primates (in non-patent documents 10 and 12, akakasaru is used. In non-patent document 11, the animals used are “ It seems that it is only described as being a “non-human” primate.) By culturing in a Neurobasal medium ™ medium developed for the maintenance of neurons-an attempt was made to form eurospheres (Non-patent documents 10 to 12). However, according to Rang KS et al., The produced -Eurosphere expresses NeuN, MAP2, NF160, etc., and thus aggregates of those differentiated into neural cells (ie, differentiated cells) are formed. Seems to be
[0009] 神経堤細胞は、神経冠細胞とも称され、遊走能を有し、神経系の分化細胞への多 分化能 (例えば、後根神経節細胞、自律神経節細胞、副腎髄質クロム親和性細胞、 シュワン細胞、外皮色素細胞への分化能を有する)を有する細胞である。したがって 、神経堤細胞は分化細胞というよりは、広義の幹細胞であるといえる。神経堤細胞は 、胎性初期に神経管辺縁 (神経堤)より生じ、胚内を広く遊走しつつ、きわめて広範 かつ特異的なレパートリーに分ィ匕する細胞群 (第 4の胚葉)として同定された。現在で は、神経堤細胞は、顔面奇形の処置に使用されている。 [0009] Neural crest cells, also called neural crest cells, have the ability to migrate and have multipotency into differentiated cells of the nervous system (eg dorsal root ganglion cells, autonomic ganglion cells, adrenal medulla chromaffinity) A cell, a Schwann cell, and a cell having a differentiation potential into a coat pigment cell). Therefore, neural crest cells can be said to be broad stem cells rather than differentiated cells. Nerve crest cells are identified as a group of cells (fourth germ layer) that originates from the limbal limbus (nerve crest) in the early fetal period and migrates widely in the embryo and divides into a very wide and specific repertoire. It was done. At present Neural crest cells have been used to treat facial malformations.
[0010] 上記のように、 Kang, KSらのグループが作製した-ユーロスフェアは、分化細胞の 凝集体であるといえ、この-ユーロスフェアには、神経系の細胞に分化する多分化能 を有する細胞は存在しておらず、ましてや神経堤細胞については、この-ユーロスフ エアには皆無であると予想される。したがって、脂肪由来の幹細胞を使用した場合、 分化の誘導が顕著に生じ、神経系の多能性細胞、例えば、神経堤細胞への誘導は 起こらな 、と考えられて 、たと 、える。  [0010] As described above, the eurosphere produced by the group of Kang, KS, etc. can be said to be an aggregate of differentiated cells, and this -eurosphere has multipotency to differentiate into cells of the nervous system. No cells are present, and even neural crest cells are expected to be completely absent from this -Eurosphere. Therefore, when adipose-derived stem cells are used, differentiation is remarkably induced and it is considered that induction into pluripotent cells of the nervous system, such as neural crest cells, does not occur.
[0011] このことについては、特許文献 1〜6および非特許文献 4、 5、 13〜22等の多数の 文献にその証拠があるといえる。これらの文献には、脂肪由来幹細胞が神経に分ィ匕 する能力を有するようであることが記載されている。しかし、これらの文献では、脂肪 由来幹細胞から神経幹細胞、神経前駆細胞、ニューロスフ ア等の神経系の多能性 細胞への誘導について実質的に記載されておらず、直接神経への分化能を有する かどうかについて検討がなされているのみである。したがって、これらの文献には、脂 肪から神経幹細胞、神経堤細胞等の神経系の多能性細胞に誘導させることができる カゝどうかについては、何ら記載されておらず、むしろ、直接神経 (すなわち、分化細胞 )への分ィ匕が試みられていることから、当該分野においては神経系の多能性細胞へ 誘導させることはできな 、と考えられて 、たと 、える。  [0011] Regarding this, it can be said that there are evidences in many documents such as Patent Documents 1 to 6 and Non-Patent Documents 4, 5, and 13-22. These documents describe that adipose-derived stem cells appear to have the ability to be divided into nerves. However, these documents do not substantially describe the induction of adipose-derived stem cells into pluripotent cells of the nervous system such as neural stem cells, neural progenitor cells, and neurospheres, and have the ability to differentiate directly into nerves. Whether or not it is being studied. Therefore, these documents do not describe anything that can be induced from fat to neural pluripotent cells such as neural stem cells, neural crest cells, etc. In other words, since it has been attempted to differentiate into differentiated cells), it is considered that it cannot be induced into pluripotent cells of the nervous system in this field.
[0012] このように、脂肪から、特に、脂肪由来幹細胞力も神経分化させることの可能性はか なり高いと思われるが、反面、脂肪から、特に、脂肪由来幹細胞から、神経系の多能 性細胞を誘導することは、困難であると考えられてきた。  [0012] Thus, it seems that the possibility of nerve differentiation from fat, particularly fat-derived stem cell force, is quite high. On the other hand, from fat, particularly from fat-derived stem cells, the pluripotency of the nervous system. Inducing cells has been considered difficult.
[0013] しかし、脂肪、特に、脂肪由来幹細胞は、供給が豊富であることから、脂肪由来、特 に、脂肪由来幹細胞を用いて、神経系の多能性細胞を生産することができれば、神 経系の疾患、障害等の治療に画期的な変化をもたらすものといえ、そのような手法に 対する需要が高まって 、ると 、える。  [0013] However, since fat, in particular, adipose-derived stem cells are abundant in supply, if adipose-derived, in particular, adipose-derived stem cells can be used to produce pluripotent cells of the nervous system, It can be said that it will bring about a revolutionary change in the treatment of diseases and disorders of the trans-system, and the demand for such methods will increase.
特許文献 1:再表 WO2003Z008592号公報  Patent Document 1: Reprinted WO2003Z008592
特許文献 2:特表 2005 - 502352号公報  Patent Document 2: Japanese Translation of Special Publication 2005-502352
特許文献 3:特表 2002— 537849号公報  Patent Document 3: Japanese Translation of Special Publication 2002-537849
特許文献 4:米国特許出願公開第 2001Z0033834号明細書 特許文献 5:特表 2003 - 523767号公報 Patent Document 4: US Patent Application Publication No. 2001Z0033834 Patent Document 5: Special Table 2003-523767
特許文献 6:特表 2005 - 502712号公報 Patent Document 6: Special Table 2005-502712
非特許文献 l:Zuk PA, Zhu M, Mizuno H, et al. Tissue Eng.2001 ;7: 2 11-228 Non-patent literature l: Zuk PA, Zhu M, Mizuno H, et al. Tissue Eng. 2001; 7: 2 11-228
非特許文献 2:Zuk PA, Zhu M, Ashjian P, et al. Mol. Biol. Cell 2002 ;1 3:4279-4295 Non-Patent Document 2: Zuk PA, Zhu M, Ashjian P, et al. Mol. Biol. Cell 2002; 1 3: 4279-4295
非特許文献 3:Yoshimura K, Shigeura T, Matsumoto D, et al. J. Cell. P hysiol.2006;208:64-76 Non-Patent Document 3: Yoshimura K, Shigeura T, Matsumoto D, et al. J. Cell. Phisiol. 2006; 208: 64-76
非特許文献 4: Ashjian PH, Elbarbary AS, Edmonds B, et al. Plast. Reco nstr. Surg.2003;111:1922-1931 Non-Patent Document 4: Ashjian PH, Elbarbary AS, Edmonds B, et al. Plast. Recon nstr. Surg. 2003; 111: 1922-1931
非特許文献 5:Kokai LE, Rubin JP, Marra KG. Plast. Reconstr. Surg.20 05;116:1453-1460 Non-Patent Document 5: Kokai LE, Rubin JP, Marra KG. Plast. Reconstr. Surg. 20 05; 116: 1453-1460
非特許文献 6:Toma JG, Akhavan M, Fernandes KJ, et al. Nat. Cell. Bio 1.2001;3:778-784 Non-Patent Document 6: Toma JG, Akhavan M, Fernandes KJ, et al. Nat. Cell. Bio 1.2001; 3: 778-784
非特許文献 7: Fernandes KJ, McKenzie IA, Mill P, et al. Nat. Cell. Biol .2004;6:1082-1093 Non-Patent Document 7: Fernandes KJ, McKenzie IA, Mill P, et al. Nat. Cell. Biol. 2004; 6: 1082-1093
非特許文献 8:Tomita Y, Matsumura K, Wakamatsu Y, et al. J. Cell. Bi ol.2005;170:1135-1146 Non-Patent Document 8: Tomita Y, Matsumura K, Wakamatsu Y, et al. J. Cell. Biol. 2005; 170: 1135-1146
非特許文献 9:岡野栄之、実験医学、第 20卷、第 9号、 2002 ;1276— 1279 非特許文献 10:Kang SK, Putnam LA, Ylostalo J, et al. J. Cell. Sci.200Non-Patent Document 9: Enoyuki Okano, Experimental Medicine, No. 20, No. 9, 2002; 1276-1279 Non-Patent Document 10: Kang SK, Putnam LA, Ylostalo J, et al. J. Cell. Sci. 200
4; Aug 15;117(Pt 18) :4289-99 4; Aug 15; 117 (Pt 18): 4289-99
非特許文献 ll:Kang SK, Putnam L, Dufour J, et al. Stem Cells 2004 ;22:1356-1372 Non-patent literature ll: Kang SK, Putnam L, Dufour J, et al. Stem Cells 2004; 22: 1356-1372
非特許文献 12: Bunnell BA, Ylostalo J, Kang SK. Biochem. Biophys. Re s. Commun.2006; May 12;343(3) :762-71 Non-Patent Document 12: Bunnell BA, Ylostalo J, Kang SK.Biochem.Biophys.Res.Commun.2006; May 12; 343 (3): 762-71
非特許文献 13: Safford KM, Hicok KC, Safford SD, et al. Biochem. Bio phys. Res. Commun.2002 ;Jun 7 ;294 (2) :371— 9 Non-Patent Document 13: Safford KM, Hicok KC, Safford SD, et al. Biochem. Biophys. Res. Commun. 2002; Jun 7; 294 (2): 371-9
非特許文献 14:Tholpady SS, Katz AJ, Ogle RC. Anat. Rec. A Discov. Mol. Cell. Evol. Biol.2003 ;May; 272 (1) : 398-402 Non-Patent Document 14: Tholpady SS, Katz AJ, Ogle RC. Anat. Rec. A Discov. Mol. Cell. Evol. Biol. 2003; May; 272 (1): 398-402
非特許文献 15:Kang SK, Lee DH, Bae YC, et al. Exp. Neurol.2003 ;0 ct;183(2) :355-66  Non-Patent Document 15: Kang SK, Lee DH, Bae YC, et al. Exp. Neurol. 2003; 0 ct; 183 (2): 355-66
非特許文献 16:水野博司 J. Nippon Med. Sch.2003; Oct ;70 (5) :428-31 非特許文献 17: Saf ford KM, Safford SD, Gimble JM, et al. Exp. Neurol .2004 ;Jun;187(2) :319-28  Non-patent document 16: Hiroshi Mizuno J. Nippon Med. Sch. 2003; Oct; 70 (5): 428-31 Non-patent document 17: Saf ford KM, Safford SD, Gimble JM, et al. Exp. Neurol. 2004; Jun; 187 (2): 319-28
非特許文献 18: Yang LY, Liu XM, Sun B, et al. Chin. Med. J. (Engl).2 004;Mar;117(3) :425— 9  Non-Patent Document 18: Yang LY, Liu XM, Sun B, et al. Chin. Med. J. (Engl) .2 004; Mar; 117 (3): 425—9
非特許文献 19: Safford KM, Rice HE. Curr. Drug. Targets.2005; Feb 6 (1) :57-62  Non-Patent Document 19: Safford KM, Rice HE. Curr. Drug. Targets. 2005; Feb 6 (1): 57-62
非特許文献 20:Fujimura J, Ogawa R, Mizuno H, et al. Biochem. Biophy s. Res. Commun.2005; Jul 22;333(1) : 116-21  Non-Patent Document 20: Fujimura J, Ogawa R, Mizuno H, et al. Biochem. Biophys. Res. Commun. 2005; Jul 22; 333 (1): 116-21
非特許文献 21:Strem BM, Hicok KC, Zhu M, et al. Keio J. Med.2005 Non-Patent Document 21: Strem BM, Hicok KC, Zhu M, et al. Keio J. Med. 2005
Sep;54(3) :132-41 Sep; 54 (3): 132-41
非特許文献 22: Parker AM, Katz AJ. Expert. Opin. Biol. Ther.2006 Jun ;6(6) :567-78  Non-Patent Document 22: Parker AM, Katz AJ. Expert. Opin. Biol. Ther. 2006 Jun; 6 (6): 567-78
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0014] このように、当該分野において、神経系疾患、障害または状態の処置に利用可能 な神経幹細胞および神経前駆細胞の制御された簡便な調製方法への需要が高まつ ている。本発明は、このような需要に応えることを課題とする。さらに、本発明は、所望 の処置効果が得られる手術法およびそれに使用する材料または医薬を提供すること を課題とする。本発明は、原料として、豊富に存在する脂肪を用いて神経系の多能 性細胞を誘導することを課題とする。より詳細には、脂肪から神経堤細胞を誘導する ことを課題とする。 [0014] Thus, there is a growing demand in the art for a controlled and simple method for preparing neural stem cells and neural progenitor cells that can be used for the treatment of nervous system diseases, disorders or conditions. An object of the present invention is to meet such a demand. Furthermore, an object of the present invention is to provide a surgical method capable of obtaining a desired treatment effect and a material or a medicine used therefor. An object of the present invention is to induce neural pluripotent cells using abundant fat as a raw material. More specifically, an object is to induce neural crest cells from fat.
課題を解決するための手段  Means for solving the problem
[0015] 本発明は、本発明者らが上記課題を鋭意検討した結果、意外にも脂肪由来または 脂肪由来幹細胞由来の神経堤細胞を調製することによって、一部完成された。 したがって、本発明は、以下を提供する。 [0015] As a result of the present inventors diligently examining the above problems, the present invention was partially completed by unexpectedly preparing a neural crest cell derived from a fat or a fat-derived stem cell. Accordingly, the present invention provides the following.
(項目 1) (Item 1)
脂肪由来の神経堤細胞。 Fat-derived neural crest cells.
(項目 2) (Item 2)
脂肪由来幹細胞から誘導された、項目 1に記載の神経堤細胞。 2. The neural crest cell according to item 1, which is derived from an adipose-derived stem cell.
(項目 3) (Item 3)
前記神経堤細胞は、遊走能を有する、項目 1に記載の神経堤細胞。 2. The neural crest cell according to item 1, wherein the neural crest cell has a migration ability.
(項目 4) (Item 4)
前記遊走能は、胚の頭部領域に移植し、その後の前頭鼻隆起、上顎隆起または鰓 弓部の間充織への遊走能を観察することによって確認される、項目 3に記載の神経 堤細胞。 4. The neural crest according to item 3, wherein the migratory ability is confirmed by transplanting to the head region of the embryo and then observing the migratory ability to the frontal nasal protuberance, maxillary protuberance, or mesenteric mesenchyme. cell.
(項目 5) (Item 5)
前記神経堤細胞は、神経幹細胞マーカーを少なくとも 1つ発現することを特徴とする 、項目 1に記載の神経堤細胞。 2. The neural crest cell according to item 1, wherein the neural crest cell expresses at least one neural stem cell marker.
(項目 6) (Item 6)
前記神経幹細胞マーカーは、 Nestin、 Musashi—1、 CD133、 notch 1, Hesl、 M ashl、 Neurogenin、 Pax6、 CD15および PDGFRからなる群より選択される、項目 5に記載の神経堤細胞。 6. The neural crest cell according to item 5, wherein the neural stem cell marker is selected from the group consisting of Nestin, Musashi-1, CD133, notch1, Hesl, Mashl, Neurogenin, Pax6, CD15 and PDGFR.
(項目 7) (Item 7)
前記神経幹細胞マーカーは、 Nestinおよび Musashi— 1を含む、項目 6に記載の 神経堤細胞。 Item 7. The neural crest cell according to item 6, wherein the neural stem cell marker includes Nestin and Musashi-1.
(項目 8) (Item 8)
前記神経堤細胞は、神経堤細胞マーカーを少なくとも 1つ発現することを特徴とする 、項目 1に記載の神経堤細胞。 2. The neural crest cell according to item 1, wherein the neural crest cell expresses at least one neural crest cell marker.
(項目 9)前記神経堤細胞マーカーは、 CRABP1、 AP2、 Slug, SoxlO、 Snail, Tw ist、 Pax3、 Pax7、 HNK1、 p75NTR、 TRP2、 Wntl、 POおよび tPAからなる群よ り選択される、項目 8に記載の神経堤細胞。  (Item 9) The neural crest cell marker is selected from the group consisting of CRABP1, AP2, Slug, SoxlO, Snail, Twist, Pax3, Pax7, HNK1, p75NTR, TRP2, Wntl, PO, and tPA. The neural crest cell according to 1.
(項目 10) 前記神経堤細胞は、 Leptinの発現が脂肪由来幹細胞のものの発現レベルよりも低 いことを特徴とする、項目 1に記載の神経堤細胞。 (Item 10) Item 2. The neural crest cell according to item 1, wherein the neural crest cell has an expression level of Leptin lower than that of an adipose-derived stem cell.
(項目 11) (Item 11)
前記神経堤細胞は、 Nestinおよび Musashi— 1を発現し、かつ、 Leptinの発現が 脂肪由来前駆細胞のものの発現レベルよりも低いことを特徴とする、項目 1に記載の 神経堤細胞。 2. The neural crest cell according to item 1, wherein the neural crest cell expresses Nestin and Musashi-1, and the expression level of Leptin is lower than the expression level of adipose-derived progenitor cells.
(項目 12) (Item 12)
前記脂肪は、哺乳動物由来である、項目 1に記載の神経堤細胞。 Item 2. The neural crest cell according to Item 1, wherein the fat is derived from a mammal.
(項目 13) (Item 13)
前記脂肪は、ヒト由来である、項目 12に記載の神経堤細胞。 Item 13. The neural crest cell according to Item 12, wherein the fat is derived from a human.
(項目 14) (Item 14)
神経堤細胞を調製するための方法であって、以下の工程: A method for preparing neural crest cells comprising the following steps:
A)脂肪力も幹細胞を得る工程;および  A) the step of obtaining fat stem cells as well
B)該幹細胞を、神経堤細胞を誘導する条件に供する工程  B) Step of subjecting the stem cells to conditions for inducing neural crest cells
を包含する、方法。 Including the method.
(項目 15)  (Item 15)
前記前幹細胞は、脂肪由来幹細胞(adipose— derived stem or stromal cell) であることを特徴とする、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein the anterior stem cell is an adipose-derived stem or stromal cell.
(項目 16) (Item 16)
前記幹細胞を得る工程は、コラゲナーゼ処理および遠心分離処理を包含する、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein the step of obtaining the stem cell comprises collagenase treatment and centrifugation treatment.
(項目 17) (Item 17)
前記幹細胞は、 PLA細胞および LAF細胞力 なる群より選択される、項目 14に記載 の方法。 Item 15. The method according to Item 14, wherein the stem cell is selected from the group consisting of PLA cell and LAF cell force.
(項目 18) (Item 18)
前記 B)工程において、前記脂肪由来前駆細胞が、約 1 X 104細胞 Zml〜約 1 X 106 細胞 Zmlの密度で播種される、項目 14に記載の方法。 15. The method of item 14, wherein in step B), the adipose-derived progenitor cells are seeded at a density of about 1 × 10 4 cells Zml to about 1 × 10 6 cells Zml.
(項目 19) 前記 B)工程において、前記脂肪由来幹細胞が、約 1 X 105細胞 Zmlの密度で播種 される、項目 18に記載の方法。 (Item 19) Item 19. The method according to Item 18, wherein in the step B), the adipose-derived stem cells are seeded at a density of about 1 × 10 5 cells Zml.
(項目 20) (Item 20)
前記 B)工程において、前記脂肪由来幹細胞が、少なくとも上皮増殖因子 (EGF)を 含む培地中で培養される、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein in the step B), the adipose-derived stem cells are cultured in a medium containing at least epidermal growth factor (EGF).
(項目 21) (Item 21)
前記 B)工程において、前記脂肪由来幹細胞が、補助成分として、上皮増殖因子、 塩基性線維芽細胞増殖因子、神経誘導因子、ペニシリンおよびストレプトマイシンを 含む培地中で培養される、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein in step B), the adipose-derived stem cells are cultured in a medium containing epidermal growth factor, basic fibroblast growth factor, nerve induction factor, penicillin, and streptomycin as auxiliary components. .
(項目 22) (Item 22)
前記神経誘導因子が、ピオチン、 L—カル-チン、コルチコステロン、エタノールアミ ン、 D ( + )—ガラクトース、ダルタチオン (還元型)、リノール酸、リノレン酸、プロゲス テロン、レチュルアセテート、セレン、トリョードサイロニン(T3)、 DL— a—トコフエ口 ール、 DL— a—トコフエロールアセテート、アルブミン(ゥシ)、カタラーゼ、インスリン 、スーパーォキシドジスムターゼ、トランスフェリンを含む、項目 21に記載の方法。 (項目 23) The nerve-inducing factor is piotin, L-carcin, corticosterone, ethanolamine, D (+)-galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retulacetate, selenium, 28. The item according to item 21, comprising triodothyronine (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin (ushi), catalase, insulin, superoxide dismutase, and transferrin. Method. (Item 23)
前記 B)工程において、前記幹細胞を培養するために用いられる培地が、グルタミン 酸およびァスパラギン酸を含む、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein in the step B), a medium used for culturing the stem cells contains glutamic acid and aspartic acid.
(項目 24) (Item 24)
前記 B)工程において、前記幹細胞を培養するために用いられる培地が、イーグル基 礎培地(BME)、最小必須培地(MEM)、ダルベッコ改変イーグル培地(DMEM)ま たは HAMF12培地、あるいはそれらの混合培地である、項目 14に記載の方法。 (項目 25) In step B), the medium used for culturing the stem cells is an eagle basic medium (BME), a minimum essential medium (MEM), a Dulbecco's modified Eagle medium (DMEM), a HAMF12 medium, or a mixture thereof. Item 15. The method according to Item 14, which is a medium. (Item 25)
前記 B)工程において、前記幹細胞を培養するために用いられる培地が、ダルベッコ 改変イーグル培地(DMEM) ZF12 (1: 1)である、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein the medium used for culturing the stem cells in Step B) is Dulbecco's Modified Eagle Medium (DMEM) ZF12 (1: 1).
(項目 26) (Item 26)
前記 B)工程は、 2〜6日目に培地を新しいものに交換することを包含することを特徴 とする、項目 14に記載の方法。 (項目 27) 15. The method according to item 14, wherein the step B) includes replacing the medium with a new medium on days 2 to 6. (Item 27)
前記 B)工程は、 4〜5日目に培地を新しいものに交換することを包含することを特徴 とする、項目 14に記載の方法。 15. The method according to item 14, wherein the step B) includes replacing the medium with a new medium on the 4th to 5th days.
(項目 28) (Item 28)
前記 B)工程は、 6〜10日目に継代することを包含することを特徴とする、項目 14に 記載の方法。 The method according to Item 14, wherein the step B) includes subculture on the 6th to the 10th day.
(項目 29) (Item 29)
前記脂肪は、哺乳動物由来である、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein the fat is derived from a mammal.
(項目 30) (Item 30)
前記脂肪は、ヒト由来である、項目 29に記載の方法。 30. The method of item 29, wherein the fat is derived from a human.
(項目 31) (Item 31)
前記 B)工程において、前記脂肪由来幹細胞は、ヒト由来であり、 In the step B), the adipose-derived stem cells are human-derived,
該脂肪由来幹細胞は、約 1 X 105細胞 Zmlの密度で播種され、 The adipose-derived stem cells are seeded at a density of about 1 × 10 5 cells Zml;
該脂肪由来幹細胞は、補助成分として、上皮増殖因子、塩基性線維芽細胞増殖 因子、神経誘導因子、ペニシリンおよびストレプトマイシンを含む培地中で培養され 、ここで、該神経誘導因子は、ピオチン、 L—カル-チン、コルチコステロン、エタノー ルァミン、 D ( + ) ガラクトース、ダルタチオン (還元型)、リノール酸、リノレン酸、プロ ゲステロン、レチ-ノレアセテート、セレン、トリョードサイロ-ン(T3)、 DL— a トコフ エロール、 DL— a—トコフェロールアセテート、アルブミン(ゥシ)、カタラーゼ、インス リン、スーパーォキシドジスムターゼ、トランスフェリンを含み、そして  The adipose-derived stem cells are cultured in a medium containing epidermal growth factor, basic fibroblast growth factor, nerve induction factor, penicillin and streptomycin as auxiliary components, wherein the nerve induction factor is piotin, L— Carcin, corticosterone, ethanolamine, D (+) galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retino-acetate, selenium, tolydothyrone (T3), DL—a Tokov Contains erol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin, and
該培地は、ダルベッコ改変イーグル培地(DMEM) ZF12 (1: 1)である、項目 14 に記載の方法。  Item 15. The method according to Item 14, wherein the medium is Dulbecco's Modified Eagle Medium (DMEM) ZF12 (1: 1).
(項目 32) (Item 32)
前記幹細胞が神経幹細胞マーカーを発現しているかどうかを確認する工程をさらに 包含する、項目 14に記載の方法。 Item 15. The method according to Item 14, further comprising the step of confirming whether the stem cell expresses a neural stem cell marker.
(項目 33) (Item 33)
前記神経幹細胞マーカーは、 Nestin、 Musashi—1、 CD133、 notch 1, Hesl、 M ashl、 Neurogenin、 Pax6、 CD15および PDGFRからなる群より選択される、項目 32に記載の方法。 The neural stem cell marker is selected from the group consisting of Nestin, Musashi-1, CD133, notch 1, Hesl, Mashl, Neurogenin, Pax6, CD15 and PDGFR The method according to 32.
(項目 34) (Item 34)
前記神経幹細胞マーカーは、 Nestinおよび Musashi— 1を含む、項目 33に記載の 方法。 34. The method of item 33, wherein the neural stem cell marker comprises Nestin and Musashi-1.
(項目 35)  (Item 35)
前記幹細胞が神経堤細胞マーカーを発現しているかどうかを確認する工程をさらに 包含する、項目 14に記載の方法。 Item 15. The method according to Item 14, further comprising the step of confirming whether the stem cells express a neural crest cell marker.
(項目 36) (Item 36)
前記細胞は、神経堤細胞マーカーを少なくとも 1つ発現することを特徴とする、項目 1 4に記載の方法。 Item 15. The method according to Item 14, wherein the cell expresses at least one neural crest cell marker.
(項目 37) (Item 37)
前記神経堤細胞マーカーは、 CRABP1、 AP2、 Slug, SoxlO、 Snail, Twist, Pax 3、 Pax7、 HNK1、 p75NTR、 TRP2、 Wntl、 POおよび tP A力もなる群より選択さ れる、項目 36に記載の方法。 37. The method of item 36, wherein the neural crest cell marker is selected from the group consisting of CRABP1, AP2, Slug, SoxlO, Snail, Twist, Pax 3, Pax7, HNK1, p75NTR, TRP2, Wntl, PO, and tPA force. .
(項目 38) (Item 38)
前記幹細胞が脂肪細胞マーカーを発現のレベルを確認する工程をさらに包含する、 項目 14に記載の方法。 Item 15. The method according to Item 14, further comprising the step of confirming the level of expression of the adipocyte marker by the stem cell.
(項目 39) (Item 39)
前記脂肪細胞マーカーのレベルの確認は、 Leptinの減少の確認である、項目 38に 記載の方法。 The method according to Item 38, wherein the confirmation of the level of the adipocyte marker is confirmation of a decrease in Leptin.
(項目 40) (Item 40)
前記神経堤細胞は、 B)工程により、神経堤細胞と脂肪由来幹細胞とを含む脂肪由 来神経堤細胞前駆体となることを特徴とする、項目 14に記載の方法。 Item 15. The method according to Item 14, wherein the neural crest cell becomes a fat-derived neural crest cell precursor containing neural crest cells and adipose-derived stem cells by the step B).
(項目 41) (Item 41)
前記幹細胞が神経幹細胞マーカーおよび神経堤細胞マーカーを発現しているかど うかを確認する工程、および前記幹細胞が脂肪細胞マーカーを発現のレベルを確認 する工程をさらに包含する、項目 14に記載の方法。 15. The method according to item 14, further comprising the step of confirming whether the stem cell expresses a neural stem cell marker and a neural crest cell marker, and confirming the level of expression of the adipocyte marker by the stem cell.
(項目 42) 前記幹細胞が遊走能を有するかどうかを確認する工程をさらに包含する、項目 14に 記載の方法。 (Item 42) Item 15. The method according to Item 14, further comprising the step of confirming whether the stem cell has a migration ability.
(項目 43) (Item 43)
前記遊走能は、胚の頭部領域に移植し、その後の遊走能を観察することによって確 認される、項目 42に記載の方法。 43. The method according to Item 42, wherein the migration ability is confirmed by transplanting to a head region of an embryo and observing the subsequent migration ability.
(項目 44) (Item 44)
前記神経幹細胞マーカーは、 Nestinおよび Musashi— 1であり、かつ、前記脂肪細 胞マーカーは、 Leptinであることを特徴とする、項目 41に記載の方法。 Item 44. The method according to Item 41, wherein the neural stem cell marker is Nestin and Musashi-1, and the fat cell marker is Leptin.
(項目 45) (Item 45)
項目 1に記載の細胞を含む、神経系疾患、障害または状態の処置のための細胞移 植のための組成物。 A composition for cell transplantation for the treatment of a nervous system disease, disorder or condition, comprising the cell according to item 1.
(項目 46) (Item 46)
前記神経系疾患、障害または状態は、神経系の分化細胞の欠損に起因する疾患、 障害または状態である、項目 45に記載の組成物。 46. The composition according to item 45, wherein the nervous system disease, disorder or condition is a disease, disorder or condition caused by a defect in differentiated cells of the nervous system.
(項目 47) (Item 47)
前記脂肪は、前記神経系疾患、障害または状態の対象の個体と同種異系の関係に ある個体由来である、項目 45に記載の組成物。 46. The composition according to item 45, wherein the fat is derived from an individual having an allogeneic relationship with an individual subject to the nervous system disease, disorder or condition.
(項目 48) (Item 48)
前記脂肪は、前記神経系疾患、障害または状態の対象の個体と異種の関係にある 個体由来である、項目 45に記載の組成物。 46. A composition according to item 45, wherein the fat is derived from an individual having a heterogeneous relationship with the subject of the nervous system disease, disorder or condition.
(項目 49) (Item 49)
前記脂肪は、前記神経系疾患、障害または状態の対象の個体と同種同系の関係に ある個体由来である、項目 45に記載の組成物。 46. The composition according to item 45, wherein the fat is derived from an individual having an allogeneic relationship with an individual subject to the nervous system disease, disorder or condition.
(項目 50) (Item 50)
項目 1に記載の細胞を投与する工程を包含する、神経系疾患、障害または状態の処 置のための細胞移植のための方法。 A method for cell transplantation for the treatment of a nervous system disease, disorder or condition comprising the step of administering the cell according to item 1.
(項目 51) (Item 51)
前記神経系疾患、障害または状態は、神経系の分化細胞の欠損に起因する疾患、 障害または状態である、項目 50に記載の方法。 Said nervous system disease, disorder or condition is a disease caused by a loss of differentiated cells of the nervous system, 51. The method of item 50, wherein the method is a disorder or condition.
(項目 52)  (Item 52)
項目 1に記載の細胞の、神経系疾患、障害または状態を処置または予防するための 医薬の調製のための使用。  Use of the cell according to item 1 for the preparation of a medicament for treating or preventing a nervous system disease, disorder or condition.
(項目 53)  (Item 53)
前記神経系疾患、障害または状態は、神経系の分化細胞の欠損に起因する疾患、 障害または状態である、項目 52に記載の使用。  53. Use according to item 52, wherein the nervous system disease, disorder or condition is a disease, disorder or condition caused by a defect in differentiated cells of the nervous system.
(項目 54)  (Item 54)
神経堤細胞と脂肪由来幹細胞とを含む、脂肪由来神経堤細胞混合物。  An adipose-derived neural crest cell mixture comprising neural crest cells and adipose-derived stem cells.
(項目 55)  (Item 55)
神経幹細胞をさらに含む、項目 54に記載の脂肪由来神経堤細胞混合物。  55. The fat-derived neural crest cell mixture according to item 54, further comprising neural stem cells.
[0017] 従って、本発明のこれらおよび他の利点は、添付の図面を参照して、以下の詳細な 説明を読めば、明白である。 [0017] Accordingly, these and other advantages of the present invention will be apparent upon reading the following detailed description with reference to the accompanying drawings.
発明の効果  The invention's effect
[0018] 本発明により、脂肪由来の神経堤細胞および脂肪前駆細胞由来の神経堤細胞を 用いて再生治療ができる。本発明は、さらに、脂肪力 前駆細胞を回収し、その前駆 細胞を神経堤細胞を単離する条件に供することにより、神経堤細胞を調製することが できる。さらに、本発明はまた、神経堤細胞を移植することにより、神経系疾患、障害 または状態の処置を容易にする。これらの処置は、ほとんど副作用が予測されないこ と、およびその供給源が豊富なことから、再生医療において簡易かつ効率のよい処 置方法を提供する。本発明により、神経堤細胞を含むニューロスフェアを顕著に速い 速度で提供することができた。脂肪を原料とすることができるので、神経堤細胞混合 物を大量に提供することができるという効果も提供される。  [0018] According to the present invention, regenerative treatment can be performed using a neural crest cell derived from fat and a neural crest cell derived from fat precursor cell. In the present invention, a neural crest cell can be further prepared by recovering a fat progenitor cell and subjecting the progenitor cell to conditions for isolating the neural crest cell. Furthermore, the present invention also facilitates treatment of nervous system diseases, disorders or conditions by transplanting neural crest cells. These treatments provide a simple and efficient treatment method in regenerative medicine because few side effects are expected and their sources are abundant. According to the present invention, neurospheres containing neural crest cells could be provided at a remarkably high rate. Since fat can be used as a raw material, an effect that a large amount of neural crest cell mixture can be provided is also provided.
図面の簡単な説明  Brief Description of Drawings
[0019] [図 1]図 1は、ニューロスフェア培養培地での培養、 5日目(Day 5 :A)、 6日目(Day  [0019] [Fig. 1] Fig. 1 shows culture in a neurosphere culture medium, Day 5 (Day 5: A), Day 6 (Day
6 : B)および 7日目(Day 7 : C)の脂肪由来幹細胞の-ユーロスフエア形成を示す 写真(X 200)である。  6: Photo (X200) showing the formation of eurosphere of adipose-derived stem cells on day 7 (Day 7: C).
[図 2]図 2は、神経幹細胞マーカー Nestin (A)および Musashi— 1 (B)、ならびに 脂肪生成分ィ匕マーカー Leptin (C)の遺伝子発現の定量的リアル タイム RT—P CR分析の結果を示す (コントロール:未分化な脂肪由来幹細胞、スフエア:ニユーロ スフエア)。アツセィは、 3連で実施した。標準誤差をエラーバーで示す。 [Figure 2] Figure 2 shows neural stem cell markers Nestin (A) and Musashi— 1 (B), and The results of quantitative real-time RT-PCR analysis of gene expression of the adipogenic marker Leptin (C) are shown (control: undifferentiated adipose-derived stem cells, sphere: Nyuro sphere). Atsey was conducted in triplicate. Standard error is indicated by an error bar.
[図 3]図 3は、インビボで培養したマウス胚に移植した、 GFPで形質転換した脂肪由 来幹細胞由来の-ユーロスフエアの神経堤様遊走を示す。(A) 8日胚カも 40時間培 養したマウス胚の写真。(B, C)胚の蛍光写真。 GFP陽性-ユーロスフェア細胞は、 列になって配置された (矢印)。これは、ニューロスフェア細胞が第二鰓弓に沿って遊 走していることを示唆する。バーは 500 μ mである。  FIG. 3 shows neural crest-like migration of GFP-transformed adipose-derived stem cells transplanted into mouse embryos cultured in vivo. (A) A photograph of a mouse embryo that was cultured for 40 hours on day 8 embryos. (B, C) Fluorescent photographs of embryos. GFP positive-Eurosphere cells were arranged in a row (arrow). This suggests that neurosphere cells are migrating along the second arch. The bar is 500 μm.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0020] 以下、本発明を説明する。本明細書の全体にわたり、単数形の冠詞 (例えば、英語 の場合は「a」、「an」、「the」など)は、特に言及しない限り、その複数形の概念をも含 むことが理解されるべきである。また、本明細書において使用される用語は、特に言 及しない限り、当該分野で通常用いられる意味で用いられることが理解されるべきで ある。矛盾する場合、本明細書 (定義を含めて)が優先する。  [0020] Hereinafter, the present invention will be described. Throughout this specification, it should be understood that singular articles (eg, “a”, “an”, “the”, etc. in English) also include the plural concept unless otherwise stated. It should be. In addition, it should be understood that the terms used in this specification are used in the meanings usually used in the art unless otherwise specified. In case of conflict, the present specification, including definitions, will control.
[0021] (用語の定義)  [0021] (Definition of terms)
以下に本明細書において特に使用される用語の定義を列挙する。  Listed below are definitions of terms particularly used in the present specification.
[0022] 本明細書において使用される「細胞」は、当該分野において用いられる最も広義の 意味として定義され、多細胞生物の組織の構成単位であって、外界から生体を隔離 する膜構造に包まれ、内部に自己再生能を備え、遺伝情報およびその発現機構を 有する生命体をいう。本発明の方法においては、どのような細胞でも対象とされ得る。 本発明で使用される「細胞」の数は、光学顕微鏡を通じて計数することができる。光 学顕微鏡を通じて計数する場合は、核の数を数えることにより計数を行う。当該組織 を組織切片スライスとし、へマトキシリン—ェォシン (HE)染色を行うことにより細胞外 マトリクス (例えば、エラスチンまたはコラーゲン)および細胞に由来する核を染め分け る。この組織切片を光学顕微鏡にて検鏡し、特定の面積 (例えば、 200 mX 200 m)あたりの核の数を細胞数と見積って計数することができる。本明細書において使 用される細胞は、天然に存在する細胞であっても、人工的に改変された細胞 (例えば 、融合細胞、遺伝子改変細胞など)であってもよい。細胞の供給源としては、例えば、 単一の細胞培養物であり得、あるいは、正常に成長したトランスジエニック動物の胚、 血液、または体組織 (例えば、脂肪組織)、または正常に成長した細胞株由来の細胞 のような細胞混合物などが挙げられるがそれらに限定されない。また、このような供給 源をそのまま細胞として用いることもできる。 [0022] The term "cell" used herein is defined as the broadest meaning used in the field, and is a structural unit of a tissue of a multicellular organism and is enclosed in a membrane structure that isolates the living body from the outside world. Rarely, an organism with self-regenerative ability and genetic information and its expression mechanism. Any cell can be targeted in the method of the present invention. The number of “cells” used in the present invention can be counted through an optical microscope. When counting through an optical microscope, count by counting the number of nuclei. The tissue is sliced into tissue slices and stained with hematoxylin-eosin (HE) to separate extracellular matrix (eg, elastin or collagen) and cell-derived nuclei. This tissue section can be examined with an optical microscope, and the number of nuclei per specific area (for example, 200 m × 200 m) can be estimated and counted. The cells used in the present specification may be naturally occurring cells or artificially modified cells (eg, fusion cells, genetically modified cells, etc.). Examples of cell sources include: It can be a single cell culture, or a cell mixture such as cells from a normally grown transgenic animal embryo, blood, or body tissue (eg, adipose tissue), or a normally grown cell line However, it is not limited to them. Further, such a supply source can be used as a cell as it is.
[0023] 本発明において使用される脂肪細胞 (fat eelほたは adipocyte)およびその対応 物は、以下の生物が脂肪細胞またはその対応物を有する限り、どの生物 (例えば、メ クラウナギ類、ャッメゥナギ類、軟骨魚類、硬骨魚類、両生類、爬虫類、鳥類、哺乳動 物など)由来であってもよい。好ましくは、哺乳動物 (例えば、単孔類、有袋類、貧歯 類、皮翼類、翼手類、食肉類、食虫類、長鼻類、奇蹄類、偶蹄類、管歯類、有鱗類、 海牛類、クジラ目、霊長類、齧歯類、ゥサギ目など)由来であってもよい。 1つの実施 形態では、霊長類 (例えば、チンパンジー、二ホンザル、ヒト)由来の細胞、特にヒト由 来の細胞が用いられるがそれに限定されな 、。 [0023] The fat cells (fat eels or adipocytes) used in the present invention and their counterparts may be any organism (for example, larvae, jellyfishes) as long as the following organisms have adipocytes or their counterparts. , Cartilaginous fish, teleosts, amphibians, reptiles, birds, mammals, etc.). Preferably, mammals (e.g. single pores, marsupials, rodents, wings, wings, carnivores, carnivores, long noses, odd-hoofed animals, even-hoofed animals, rodents, It may be derived from scales, sea cattle, cetaceans, primates, rodents, maggots, etc.). In one embodiment, cells derived from primates (eg, chimpanzee, dihonosa, human), particularly cells derived from humans, are used, but are not limited thereto.
[0024] 本明細書において使用される場合、用語「幹細胞」とは、分化細胞の前駆細胞 (pr ecursorまたは progenitor)をいい、これは、単分化能性(monopotency)、多能性 (multipotency)、全能性(totipotency)を有する細胞をいう。本明細書では、「幹 細胞」は、「前駆細胞」と交換可能に用いられ得る。幹細胞は、特定の刺激に応答し て分化され得る。幹細胞は通常、組織が傷害を受けたときにその組織を再生すること ができる。本明細書で使用される幹細胞は、胚性幹 (ES)細胞または組織幹細胞 (組 織性幹細胞、組織特異的幹細胞または体性幹細胞とも 、う)または他の前駆細胞で あり得るがそれらに限定されない。また、上述の能力を有している限り、人工的に作 製した細胞 (例えば、本明細書において使用される融合細胞、再プログラム化された 細胞など)もまた、幹細胞であり得る。胚性幹細胞とは初期胚に由来する多能性幹細 胞(pluripotent stem cell)である。胚性幹細胞は、 1981年に初めて榭立され、 1 989年以降ノックアウトマウス作製にも応用されている。 1998年にはヒト胚性幹細胞 が榭立されており、再生医学にも利用されつつある。組織幹細胞は、胚性幹細胞と は異なり、分ィ匕の程度が比較的限定されている細胞であり、組織中に存在し、未分 化な細胞内構造をしている。組織幹細胞は、核 Z細胞質比が高ぐ細胞内小器官が 乏しい。組織幹細胞は、概して、多能性 (pluripotency)を有し、細胞周期が遅ぐ個 体の一生以上に増殖能を維持する。本明細書において使用される場合は、幹細胞 は好ましくは胚性幹細胞であり得るが、状況に応じて組織幹細胞も使用され得る。 [0024] As used herein, the term "stem cell" refers to a precursor cell of a differentiated cell (pr ecursor or progenitor), which is monopotency, multipotency. Refers to a cell having totipotency. As used herein, “stem cell” can be used interchangeably with “progenitor cell”. Stem cells can be differentiated in response to specific stimuli. Stem cells are usually able to regenerate the tissue when it is damaged. Stem cells as used herein can be, but are not limited to, embryonic stem (ES) cells or tissue stem cells (including tissue stem cells, tissue-specific stem cells or somatic stem cells) or other progenitor cells. Not. Also, an artificially produced cell (eg, a fused cell, a reprogrammed cell, etc. used herein) can also be a stem cell as long as it has the above-mentioned ability. Embryonic stem cells are pluripotent stem cells derived from early embryos. Embryonic stem cells were first established in 1981 and have been applied since 1989 to the production of knockout mice. In 1998, human embryonic stem cells were established and are being used in regenerative medicine. Unlike embryonic stem cells, tissue stem cells are cells that have a relatively limited degree of separation, exist in the tissue, and have an undifferentiated intracellular structure. Tissue stem cells are poor in organelles with a high nuclear Z cytoplasm ratio. Tissue stem cells are generally individuals with pluripotency and slow cell cycle Maintains proliferative capacity over the life of the body. As used herein, stem cells can preferably be embryonic stem cells, although tissue stem cells can also be used depending on the circumstances.
[0025] 本明細書において使用される場合、用語「幹細胞」とは、幹細胞または前駆細胞を 少なくとも一定量含む組織をさすことがある。従って、幹細胞は、コラゲナーゼ処理し て脂肪組織力 採取した幹細胞 (例えば、以下の実施例において使用される脂肪由 来幹細胞など)を用いることができるがそれらに限定されない。  [0025] As used herein, the term "stem cell" may refer to a tissue containing at least a certain amount of stem cells or progenitor cells. Therefore, the stem cells can be stem cells (eg, adipose-derived stem cells used in the following examples) that have been treated with collagenase and collected adipose tissue strength, but are not limited thereto.
[0026] 細胞が由来する部位により分類すると、組織幹細胞は、例えば、皮膚系、消化器系 、骨髄系、神経系などに分けられる。皮膚系の組織幹細胞としては、表皮幹細胞、毛 嚢幹細胞などが挙げられる。消ィ匕器系の組織幹細胞としては、膝 (共通)幹細胞、肝 幹細胞などが挙げられる。骨髄系の組織幹細胞としては、造血幹細胞、間葉系幹細 胞などが挙げられる。神経系の組織幹細胞としては、神経幹細胞、網膜幹細胞など が挙げられる。  When classified according to the site from which the cells are derived, tissue stem cells are classified into, for example, the skin system, digestive system, myeloid system, nervous system and the like. Skin tissue stem cells include epidermal stem cells and hair follicle stem cells. Examples of tissue stem cells of the extinct system include knee (common) stem cells and liver stem cells. Examples of myeloid tissue stem cells include hematopoietic stem cells and mesenchymal stem cells. Neural tissue stem cells include neural stem cells and retinal stem cells.
[0027] 本明細書において使用される場合、用語「間葉系幹細胞」とは、間葉に見出される 幹細胞をいう。用語「間葉系幹細胞」とは、本明細書では「MSC」と略されることがあ る。ここで、間葉とは、多細胞動物の発生各期に認められる、上皮組織間の間隙をう める星状または不規則な突起をもつ遊離細胞の集団と,それに伴う細胞間質によつ て形成される組織をいう。間葉系幹細胞は、増殖能と、骨細胞、軟骨細胞、筋細胞、 ストローマ細胞、腱細胞、脂肪細胞への分化能を有する。間葉系幹細胞は、患者か ら採取した骨髄細胞等を培養または増殖、軟骨細胞ある!/、は骨芽細胞に分化させる ために使用され、また、例えば、歯槽骨、関節症等の骨、軟骨または関節などの再建 材料として使用されており、その需要は大きい。また、間葉系幹細胞は、血液細胞、リ ンパ系細胞へも分ィ匕し得ることから、その需要がますます高まって ヽる。  [0027] As used herein, the term "mesenchymal stem cell" refers to a stem cell found in the mesenchyme. The term “mesenchymal stem cell” may be abbreviated herein as “MSC”. Here, mesenchyme is defined by a population of free cells with stellate or irregular projections that fill the gaps between epithelial tissues, which are observed at various stages of development of multicellular animals, and the associated cytoplasm. This refers to the organization that is formed. Mesenchymal stem cells have the ability to proliferate and differentiate into bone cells, chondrocytes, muscle cells, stromal cells, tendon cells, and adipocytes. Mesenchymal stem cells are used to cultivate or proliferate bone marrow cells collected from patients, have chondrocytes! /, To differentiate into osteoblasts, and, for example, bones such as alveolar bone, arthropathy, It is used as a reconstruction material for cartilage or joints, and its demand is great. In addition, since mesenchymal stem cells can be separated into blood cells and lymphoid cells, the demand for them is increasing.
[0028] 本明細書において使用される場合、用語「脂肪由来幹細胞」とは、脂肪吸引により 得られる幹細胞をいい、そしてまた、他の前駆細胞 (例えば、末梢血もしくは血管スト ローマ細胞 (vascular - stromal cell) (脂肪前駆細胞 (preadipocyte) )由来の幹 細胞)をいう。脂肪由来幹細胞は、脂肪組織に由来するか、または脂肪吸引手順に より得られた任意の多能性前駆細胞(multipotent precursor cell) )もしくは単分 化能性前駆細胞(monopotent precursor cell)の集団を意味する。これらとして は、脂肪由来血管ストローマ細胞 (脂肪前駆細胞 (preadipocyte)、脂肪由来間質 糸田胞 (adipose— derived interstitial cellよには adipose— derived stromal c ell) )、脂肪由来幹細胞、脂肪由来前駆細胞、脂肪幹細胞、内皮前駆細胞、造血幹 細胞などが挙げられる。このような幹細胞の分離方法の一部は公知であり、例えば、 非特許文献 1、特許文献 1および 2に記載される。これらの文献に記載された事項は 、本明細書において特に関連する場所が参考として援用される。本明細書において 使用される場合、用語「脂肪由来幹細胞」とは、これらの公知の分離方法によって得 られる脂肪組織由来幹細胞を含む、すべての脂肪組織由来幹細胞のことを指す。本 明細書において使用される場合、用語「前駆細胞」とは、多能性未分化細胞だけで なぐ単分化能性未分化細胞も含まれる。本明細書において使用される場合、用語「 幹細胞」とは、前駆細胞を含む。 [0028] As used herein, the term "adipose-derived stem cell" refers to a stem cell obtained by liposuction and also other progenitor cells (eg, peripheral blood or vascular stromal cells (vascular- stromal cell) (stem cell derived from preadipocyte). Adipose-derived stem cells are derived from adipose tissue or from any multipotent precursor cell) or monopotent precursor cell population obtained by a liposuction procedure. means. As these Adipose-derived vascular stromal cells (adipose—derived interstitial cells, adipose—derived stromal cells), adipose-derived stem cells, adipose-derived progenitor cells, adipose stem cells , Endothelial progenitor cells, hematopoietic stem cells, and the like. Some of such stem cell separation methods are known, and are described, for example, in Non-Patent Document 1, Patent Documents 1 and 2. The matters described in these documents are hereby incorporated by reference in particular in the present specification. As used herein, the term “adipose-derived stem cells” refers to all adipose tissue-derived stem cells including adipose tissue-derived stem cells obtained by these known separation methods. As used herein, the term “progenitor cell” includes unipotent undifferentiated cells as well as pluripotent undifferentiated cells. As used herein, the term “stem cell” includes progenitor cells.
[0029] 脂肪吸引で採取される吸引物は、吸引瓶の中で二層に分離される。吸引物の上層 は、浮遊する脂肪部分 (吸引脂肪 (lipoaspimte) )から成り、下層は液体部分 (吸引 廃揿 (liquid— aspirateま 7こは liposcution— aspirate fluid) )力ら構成 れる。本 明細書において使用される場合、用語「PLA (吸引脂肪由来細胞 (processed lipo aspirate cell) )」とは、脂肪吸引による吸引物の吸引脂肪より得られる前駆細胞を いう。本明細書において使用される場合、用語「LAF細胞(吸引廃液細胞 (liquid— aspirate [0029] Aspirated material collected by liposuction is separated into two layers in a suction bottle. The upper layer of aspirate is composed of floating fat (lipoaspimte) and the lower layer is composed of liquid (liquid-aspirate or liposcution-aspirate fluid) forces. As used herein, the term “PLA (processed lipo aspirate cell)” refers to a progenitor cell obtained from the aspirated fat of the aspirate by liposuction. As used herein, the term “LAF cells (liquid—aspirate
eelほたは liposcution— aspirate fluid cell) )とは、脂肪吸引による吸引物の 液体部分に由来する前駆細胞をいう。脂肪由来幹細胞は、 PLA細胞および LAF細 胞を含む。  eel is a liposcution—aspirate fluid cell)), which is a progenitor cell derived from the liquid part of the aspirate. Adipose-derived stem cells include PLA cells and LAF cells.
[0030] 本明細書において使用される場合、用語「体細胞」は、卵子、精子などの生殖細胞 以外の細胞であり、その DNAを次世代に引き渡さない全ての細胞をいう。体細胞は 通常、多能性が限定されている力または消失している。本明細書において使用され る体細胞は、意図された処置を達成し得る限り、天然に存在するものであってもよぐ 遺伝子改変されたものであってもょ 、。  [0030] As used herein, the term "somatic cell" refers to all cells that are cells other than germ cells, such as eggs and sperm, and that do not deliver the DNA to the next generation. Somatic cells usually have power or loss of limited pluripotency. Somatic cells as used herein may be naturally occurring or genetically modified as long as the intended treatment can be achieved.
[0031] 本明細書において使用される場合、「神経幹細胞」とは、神経系に分化することが でき、かつ、自己複製能と、多能性を併せもつ細胞をいう。神経幹細胞は、神経幹細 胞の細胞マーカー、自己複製能および多能性を確認することなどにより同定すること ができる。 [0031] As used herein, "neural stem cell" refers to a cell that can differentiate into the nervous system and has both self-renewal ability and pluripotency. Neural stem cells It can be identified by confirming cell markers, self-renewal ability, and pluripotency of vesicles.
[0032] 本明細書にぉ 、て使用される場合、神経幹細胞の「自己複製能」とは、分裂を繰り 返して自己と同じ細胞集団を生成する能力を 、う)。神経幹細胞の「自己複製能」は 、例えば、ニューロスフェア法により確認することができる。ニューロスフェア法は、 Re ynoldsら、 J Neurosci 1992 ; 12 :4565— 4574および Science 1992 ; 255 : 17 07— 1710によって、もともと報告された方法である。ニューロスフェア法は、胚または 成体中枢神経系から神経幹細胞を単離するために最も頻繁に使用される方法のうち の 1つである。ニューロスフェア法の一例としては、増殖因子として上皮増殖因子 (E GF)および塩基性線維芽細胞増殖因子(basic— FGF、 b— FGF)の存在下で、神 経幹細胞を浮遊状態で培養し、単一細胞力 なる-ユーロスフェアを形成させる工程 、形成された-ユーロスフェアを解離させて、解離した細胞を新しい培地に播種する 工程、およびこの解離した細胞が再び-ユーロスフェアを形成する力否かを確認する 工程を包含する方法がある。この方法では、解離した細胞が再び-ユーロスフェアを 形成した場合に、神経幹細胞と判定される。  [0032] As used herein, the "self-renewal ability" of a neural stem cell refers to the ability to repeat division and generate the same cell population as self). The “self-renewal ability” of a neural stem cell can be confirmed by, for example, the neurosphere method. The neurosphere method is the method originally reported by Reynolds et al., J Neurosci 1992; 12: 4565-4574 and Science 1992; 255: 17 07-1710. The neurosphere method is one of the most frequently used methods for isolating neural stem cells from the embryonic or adult central nervous system. As an example of the neurosphere method, neural stem cells are cultured in a floating state in the presence of epidermal growth factor (E GF) and basic fibroblast growth factor (basic-FGF, b-FGF) as growth factors. Single cell force-forming the eurosphere, dissociating the formed eurosphere, seeding the dissociated cell in a new medium, and the ability of the dissociated cell to form the eurosphere again There is a method including a step of confirming whether or not. In this method, a dissociated cell forms a neurosphere again when it forms a eurosphere.
[0033] 本明細書において使用される場合、神経系細胞について、「多能性」または「多分 化能」とは、神経細胞、グリア系細胞 (ァストロサイト、オリゴデンドロサイト等)の神経 系の複数の細胞へ分ィ匕する能力をいう。神経幹細胞の多能性は、例えば、ニューロ スフエアを神経系の細胞への分ィ匕条件下に置いた場合に、神経細胞、グリア系細胞 (ァストロサイト、オリゴデンドロサイト等)の神経系へと分ィ匕した場合、その細胞は多 能性を有すると判定する。好ましくは、多能性は、複数の神経系の細胞へ分化する 能力を有することをいうが、これに限定されない。神経系の細胞への分ィ匕条件として は、例えば、神経系への分ィ匕培地 (例えば、神経誘導培地(50ml)中、 DMEMZF 12 :49ml、神経誘導因子 (例えば、 B27 (GIBCO) ): lml、抗生物質を含み得る。 ) における 1週間の培養 (2日おきに培地の半量を交換しながら培養する)が挙げられ る力 これらに限定されない。グリア系細胞への分ィ匕条件としては、例えば、グリア誘 導培地(培地 50ml中、 DMEMZF12 :45ml、 FBS : 5ml、抗生物質を含み得る。 ) における 1週間の培養(2日おきに半量の培地を交換する。)が挙げられる力 これら に限定されない。本明細書において使用され得る神経栄養因子は、神経の誘導を 補助または促進する作用を有しさえすればどのようなものであってもよい。 1つの実施 形態において、神経誘導因子は、ピオチン、 L—カル-チン、コルチコステロン、エタ ノールァミン、 D ( + )—ガラクトース、ダルタチオン (還元型)、リノール酸、リノレン酸、 プロゲステロン、レチュルアセテート、セレン、トリョードサイロ-ン(T3)、 DL— aート コフエロール、 DL— a—トコフエロールアセテート、アルブミン(ゥシ)、カタラーゼ、ィ ンスリン、スーパーォキシドジスムターゼ、トランスフェリンを含む。神経系の多能性細 胞としては、例えば、神経幹細胞、神経堤細胞、神経前駆細胞等を挙げることができ る。 [0033] As used herein, with respect to nervous system cells, "pluripotency" or "multipotency" refers to the nervous system of nerve cells and glial cells (astrocytes, oligodendrocytes, etc.). The ability to divide into multiple cells. The pluripotency of neural stem cells is, for example, that when neurospheres are placed under the condition of dividing them into cells of the nervous system, they enter the nervous system of neurons and glial cells (astrocytes, oligodendrocytes, etc.). If it is determined that the cell is pluripotent. Preferably, pluripotency refers to having the ability to differentiate into cells of multiple nervous systems, but is not limited thereto. Examples of conditions for separating cells of the nervous system include, for example, a medium for separating the nervous system (for example, DMEMZF 12:49 ml in a nerve induction medium (50 ml), a nerve inducer (for example, B27 (GIBCO)) : Lml, which may contain antibiotics) Powers including, but not limited to, 1 week of culture (culture with half the medium every 2 days). As the conditions for separating cells into glial cells, for example, a glial induction medium (50 ml of medium, DMEMZF12: 45 ml, FBS: 5 ml, may contain antibiotics) is cultured for 1 week (half volume every 2 days). Replace the medium.) It is not limited to. The neurotrophic factor that can be used herein may be any as long as it has an action of assisting or promoting nerve induction. In one embodiment, the neuroinductive factor is piotin, L-carthine, corticosterone, ethanolamine, D (+)-galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retort acetate , Selenium, tolyothyrone (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin. Examples of neural pluripotent cells include neural stem cells, neural crest cells, neural progenitor cells, and the like.
[0034] 神経幹細胞は、神経幹細胞マーカーのみを用いても同定することができる。  [0034] Neural stem cells can also be identified using only neural stem cell markers.
[0035] 本明細書において使用される場合、「神経幹細胞マーカー」とは、神経幹細胞にお V、て、その局在または発現が神経幹細胞を同定するにお 、て補助となるものを 、う。 好ましくは、その局在または発現(例えば、 Nestin (NM006617(t ) )、Musashi - 1 (NM002442 (ヒ卜))、 CD133 (NM0006017 (ヒ卜))、 notchl (NM017617 ( ヒト))ゝ Hes l (NM005524 (ヒト))ゝ Mashl (NM004316 (ヒト))ゝ Neurogenin (N M006161 (ヒト))、 Pax6 (NM001604 (ヒト))、 CD15 (NM002033 (ヒト))、 PDG FR (NM002609 (ヒト)の局在または発現)によって神経幹細胞であることが同定で きるものをいう。このように、神経幹細胞の細胞マーカーとしては、例えば、 Nestin (N M006617 (ヒ卜))、 Musashi— 1 (NM002442 (ヒ卜))、 CD133 (NM0006017 (ヒ ト) )、 notch 1 (NM017617 (ヒト) )、 Hesl (NM005524 (ヒト) )、 Mashl (NM004 316 (ヒ卜))、 Neurogenin (NM006161 (ヒ卜))、 Pax6 (NM001604 (ヒ卜))、 CD1 5 (NM002033 (ヒト) )、 PDGFR (NM002609 (ヒト)などを挙げることができるがそ れらに限定されない。 [0035] As used herein, a "neural stem cell marker" refers to a neural stem cell that assists in identifying a neural stem cell by its localization or expression. . Preferably, its localization or expression (for example, Nestin (NM006617 (t)), Musashi-1 (NM002442 (human)), CD133 (NM0006017 (human)), notchl (NM017617 (human))) Hes l ( NM005524 (human)) ゝ Mashl (NM004316 (human)) ゝ Neurogenin (N M006161 (human)), Pax6 (NM001604 (human)), CD15 (NM002033 (human)), PDG FR (NM002609 (human)) In this way, neural stem cell markers include, for example, Nestin (N M006617 (Hin)), Musashi-1 (NM002442 (Hin))) , CD133 (NM0006017 (human)), notch 1 (NM017617 (human)), Hesl (NM005524 (human)), Mashl (NM004 316 (human)), Neurogenin (NM006161 (human)), Pax6 (NM001604 (human))卜)), CD15 (NM002033 (human)), PDGFR (NM002609 (human)) and the like, but are not limited thereto.
[0036] これらの神経幹細胞マーカーによる判定は、当該分野において公知の方法 (例え ば、 RT— PCR、ノーザンブロット、ウェスタンブロット、免疫染色、 FACSなど)を用い て行うことができる。ここで、発現しているかしていないかは、 RT— PCR、ノーザンブ ロット、ウェスタンブロット、免疫染色、 FACSなどによって、適宜選択することができる [0037] 本明細書において、「神経堤細胞(neural crest cell:NCC)」は、独自の遊走能 および少なくとも 1つの神経堤細胞の細胞マーカーの発現力 なる群より選択される 少なくとも 1つの特徴を有する細胞をいう。好ましくは、神経堤細胞は、独自の遊走能 および少なくとも 1つの神経堤細胞の細胞マーカーの発現の両方の特徴を確認する ことによって同定される。この独特の遊走能は、胚の頭部領域に移植し、その後前頭 鼻隆起、上顎隆起、鰓弓の間充織に遊走することを観察することによって確認できる 。神経堤細胞の細胞マーカーとしては、例えば、 CRABP1 (NM004378 (ヒト))、 A P2 (NM002097 (ヒ卜))、 Slug (NM003068 (ヒ卜))、 SoxlO (NM006941 (ヒ卜))、 Snail (NM005985 (ヒ卜))、 Twist (NM000474 (ヒ卜))、 Pax3 (NM000438 (ヒ卜) )、 Pax7 (NM002584 (ヒト) )、 HNKl (NM004854 (ヒト) )、 p75NTR (NM0025 07 (ヒト))、 TRP2 (NM006267 (ヒト))、 Wntl (NM005430 (ヒト))、 PO (NM002 723 (ヒト) )、 tPA (NM000930 (ヒト) )などを挙げることができるがこれらに限定され ない。この神経堤細胞は、神経幹細胞マーカーを少なくとも 1つ発現することを観察 することによつても確認することができる。例えば、神経幹細胞マーカーは、 Nestin ( NM006617 (ヒ卜))、 Musashi— 1 (NM002442 (ヒ卜))、 CD133 (NM0006017 ( ヒト) )、 notch 1 (NM017617 (ヒト) )、 Hesl (NM005524 (ヒト) )、 Mashl (NM00 4316 (ヒ卜))、Neurogenin(NM006161 (ヒ卜))、Pax6 (NM001604 (ヒ卜))、 CD 15 (NM002033 (ヒト) )、 PDGFR (NM002609 (ヒト) )などが挙げられるがこれらに 限定されない。好ましくは、使用され得る神経幹細胞マーカーは、 Nestinおよび Mu sashi— 1を含む。 [0036] Determination with these neural stem cell markers can be performed using methods known in the art (eg, RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.). Here, whether it is expressed or not can be appropriately selected by RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc. [0037] In the present specification, "neural crest cell (NCC)" has at least one characteristic selected from the group consisting of a unique migratory ability and the expression of a cell marker of at least one neural crest cell. Cell. Preferably, neural crest cells are identified by confirming characteristics of both unique migration ability and expression of cell markers of at least one neural crest cell. This unique migratory ability can be confirmed by transplanting into the head region of the embryo and then observing that it migrates to the frontal nasal protuberance, maxillary protuberance, and arch mesenchyme. As cell markers of neural crest cells, for example, CRABP1 (NM004378 (human)), A P2 (NM002097 (chicken)), Slug (NM003068 (chicken)), SoxlO (NM006941 (chicken)), Snail (NM005985) (Chicken)), Twist (NM000474 (chicken)), Pax3 (NM000438 (chicken)), Pax7 (NM002584 (human)), HNKl (NM004854 (human)), p75NTR (NM0025 07 (human)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002 723 (human)), tPA (NM000930 (human)), and the like, but are not limited thereto. This neural crest cell can also be confirmed by observing that at least one neural stem cell marker is expressed. For example, neural stem cell markers are Nestin (NM006617 (Human)), Musashi-1 (NM002442 (Human)), CD133 (NM0006017 (Human)), Notch 1 (NM017617 (Human)), Hesl (NM005524 (Human)) ), Mashl (NM00 4316 (chicken)), Neurogenin (NM006161 (chicken)), Pax6 (NM001604 (chicken)), CD 15 (NM002033 (human)), PDGFR (NM002609 (human)), etc. However, it is not limited to these. Preferably, neural stem cell markers that can be used include Nestin and Musashi-1.
[0038] 神経堤細胞は、胎性初期に神経管辺縁 (神経堤)より生じ、胚内を広く遊走しつつ 、きわめて広範かつ特異的なレパートリーに分ィ匕する細胞群 (第 4の胚葉)として同定 された。神経堤細胞は、例えば、後根神経節細胞、自律神経節細胞、副腎髄質クロ ム親和性細胞、シュワン細胞、外皮色素細胞などに分化し得る。神経堤細胞は、神 経冠細胞とも称される。神経堤細胞は、自己複製能と多分化能とを有し、神経幹細 胞と非常に類似している。  [0038] Neural crest cells originate from the edge of the neural tube (nerve crest) in the early fetal period and migrate widely in the embryo, and are divided into an extremely wide and specific repertoire (fourth germ layer). ) Was identified. Neural crest cells can differentiate into, for example, dorsal root ganglion cells, autonomic ganglion cells, adrenal medulla chromaffin cells, Schwann cells, and outer pigment cells. Neural crest cells are also called neural crest cells. Neural crest cells have self-renewal and pluripotency and are very similar to neural stem cells.
[0039] 好ま 、実施形態では、本発明の神経堤細胞は、脂肪細胞マーカーの発現が脂 肪由来幹細胞のものの発現レベルよりも低 、と 、う特徴を有し得る。本方法にお!、て 用いられる脂肪細胞マーカーは、脂肪細胞を判定できるものであれば、どのようなも のであってもよいが、好ましくは、 Leptin(NM000230 (ヒト))が使用され得る力 こ れに限定されない。従って、 Leptinの発現が脂肪由来幹細胞のものの発現レベルよ りも低いという特徴を有することが好ましい。本発明の神経堤細胞は、 Nestinおよび Musashi— 1を発現し、かつ、脂肪細胞マーカーである Leptinの発現が脂肪由来幹 細胞のものの発現レベルよりも低いことを特徴とし得る。 [0039] Preferably, in an embodiment, the neural crest cell of the present invention may have a characteristic that expression of an adipocyte marker is lower than that of a fat-derived stem cell. This method! The adipocyte marker used may be any as long as it can determine adipocytes, but is preferably not limited to the force with which Leptin (NM000230 (human)) can be used. Accordingly, it is preferable that Leptin expression is lower than that of adipose-derived stem cells. The neural crest cells of the present invention can be characterized by expressing Nestin and Musashi-1 and expressing Leptin, which is an adipocyte marker, lower than the expression level of adipose-derived stem cells.
[0040] 本明細書において使用される場合「神経堤細胞マーカー」とは、神経堤細胞にお V、て、その局在または発現が神経堤細胞を同定するにお 、て補助となるものを 、う。 好ましくは、その局在または発現(例えば、 CRABP1 (NM004378 (ヒト))、 AP2 (N M002097 (ヒ卜))、 Slug (NM003068 (ヒ卜))、 SoxlO (NM006941 (ヒ卜))、 Snail (NM005985 (ヒト) )、 Twist (NM000474 (ヒト) )、 Pax3 (NM000438 (ヒト) )、 Pa x7 (NM002584 (ヒ卜) )、 HNKl (NM004854 (ヒ卜) )、 p75NTR (NM002507 (ヒ ト))、 TRP2 (NM006267 (ヒト))、 Wntl (NM005430 (ヒト))、 PO (NM002723 ( ヒト);)、 tPA (NM000930 (ヒト))によって神経堤細胞であることが同定できるものを いう。このように、神経堤細胞の細胞マーカーとしては、例えば、 CRABP1 (NM004 378 (ヒ卜))、 AP2 (NM002097 (ヒ卜))、 Slug (NM003068 (ヒ卜))、 SoxlO (NM0 06941 (ヒ卜))、 Snail (NM005985 (ヒ卜))、 Twist (NM000474 (ヒ卜))、 Pax3 (N M000438 (ヒ卜))、 Pax7 (NM002584 (ヒ卜))、 HNKl (NM004854 (ヒ卜))、 p75 NTR (NM002507 (ヒト) )、 TRP2 (NM006267 (ヒト) )、 Wntl (NM005430 (ヒト) )、 PO (NM002723 (ヒト) )、 tPA (NM000930 (ヒト) )などを挙げることができるがそ れらに限定されない。 [0040] As used herein, a "neural crest cell marker" is a neural crest cell that has V, and its localization or expression assists in identifying the neural crest cell. Uh. Preferably, its localization or expression (eg, CRABP1 (NM004378 (human)), AP2 (N M002097 (human)), Slug (NM003068 (human)), SoxlO (NM006941 (human)), Snail (NM005985 (Human)), Twist (NM000474 (Human)), Pax3 (NM000438 (Human)), Pa x7 (NM002584 (Human)), HNKl (NM004854 (Human)), p75NTR (NM002507 (Human)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002723 (human);), tPA (NM000930 (human)), which can be identified as neural crest cells. Examples of cell markers for crest cells include CRABP1 (NM004 378 (Hin)), AP2 (NM002097 (Hin)), Slug (NM003068 (Hin)), SoxlO (NM0 06941 (Hin)), Snail ( NM005985 (chicken)), Twist (NM000474 (chicken)), Pax3 (N M000438 (chicken)), Pax7 (NM002584 (chicken)), HNKl (NM004854 (chicken)), p75 NTR (NM002507 (human) )), TR P2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002723 (human)), tPA (NM000930 (human)) and the like can be mentioned, but not limited thereto.
[0041] これらの神経堤細胞マーカー、神経幹細胞マーカーまたは脂肪細胞マーカーによ る判定は、当該分野において公知の方法 (例えば、 RT— PCR、ノーザンブロット、ゥ エスタンブロット、免疫染色、 FACSなど)を用いて行うことができる。ここで、発現して いるかまたはしていないかは、 RT—PCR、ノーザンブロット、ウェスタンブロット、免疫 染色、 FACSなどによって、適宜選択することができる。  [0041] Determination by these neural crest cell marker, neural stem cell marker or adipocyte marker is a method known in the art (for example, RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.) Can be used. Here, whether it is expressed or not can be appropriately selected by RT-PCR, Northern blot, Western blot, immunostaining, FACS and the like.
[0042] 以下に、本発明において使用され得る神経堤細胞マーカー、神経幹細胞マーカー および脂肪細胞マーカーを列挙する。 [0043] [表 1] [0042] The neural crest cell marker, neural stem cell marker, and adipocyte marker that can be used in the present invention are listed below. [0043] [Table 1]
(表 1.神経堤細胞マーカ一)
Figure imgf000022_0001
(Table 1. Neural crest cell markers)
Figure imgf000022_0001
[0044] [表 2] (表 2.神経幹細胞マーカー) [0044] [Table 2] (Table 2. Neural stem cell markers)
神経幹細胞マーカー ァクセッション番号  Neural stem cell marker accession number
Nesti n NM006617(ヒ卜) Nesti n NM006617
Musashi— 1 NM002442(ヒ卜) Musashi— 1 NM002442
CD133 NM0006017 (ヒ卜) notch! NM017617(ヒ卜) CD133 NM0006017 (Hui) notch! NM017617 (Hige)
Hes1 NM005524(ヒ卜) Hes1 NM005524
Mashl NM004316(ヒ卜) Mashl NM004316
Neur ogenin NM006161 (ヒ卜) Neur ogenin NM006161
Pax6 NM001604(ヒ卜) Pax6 NM001604 (Hui)
CD15 NM002033(ヒ卜) CD15 NM002033
PDGFR NM002609(ヒ卜) PDGFR NM002609 (Hui)
13] 13]
(表 3.脂肪細胞マーカー)  (Table 3. Adipocyte marker)
脂肪細胞マーカー ァクセッション番号  Adipocyte marker accession number
Lepti n NM000230(ヒ卜) 本明細書において使用される場合、「胚」または「胚細胞」とは、多細胞生物におけ る発生の初期の時代の状態をいう。本明細書において使用される場合、胚の「頭部 領域」とは、後脳、特に、第 2鰓弓に遊走する部位、すなわちロンボメァ 3, 4の周辺を いう。 Leptin NM000230 (Han) As used herein, “embryo” or “embryonic cell” refers to the state of an early era of development in a multicellular organism. As used herein, the “head region” of the embryo refers to the region that migrates to the hindbrain, in particular, the second arch, that is, around Lombomea 3 and 4.
本明細書において使用される場合、用語「分ィ匕 (した)細胞」とは、機能および形態 が特殊ィ匕した細胞 (例えば、筋細胞、神経細胞など)をいい、幹細胞とは異なり、多能 性はないか、またはほとんどない。分化した細胞としては、例えば、表皮細胞、脾実 質細胞、膝管細胞、肝細胞、血液細胞、心筋細胞、骨格筋細胞、骨芽細胞、骨格筋 芽細胞、神経細胞、血管内皮細胞、色素細胞、平滑筋細胞、脂肪細胞、骨細胞、軟 骨細胞などが挙げられる。本発明において用いられる場合、分ィ匕細胞は、集団また は組織の形態であってもよ!/、。 As used herein, the term “divided cell” refers to a cell with a special function and morphology (eg, muscle cell, nerve cell, etc.), and unlike stem cells, There is little or no performance. Differentiated cells include, for example, epidermal cells, spleen Stromal cells, knee duct cells, hepatocytes, blood cells, cardiomyocytes, skeletal muscle cells, osteoblasts, skeletal myoblasts, neurons, vascular endothelial cells, pigment cells, smooth muscle cells, fat cells, bone cells, soft Examples include bone cells. When used in the present invention, sperm cells may be in the form of a population or tissue! /.
[0047] 幹細胞の由来は、外胚葉、中胚葉および内胚葉に分類され得る。外胚葉由来の幹 細胞は、主に脳に存在し、神経幹細胞が含まれる。中胚葉由来の幹細胞は、主に骨 髄に存在し、血管幹細胞およびその分化細胞、造血幹細胞およびその分化細胞な らびに間葉系幹細胞およびその分ィ匕細胞などが含まれる。内胚葉由来の幹細胞は 主に臓器に存在し、肝幹細胞およびその分ィ匕細胞、脾幹細胞およびその分ィ匕細胞 などが含まれる。本明細書では、体細胞はどのような胚葉由来でもよい。好ましくは、 体細胞は、間葉系由来の細胞が使用され得る。  [0047] The stem cell origin can be classified into ectoderm, mesoderm and endoderm. Stem cells derived from ectoderm are mainly present in the brain and include neural stem cells. Stem cells derived from mesoderm are mainly present in bone marrow, and include vascular stem cells and their differentiated cells, hematopoietic stem cells and their differentiated cells, and mesenchymal stem cells and their differentiation cells. Endoderm-derived stem cells exist mainly in organs, and include hepatic stem cells and their differentiation cells, spleen stem cells and their differentiation cells. As used herein, somatic cells may be derived from any germ layer. Preferably, mesenchymal cells can be used as the somatic cells.
[0048] 本明細書において使用される場合、用語「脂肪細胞」とは、組織間に散在、あるい は疎性結合組織の一つとして毛細血管の走行に沿う集団として脂肪組織を形成する 、多量の脂肪を含む細胞をいう。脂肪細胞には、黄色脂肪細胞と褐色脂肪細胞とが あるが、本明細書では、いずれも等価に用いることができる。細胞内の脂肪はスダン I Πまたは四酸ィ匕オスミウムにより容易に検出され得る。  [0048] As used herein, the term "adipocyte" refers to interstitial tissues, or forms adipose tissue as a population along a capillary run as one of the loose connective tissues. A cell containing a large amount of fat. Adipocytes include yellow fat cells and brown fat cells, and any of these can be used equivalently in this specification. Intracellular fat can be easily detected by Sudan I or osmium tetroxide.
[0049] 本明細書において使用される場合、用語「所望の部位」とは、被検体における任意 の部位であって、処置が望まれる部位をいう。本発明では、そのような所望の部位は 、被検体における任意の臓器または組織における部位が選択され得ることが理解さ れる。  [0049] As used herein, the term "desired site" refers to any site in a subject for which treatment is desired. In the present invention, it is understood that such a desired site can be selected from any organ or tissue in the subject.
[0050] 本明細書において使用される場合、用語「組織」 (tissue)とは、多細胞生物におい て、実質的に同一の機能および Zまたは形態をもつ細胞集団をいう。通常「組織」は [0050] As used herein, the term "tissue" refers to a population of cells having substantially the same function and Z or morphology in a multicellular organism. “Organization” is usually
、同じ起源を有する細胞集団であるが、異なる起源を持つ細胞集団であっても、同一 の機能および Zまたは形態を有するのであればよい。従って、本発明の幹細胞を用However, cell populations having the same origin, but cell populations having different origins, may have the same function and Z or morphology. Therefore, the stem cell of the present invention is used.
Vヽて組織を再生する場合、 2以上の異なる起源を有する細胞集団が一つの組織を構 成し得る。通常、組織は、臓器の一部を構成する。動物の組織は,形態的、機能的ま たは発生的根拠に基づき、上皮組織、結合組織、筋肉組織、神経組織などに区別さ れる。植物組織では、組織を構成する細胞の発達段階によって分裂組織と永久組織 とに大別され、また構成細胞の種類によって単一組織と複合組織とに分けるなど、い ろいろな分類が行われている。本明細書において、任意の組織が処置される対象と して意図され得る。 When regenerating a tissue by V, cell populations having two or more different origins can constitute one tissue. Usually, tissue constitutes part of an organ. Animal tissues are classified into epithelial tissues, connective tissues, muscle tissues, and nerve tissues based on morphological, functional, or developmental basis. In plant tissue, meristem and permanent tissue depend on the stage of development of the cells that make up the tissue. Various classifications are made, such as dividing into single tissue and composite tissue according to the type of constituent cells. As used herein, any tissue can be intended as a subject to be treated.
[0051] 本発明において、臓器が対象とされる場合、そのような臓器はどのような臓器でもよ ぐまた本発明が対象とする組織または細胞は、生物のどの臓器または器官に由来 するものでもよい。本明細書において使用される場合、用語「臓器」は、生物個体の ある機能が個体内の特定の部分に局在して営まれ,かつその部分が形態的に独立 性をもっている構造体をいう。多細胞生物(例えば、動物、植物)では臓器は特定の 空間的配置をもついくつかの組織からなり、各組織は多数の細胞力もなる。そのよう な臓器としては、血管系に関連する臓器が挙げられる。 1つの実施形態では、本発明 が対象とする臓器は、皮膚、血管、角膜、腎臓、心臓、肝臓、臍帯、腸、神経、肺、胎 盤、脾臓、脳、四肢末梢、網膜などが挙げられるがそれらに限定されない。本明細書 において臓器が対象として使用される場合、どのような臓器が対象であってもよいが 、好ましくは、間葉系の組織 (例えば、脂肪、骨、靭帯など)が対象とされ得るがそれ に限定されない。  [0051] In the present invention, when an organ is a target, such an organ may be any organ, and the tissue or cell targeted by the present invention may be derived from any organ or organ of an organism. Good. As used herein, the term “organ” refers to a structure in which a function of an individual organism is localized and operates in a specific part of the individual, and that part is morphologically independent. . In multicellular organisms (eg animals, plants), an organ consists of several tissues with a specific spatial arrangement, and each tissue also has a number of cellular forces. Such organs include those related to the vascular system. In one embodiment, organs targeted by the present invention include skin, blood vessels, cornea, kidney, heart, liver, umbilical cord, intestine, nerve, lung, placenta, spleen, brain, peripheral limbs, retina, and the like. Is not limited to them. In the present specification, when an organ is used as a target, any organ may be a target, but preferably a mesenchymal tissue (for example, fat, bone, ligament, etc.) may be a target. It is not limited to that.
[0052] 本明細書において使用される場合、用語「神経堤細胞を誘導する条件」とは、神経 堤細胞が誘導できるような時間、培地、温度、湿度またはそれらの組み合わせなどを 指す。本発明では、脂肪由来幹細胞から神経堤細胞を調製することが初めて見出さ れたが、本明細書の内容を考慮すれば、このような条件は、脂肪由来幹細胞または 神経幹細胞を単独で維持するような条件と重複し得ることが理解される。したがって、 そのような条件は、適宜変更することができる。また、いったんそのような好ましい条 件が設定されると、以後、そのような条件は、同様の神経堤細胞を誘導するために用 いられ得る。本発明では、このような神経堤細胞を誘導する十分な条件は、インビトロ であってもインビボであってもェキソビボであっても使用され得る。インビボの場合は 、移植された体内の部分における条件がそのまま適用される。本発明では、幹細胞と 分化細胞とを混合した後は、すぐに移植してインビボの環境においてもよぐインビト 口で混合培養をしてもよい。自家移植の場合はェキソビボ移植とも呼ばれ得る。本発 明では、「神経堤細胞を誘導する条件」として、従来神経系の組織力 分離した幹細 胞を-ユーロスフェアに分ィ匕させる条件として知られる条件を挙げることができる。こ のような「神経系の組織力 分離した幹細胞を-ユーロスフェアに分ィ匕させる条件」が 脂肪由来幹細胞を神経堤細胞へ誘導させる条件として有効であることは本発明にお いて初めて見出された効果である。 [0052] As used herein, the term "conditions for inducing neural crest cells" refers to time, medium, temperature, humidity, or a combination thereof, etc. that allow neural crest cells to be induced. In the present invention, it has been found for the first time to prepare neural crest cells from adipose-derived stem cells. However, in view of the contents of this specification, such a condition seems to maintain adipose-derived stem cells or neural stem cells alone. It is understood that this can overlap with other conditions. Therefore, such conditions can be changed as appropriate. Also, once such favorable conditions have been established, such conditions can subsequently be used to induce similar neural crest cells. In the present invention, sufficient conditions for inducing such neural crest cells can be used in vitro, in vivo, or ex vivo. In vivo, the conditions in the transplanted body part are applied as they are. In the present invention, after the stem cells and the differentiated cells are mixed, they may be immediately transplanted and mixed and cultured in an in-vitro environment in an in vivo environment. Autotransplantation can also be called ex vivo transplantation. In the present invention, as a “condition for inducing neural crest cells”, the trunk cells that have been separated from the conventional tissue strength of the nervous system are used. A condition known as a condition for dividing the cell into -eurosphere can be mentioned. It has been found for the first time in the present invention that such “conditions for separating the stem cells of the nervous system into the eurospheres” are effective as conditions for inducing adipose-derived stem cells into neural crest cells. Effect.
[0053] 本明細書において「神経系の組織力 分離した幹細胞を-ユーロスフェアに分ィ匕さ せる条件」としては、実施例に記載されている条件(「ニューロスフェア培養培地」中で の培養)のほか、以下の条件を有するものを挙げることができる。  [0053] In the present specification, the "conditions for separating the stem cells separated from the nervous system into -eurospheres" are the conditions described in the examples (cultivation in "neurosphere culture medium"). In addition to the above, those having the following conditions can be mentioned.
[0054] a)約 1 X 104細胞 Zml〜約 1 X 106細胞 Zml [0054] a) about 1 X 10 4 cells Zml to about 1 X 10 6 cells Zml
b)上皮増殖因子 (EGF)の存在 <例えば、上皮増殖因子、塩基性線維芽細胞増 殖因子、 B27 supplement (GIBCO) >  b) Presence of epidermal growth factor (EGF) <e.g. epidermal growth factor, basic fibroblast growth factor, B27 supplement (GIBCO)>
c)イーグル基礎培地(BME)、最小必須培地(MEM)、ダルベッコ改変イーグル培 地(DMEM)または HAMF12培地、ある!/、はそれらの混合培地  c) Eagle Basal Medium (BME), Minimum Essential Medium (MEM), Dulbecco's Modified Eagle Medium (DMEM) or HAMF12 medium, there is! /, or their mixed medium
d) 2〜6日目(例えば、 4〜5日目)に培地を新しいものに交換すること  d) Replace the medium with a new one on the 2nd to 6th day (eg 4th to 5th day).
e) 6〜: LO日目(例えば、 8日目)に継代すること、  e) 6 ~: Passing on the LO day (eg, day 8),
およびこれらの組み合わせ。  And combinations of these.
[0055] この条件は、 Kanemura Y, Mori H, Kobayashi S, et al. J. Neurosci. Re s. 2002 ; 69 : 869— 879を参考にして当業者により調製することができる。  [0055] This condition can be prepared by those skilled in the art with reference to Kanemura Y, Mori H, Kobayashi S, et al. J. Neurosci. Res. 2002; 69: 869-879.
[0056] 本明細書において使用される「神経系の組織力 分離した幹細胞をニューロスフエ ァに分化させる条件」において、好ましい実施形態では、 a)細胞密度の条件は、 Ka ng KSらの条件 (非特許文献 10〜12; 10— 20細胞 Zcm2の密度)と大きく異なつ ており、蜜である。また、 c)培地条件は、 Rang KSらは、神経細胞の維持のために 開発され、特に、グルタミン酸、ァスパラギン酸、硫酸鉄が除かれている。他方、本発 明では、特に好ましい実施形態において、 DMEMZF12 (1 : 1)を使用しており、グ ルタミン酸、ァスパラギン酸、硫酸鉄を含むことがより有利であることが判明している。 この好ましい実施形態では、特に、本発明では、興奮性のアミノ酸である、グルタミン 酸またはァスパラギン酸が含まれて 、るので、分化した神経細胞の維持に使用され る Neurobasal medium™とは環境が全く異なるといえる。理論に束縛されることを 望まないが、興奮性のアミノ酸の存在は、神経細胞の維持において、エネルギーの 浪費等の悪影響を与えると考えられるが、他方、脂肪から神経系の多能性細胞 (例 えば、神経堤細胞)への誘導には好影響を与えるものと考えられる。 [0056] As used herein, in the "conditions for differentiating stem cells of the nervous system to differentiate into neurospheres", in a preferred embodiment, a) the cell density condition is the condition of Kang KS et al. Patent Documents 10 to 12; 10-20 cells (density of Zcm 2 )) and is nectar. C) Medium conditions were developed by Rang KS et al. For the maintenance of nerve cells, with the exception of glutamic acid, aspartic acid, and iron sulfate. On the other hand, in the present invention, in a particularly preferred embodiment, DMEMZF12 (1: 1) is used, and it has been found that it is more advantageous to include glutamic acid, aspartic acid, and iron sulfate. In this preferred embodiment, in particular, the present invention includes an excitatory amino acid, glutamate or aspartate, so the environment is completely different from the Neurobasal medium ™ used to maintain differentiated neurons. It's different. Although not wishing to be bound by theory, the presence of excitable amino acids is the Although it is thought to have an adverse effect such as wasting, on the other hand, it is considered to have a positive effect on the induction from fat into pluripotent cells of the nervous system (for example, neural crest cells).
本明細書において使用される場合、用語「インビボ」 (in vivo)は、生物の内部を いう。特定の文脈において、「インビボ」は、対象とする組織または臓器が配置される べき位置 (例えば、本明細書に!/、う所望の部位)を 、う。  As used herein, the term “in vivo” refers to the interior of an organism. In a particular context, “in vivo” refers to the location (eg, desired site! / Where desired) where the tissue or organ of interest is to be placed.
[0057] 本明細書において使用される場合、用語「インビトロ」 (in vitro)は、種々の研究目 的のために生体の一部分が「生体外に」(例えば、試験管内に)摘出または遊離され て 、る状態を 、う。インビボと対照をなす用語である。  [0057] As used herein, the term "in vitro" refers to the removal or release of a portion of a living organism "ex vivo" (eg, in a test tube) for various research purposes. Let's see the state. A term that contrasts with in vivo.
[0058] 本明細書において使用される場合、用語「ェキソビボ」(ex vivo)は、遺伝子導入 を行うための標的細胞を被験体より抽出し、インビトロで治療遺伝子を細胞に導入し た後に、再び同一被験体に戻す場合、一連の動作をェキソビボという。 [0058] As used herein, the term " ex vivo" refers to the extraction of a target cell for gene transfer from a subject and the introduction of a therapeutic gene into the cell in vitro, and then again. When returning to the same subject, a series of operations is called ex vivo.
本明細書において「自己移植片 (組織、細胞、臓器など)」というときは、広義には遺 伝的に同じ他個体 (例えば一卵性双生児)からの移植片 (組織、細胞、臓器など)を も含み得る。本明細書では、このような自己との表現は、被験体に由来すると交換可 能に使用される。従って、本明細書では、ある被験体に由来しないとの表現は、自己 ではない(すなわち、非自己)と同一の意味を有する。  In this specification, the term “autograft (tissue, cell, organ, etc.)” refers to a graft (tissue, cell, organ, etc.) from another genetically identical individual (eg, monozygotic twin) in a broad sense. Can also be included. As used herein, such self expression is used interchangeably when derived from a subject. Thus, as used herein, the expression not derived from a subject has the same meaning as not being self (ie, non-self).
[0059] 本明細書において「同種(同種異系)(移植片、組織、細胞、臓器など)」とは、同種 であっても遺伝的には異なる他個体力 移植されるもの(移植片、組織、細胞、臓器 など)をいう。遺伝的に異なることから、同種異系のもの (移植片、組織、細胞、臓器な ど)は、移植された個体 (レシピエント)において免疫反応を惹起し得る。そのようなも の(移植片、組織、細胞、臓器など)の例としては、親由来のもの(移植片、組織、細 胞、臓器など)などが挙げられるがそれらに限定されない。  [0059] In the present specification, "same species (allogeneic) (grafts, tissues, cells, organs, etc.)" refers to those that are transplanted to other individual forces that are genetically different even if they are the same species. Tissue, cell, organ, etc.). Because of genetic differences, allogeneic ones (grafts, tissues, cells, organs, etc.) can elicit an immune response in the transplanted individual (recipient). Examples of such (grafts, tissues, cells, organs, etc.) include, but are not limited to, those derived from the parent (grafts, tissues, cells, organs, etc.).
[0060] 本明細書にぉ 、て「異種 (移植片、組織、細胞、臓器など)」とは、異種個体から移 植されるもの (移植片、組織、細胞、臓器など)をいう。従って、例えば、マウスがレシ ピエントである場合、ヒト由来のもの(移植片、組織、細胞、臓器など)は異種 (組織、 細胞、臓器など)という。本発明では、異種でも使用可能であり、良好な移植成績が 立証されて 、ると 、う意味で注目されるべきである。  As used herein, the term “heterologous (graft, tissue, cell, organ, etc.)” refers to a transplant (graft, tissue, cell, organ, etc.) transplanted from a heterogeneous individual. Thus, for example, when a mouse is a recipient, a human-derived one (graft, tissue, cell, organ, etc.) is said to be heterogeneous (tissue, cell, organ, etc.). In the present invention, it should be noted in a sense that it can be used in different species and has demonstrated good transplantation results.
[0061] そのような神経幹細胞を単離する条件としては、それぞれ独立して、約 1 X 104細胞 Zml〜約 1 X 106細胞 Zml。(例えば、約 1 X 105細胞 Zml)の密度、増殖因子 (例 えば、 EGF、 bFGF)の添加、神経誘導因子(例えば、ピオチン、 L—カル-チン、コ ルチコステロン、エタノールァミン、 D ( + )—ガラクトース、ダルタチオン(還元型)、リノ 一ノレ酸、リノレン酸、プロゲステロン、レチニノレアセテート、セレン、トリョードサイロニン (T3)、 DL— a—トコフエロール、 DL- a—トコフエロールアセテート、アルブミン(ゥ シ)、カタラーゼ、インスリン、スーパーォキシドジスムターゼ、トランスフェリン)の添カロ 、ペニシリンおよびストレプトマイシンの添力 []、ペニシリンの添力!]、ストレプトマイシンの 添加、培地の選択(BME、 MEM、 DMEMまたは HAMF12培地、あるいはそれら の混合培地(例えば、 DMEMZHAMF12 (1: 1) )、 2〜6日間(例えば、 4〜5日間 )に 1度の培地交換、 6〜10日間(例えば、 8日間)に 1度の継代、 20°C〜40°Cの温 度 (例えば、 37°C)、炭酸ガス 5%、 80%以上の湿度 (例えば、 100%)、およびコート していないディッシュの使用力 選択される。一例として、 1 X 105細胞 Zmlの密度、 EGFの添加、 bFGFの添加、神経栄養因子(例えば、 B27 supplement (GIBCO) )の添加、ペニシリンおよびストレプトマイシンの添加、 DMEMZHAMF12 (1 : 1)の 使用、 4〜5日間に 1度の培地交換、 8日間に 1度の継代、 37°C、炭酸ガス 5%、 80 %以上の湿度、コートしていないディッシュの使用にて培養するという条件である。本 明細書において使用され得る神経栄養因子は、神経の誘導を補助または促進する 作用を有しさえすればどのようなものであってもよい。 1つの実施形態において、神経 誘導因子は、ピオチン、 L—カル-チン、コルチコステロン、エタノールァミン、 D ( + ) ガラクトース、ダルタチオン (還元型)、リノール酸、リノレン酸、プロゲステロン、レチ -ノレアセテート、セレン、トリョードサイロ-ン(T3)、 DL— a トコフェローノレ、 DL- a トコフエロールアセテート、アルブミン(ゥシ)、カタラーゼ、インスリン、スーパーォ キシドジスムターゼ、トランスフェリンを含む。 1つの実施形態において、神経誘導因 子は、 B27 supplement (GIBCO)であり得る。 [0061] The conditions for isolating such neural stem cells are independently about 1 X 10 4 cells. Zml to about 1 X 10 6 cells Zml. Density (eg, about 1 X 10 5 cells Zml), addition of growth factors (eg, EGF, bFGF), nerve inducers (eg, piotin, L-carcin, corticosterone, ethanolamine, D ( +) —Galactose, dartathione (reduced form), linolenoreic acid, linolenic acid, progesterone, retinoreacetate, selenium, triodothyronine (T3), DL—a—tocopherol, DL-a—tocopherol acetate, Addition of albumin (us), catalase, insulin, superoxide dismutase, transferrin), penicillin and streptomycin [], penicillin! ], Addition of streptomycin, media selection (BME, MEM, DMEM or HAMF12 media, or mixed media thereof (eg, DMEMZHAMF12 (1: 1)), once every 2-6 days (eg, 4-5 days) Medium change, passage once every 6-10 days (eg 8 days), temperature 20 ° C-40 ° C (eg 37 ° C), carbon dioxide 5%, humidity above 80% ( For example, 100%), and the use of uncoated dishes, such as 1 x 10 5 cell Zml density, EGF addition, bFGF addition, neurotrophic factor (eg B27 supplement (GIBCO)) ), Penicillin and streptomycin, DMEMZHAMF12 (1: 1), medium change once every 4-5 days, passage once every 8 days, 37 ° C, carbon dioxide 5%, 80% It is the condition that the culture is performed with the above humidity and the use of an uncoated dish. The trophic factor may be any as long as it has the effect of assisting or promoting nerve induction, In one embodiment, the nerve inducer may be piotin, L-carcin, corti Costerone, ethanolamine, D (+) galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retino-acetate, selenium, tolydothyrone (T3), DL—a Tocopheronole, DL- a Tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin In one embodiment, the nerve inducer may be B27 supplement (GIBCO).
[0062] 上記の条件は、神経幹細胞、神経堤細胞などを維持するために採用され得るがそ れに限定されない。 [0062] The above conditions may be employed to maintain neural stem cells, neural crest cells, and the like, but are not limited thereto.
[0063] 脂肪由来幹細胞から神経堤細胞を誘導するには、脂肪由来幹細胞から神経堤細 胞への誘導を促進する因子を含む任意の培養培地を用いることができる。そのような 培地としては、例えば、 DMEM/HAMF12 (1: 1)にヒト組換え EGF (20ngZml, PeproTech 、ヒト糸且換 basic— FGF (20ng/ ml, Kaken Pharmaceutical, Ja pan)、 2% B27 supplement (GIBCO)、 lOOUZmlペニシリンおよび 100 gZ mlストレプトマイシンをカ卩えた培地中での培養が挙げられ得るがそれらに限定されな い。この培地は、 37°C、酸素 20%、炭酸ガス 5%および 80%以上の湿度で使用され 得る。 [0063] In order to induce neural crest cells from adipose-derived stem cells, any culture medium containing a factor that promotes induction from adipose-derived stem cells to neural crest cells can be used. like that As the medium, for example, DMEM / HAMF12 (1: 1) and human recombinant EGF (20 ng Zml, PeproTech, human thread and basic-FGF (20 ng / ml, Kaken Pharmaceutical, Japan), 2% B27 supplement (GIBCO) Incubation in a medium supplemented with lOOUZml penicillin and 100 gZ ml streptomycin includes, but is not limited to, 37 ° C, oxygen 20%, carbon dioxide 5% and 80% or more. Can be used in humidity.
[0064] 本明細書において使用される場合、用語「神経堤細胞への誘導を促進する因子」 または「神経堤細胞誘導促進因子」とは、神経堤細胞への誘導を促進する因子 (例 えば、化学物質、温度など)であれば、どのような因子であってもよい。そのような因子 としては、例えば、種々の環境要因を挙げることができ、そのような因子としては、例 えば、温度、湿度、 pH、塩濃度、栄養、金属、ガス、有機溶媒、圧力、化学物質 (例 えば、ステロイド、抗生物質など)などまたはそれらの任意の組み合わせが挙げられる がそれらに限定されない。そのような因子のうち代表的なものとしては、上皮増殖因 子 (EGF)、塩基性線維芽細胞増殖因子 (basic— FGF、 b— FGF)のような増殖因 子(特に、 EGF)、内生因子としてBMP—4 (NM001202 (ヒト))、Noelinl (NM01 4279 (ヒト) )などが挙げられるがそれらに限定されな!、。  [0064] As used herein, the term "factor that promotes induction into neural crest cells" or "nerve crest cell induction promoting factor" refers to a factor that promotes induction into neural crest cells (eg, As long as it is a chemical substance, temperature, etc.). Examples of such factors include various environmental factors. Examples of such factors include temperature, humidity, pH, salt concentration, nutrition, metal, gas, organic solvent, pressure, chemical Examples include, but are not limited to, substances (eg, steroids, antibiotics, etc.) or any combination thereof. Typical examples of such factors include epidermal growth factor (EGF), growth factors such as basic fibroblast growth factor (basic—FGF, b—FGF) (especially EGF), Examples of biofactors include BMP-4 (NM001202 (human)), Noelinl (NM01 4279 (human)), but are not limited to these.
[0065] 本発明では、「神経系の組織力 分離した幹細胞をニューロスフェアに分ィ匕させる 条件」が脂肪由来幹細胞を神経堤細胞へ誘導させる条件として有効であることが初 めて見出されており、実質的に「神経堤細胞への誘導を促進する因子」または「神経 堤細胞誘導促進因子」は、「神経系の組織力 分離した幹細胞を-ユーロスフェアに 分化させる条件」に基づいて決定することができる。  [0065] In the present invention, it has been found for the first time that "conditions for separating neuronal tissue from separated stem cells into neurospheres" are effective as conditions for inducing adipose-derived stem cells to neural crest cells. The “factor that promotes induction into neural crest cells” or “factor that promotes neural crest cell induction” is essentially based on “conditions for differentiating stem cells from the nervous system to differentiate into eurospheres”. Can be determined.
[0066] 本明細書において使用される場合、用語「脂肪由来神経堤細胞混合物」とは、本 発明の神経堤細胞と脂肪幹細胞とを含む混合物をいう。本発明の混合物は、神経堤 細胞と脂肪由来肝細胞とを含んでさえいれば、どのような形態であってもよい。本発 明の混合物は、神経幹細胞をさらに含み得る。  [0066] As used herein, the term "adipose-derived neural crest cell mixture" refers to a mixture comprising the neural crest cells of the present invention and adipose stem cells. The mixture of the present invention may be in any form as long as it contains neural crest cells and fat-derived hepatocytes. The mixture of the present invention may further comprise neural stem cells.
[0067] 本明細書において使用される場合、用語「移植」とは、本発明の細胞、混合物、組 成物、医薬などを、単独で、または他の治療剤と組み合わせて体内に移入することを いう。本発明は、以下のような治療部位 (例えば、骨など)への導入方法,導入形態 および導入量が使用され得る:本発明の医薬などを障害部位へ直接注入し、貼付後 に縫合し、挿入する等の方法があげられる。本発明の脂肪由来幹細胞と、分化細胞 との組み合わせは、例えば、混合物として同時に、別々であるが同時にもしくは並行 して;または逐次的にかのいずれかで投与され得る。これは、組み合わされた薬剤が 、治療混合物としてともに投与される提示を含み、そして糸且み合わせた薬剤が、別々 であるが同時に (例えば、分ィ匕促進因子など)投与される手順もまた含む。「組み合わ せ」投与は、第 1に与えられ、続いて第 2に与えられる化合物または薬剤のうちの 1つ を別々に投与することをさらに含む。 [0067] As used herein, the term "transplant" refers to the transfer of a cell, mixture, composition, medicament, etc. of the present invention into the body alone or in combination with other therapeutic agents. Say. The present invention relates to the following method and mode of introduction to the treatment site (for example, bone). And the amount introduced can be used: a method of directly injecting the pharmaceutical agent of the present invention into an injured site, suturing after insertion, and inserting. The combination of adipose-derived stem cells of the invention and differentiated cells can be administered, for example, either simultaneously as a mixture, separately but simultaneously or concurrently; or sequentially. This includes the presentation that the combined drugs are administered together as a therapeutic mixture, and the procedure in which the combined drugs are administered separately but at the same time (for example, a protein enhancer) is also included. Including. “Combination” administration further includes the separate administration of one of the compounds or agents given first, followed by the second.
[0068] 本明細書において使用される場合、用語「自己」または「自家」とは、ある個体につ いていうとき、その個体に由来する個体またはその一部(例えば、細胞、糸且織、臓器 など)をいう。本明細書において使用される場合、用語「自己」または「自家」というとき は、広義には遺伝的に同じ他個体 (例えば一卵性双生児)からの移植片をも含み得 る。 [0068] As used herein, the term "self" or "self" refers to an individual, or an individual derived from the individual or a part thereof (eg, cell, thread and fabric, Organ). As used herein, the term “self” or “self” can broadly include grafts from other genetically identical individuals (eg, identical twins).
[0069] 本明細書において使用される場合、用語「同種」(同種異系)とは、同種であっても 遺伝的には異なる他個体力 移植される個体またはその一部(例えば、細胞、組織、 臓器など)をいう。同種異系の個体は、遺伝的に異なることから、同種異系のものは、 移植された個体 (レシピエント)において免疫反応を惹起し得る。そのような細胞など の例としては、親由来の細胞などが挙げられるがそれらに限定されない。  [0069] As used herein, the term "homologous" (homologous) refers to another individual force that is homogenous but genetically different (eg, cell, Tissue, organ, etc.). Because allogeneic individuals are genetically different, allogeneic individuals can elicit an immune response in the transplanted individual (recipient). Examples of such cells include, but are not limited to, parent-derived cells.
[0070] 本明細書において使用される場合、用語「異種」とは、異種個体から移植されるもの をいう。従って、例えば、ヒトがレシピエントである場合、ブタからの移植物は異種移植 物と ヽつ。  [0070] As used herein, the term "heterologous" refers to something that is transplanted from a heterologous individual. Thus, for example, if a human is the recipient, a transplant from a pig is a xenotransplant.
[0071] 本明細書において使用される場合、用語「レシピエント」(受容者)とは、移植される 細胞などを受け取る個体といい、「宿主」とも呼ばれる。これに対し、移植される細胞 などを提供する個体は、「ドナー」(供与者)という。レシピエントとドナーとは同じであ つても異なっていてもよい。  [0071] As used herein, the term "recipient" (recipient) refers to an individual that receives cells to be transplanted and the like, and is also referred to as "host". In contrast, an individual that provides cells to be transplanted is called a “donor”. The recipient and donor can be the same or different.
[0072] 本発明で使用される細胞は、同系由来(自己(自家)由来)でも、同種異系由来 (他 個体 (他家)由来)でも、異種由来でもよい。拒絶反応が考えられることから、自己由 来の細胞が好ましいが、拒絶反応が問題でない場合、同種異系由来であってもよい [0073] 本明細書において「神経系疾患、障害または状態」とは、神経系の細胞が関与する 任意の疾患、障害または異常状態をいう。 [0072] The cells used in the present invention may be derived from the same line (derived from the self (autologous)), from the same lineage (derived from another individual (other house)), or from a different species. Self-derived cells are preferred because of possible rejection, but may be from allogeneic if rejection is not a problem [0073] As used herein, "nervous disease, disorder or condition" refers to any disease, disorder or abnormal condition involving cells of the nervous system.
[0074] 本明細書において使用される場合、用語「診断、予防、処置または予後上有効な 量」とは、それぞれ、診断、予防、処置 (または治療)または予後において、医療上有 効であると認められる程度の量をいう。このような量は、当該分野において周知の技 法を用いて当業者が種々のパラメータを参酌しながら決定することができる。  [0074] As used herein, the term "diagnosis, prevention, treatment or prognostic effective amount" is medically effective in diagnosis, prevention, treatment (or therapy) or prognosis, respectively. This is the amount that can be recognized. Such an amount can be determined by a person skilled in the art using techniques well known in the art, taking into account various parameters.
[0075] 本発明が対象とする動物は、脂肪細胞を有する動物であれば、どのような動物 (例 えば、メタラウナギ類、ャッメゥナギ類、軟骨魚類、硬骨魚類、両生類、爬虫類、鳥類 、哺乳動物など)であってもよい。好ましくは、そのような動物は、哺乳動物(例えば、 単孔類、有袋類、貧歯類、皮翼類、翼手類、食肉類、食虫類、長鼻類、奇蹄類、偶 蹄類、管歯類、有鱗類、海牛類、クジラ目、霊長類、齧歯類、ゥサギ目など)であり得 る。例示的な被験体としては、例えば、ゥシ、ブタ、ゥマ、ニヮトリ、ネコ、ィヌなどの動 物が挙げられるがそれらに限定されない。さらに好ましくは、霊長類 (例えば、チンパ ンジ一、二ホンザル、ヒトなど)が用いられる。最も好ましくはヒトが用いられる。  [0075] As long as the animal targeted by the present invention is an animal having adipocytes, any animal (for example, metal eels, shark eels, cartilaginous fish, teleosts, amphibians, reptiles, birds, mammals, etc.) ). Preferably, such animals are mammals (eg, single pores, marsupials, rodents, wings, wings, carnivores, carnivores, long noses, odd hoofed animals, even hoofs, even Hoofs, rodents, scales, sea cattle, cetaceans, primates, rodents, maggots). Exemplary subjects include, but are not limited to, animals such as rabbits, pigs, horses, chickens, cats and dogs. More preferably, a primate (for example, a chimpanzee, a second monkey, a human, etc.) is used. Most preferably, a human is used.
[0076] 本発明が医薬として使用される場合、そのような医薬は、薬学的に受容可能なキヤ リアなどをさらに含み得る。本発明の医薬において、当該分野において公知の任意 の薬学的に受容可能なキャリアが使用され得る。  [0076] When the present invention is used as a medicament, such medicament may further comprise a pharmaceutically acceptable carrier and the like. In the medicament of the present invention, any pharmaceutically acceptable carrier known in the art can be used.
[0077] 適切な処方材料または薬学的に受容可能なキャリアとしては、抗酸化剤、保存剤、 着色料、風味料、希釈剤、乳化剤、懸濁化剤、溶媒、フィラー、増量剤、緩衝剤、送 達ビヒクルおよび Zまたは薬学的アジュバントが挙げられるがそれらに限定されない 。代表的には、本発明の医薬は、本発明の細胞および他の有効成分を、少なくとも 1 つの生理的に受容可能なキャリア、賦形剤または希釈剤とともに含む組成物の形態 で投与される。例えば、適切なビヒクルは、注射溶液、生理的溶液、または人工脳脊 髄液であり得、これらには、非経口送達のための組成物に一般的に使用される他の 物質を補充することが可能である。  [0077] Suitable formulation materials or pharmaceutically acceptable carriers include antioxidants, preservatives, colorants, flavors, diluents, emulsifiers, suspending agents, solvents, fillers, bulking agents, buffering agents. Including, but not limited to, delivery vehicles and Z or pharmaceutical adjuvants. Typically, the medicament of the present invention is administered in the form of a composition comprising the cells of the present invention and other active ingredients together with at least one physiologically acceptable carrier, excipient or diluent. For example, a suitable vehicle may be an injection solution, a physiological solution, or an artificial cerebrospinal fluid that is supplemented with other substances commonly used in compositions for parenteral delivery. Is possible.
[0078] 本明細書で使用される受容可能なキャリア、賦形剤または安定化剤は、好ましくは 、レシピエントに対して非毒性であり、そして好ましくは、使用される投薬量および濃 度において不活性であり、好ましくは、例えば、リン酸塩、クェン酸塩、または他の有 機酸;ァスコルビン酸、 OC トコフエロール;低分子量ポリペプチド;タンパク質(例え ば、血清アルブミン、ゼラチンまたは免疫グロブリン);親水性ポリマー(例えば、ポリビ -ルピロリドン);アミノ酸(例えば、グリシン、グルタミン、ァスパラギン、アルギニンまた はリジン);モノサッカリド、ジサッカリドおよび他の炭水化物(グルコース、マンノース、 またはデキストリン);キレート剤(例えば、 EDTA);糖アルコール (例えば、マン-トー ルまたはソルビトール);塩形成対イオン (例えば、ナトリウム);ならびに Zあるいは非 イオン性表面活性化剤(例えば、 Tween、プル口ニック (pluronic)またはポリエチレ ングリコール (PEG) )などが挙げられる。 [0078] Acceptable carriers, excipients or stabilizers used herein are preferably non-toxic to the recipient, and preferably the dosage and concentration used. Inactive, preferably, for example, phosphate, citrate, or other organic acids; ascorbic acid, OC tocopherol; low molecular weight polypeptides; proteins (eg, serum albumin, gelatin or immunoglobulins) Hydrophilic polymers (eg polyvinylpyrrolidone); amino acids (eg glycine, glutamine, asparagine, arginine or lysine); monosaccharides, disaccharides and other carbohydrates (glucose, mannose or dextrin); chelating agents ( Eg, EDTA); sugar alcohols (eg, mantol or sorbitol); salt-forming counterions (eg, sodium); and Z or non-ionic surfactants (eg, Tween, pluronic or Polyethylene glycol (PEG)) I can get lost.
[0079] 例示の適切なキャリアとしては、中性緩衝化生理食塩水、または血清アルブミンと 混合された生理食塩水が挙げられる。好ましくは、その生成物は、適切な賦形剤 (例 えば、スクロース)を用いて凍結乾燥剤として処方される。他の標準的なキャリア、希 釈剤および賦形剤は所望に応じて含まれ得る。他の例示的な組成物は、 pH約 7. 0 —8. 5の Tris緩衝剤または pH約 4. 0— 5. 5の酢酸緩衝剤を含み、これらは、さらに 、ソルビトールまたはその適切な代替物を含み得る。  [0079] Exemplary suitable carriers include neutral buffered saline or saline mixed with serum albumin. Preferably, the product is formulated as a lyophilizer using a suitable excipient (eg, sucrose). Other standard carriers, diluents and excipients may be included as desired. Other exemplary compositions include a Tris buffer having a pH of about 7.0 to 8.5 or an acetate buffer having a pH of about 4.0 to 5.5, which further includes sorbitol or a suitable replacement thereof. Can contain things.
[0080] 以下に本発明の医薬組成物の一般的な調製法を示す。なお、動物薬組成物、医 薬部外品、水産薬組成物、食品組成物およびィヒ粧品組成物等についても公知の調 製法により製造することができることに注意されたい。  [0080] A general method for preparing the pharmaceutical composition of the present invention is shown below. It should be noted that veterinary pharmaceutical compositions, quasi-drugs, marine pharmaceutical compositions, food compositions, baboon cosmetic compositions, and the like can also be produced by known preparation methods.
[0081] 本発明の細胞などは、薬学的に受容可能なキャリアと必要に応じて配合し、例えば 、注射剤、懸濁剤、溶液剤、スプレー剤等の液状製剤として非経口的に投与すること ができる。薬学的に受容可能なキャリアの例としては、賦形剤、潤滑剤、結合剤、崩 壊剤、崩壊阻害剤、吸収促進剤、吸収剤、湿潤剤、溶剤、溶解補助剤、懸濁化剤、 等張化剤、緩衝剤、無痛化剤等が挙げられる。また、必要に応じ、防腐剤、抗酸化剤 、着色剤、甘味剤等の製剤添加物を用いることができる。また、本発明の組成物には 本発明のポリヌクレオチド、ポリペプチドなど以外の物質を配合することも可能である 。非経口の投与経路としては、静脈内、筋肉内、皮下投与、皮内投与、粘膜投与、直 腸内投与、膣内投与、局所投与、皮膚投与など等が挙げられるがそれらに限定され ない。全身投与されるとき、本発明において使用される医薬は、発熱物質を含まない 、薬学的に受容可能な水溶液の形態であり得る。そのような薬学的に受容可能な組 成物の調製について、 pH、等張性、安定性などを考慮することは、当業者の技術範 囲内である。 [0081] The cells of the present invention are blended with a pharmaceutically acceptable carrier as needed, and are administered parenterally, for example, as liquid preparations such as injections, suspensions, solutions and sprays. be able to. Examples of pharmaceutically acceptable carriers include excipients, lubricants, binders, disintegrants, disintegration inhibitors, absorption enhancers, absorbents, wetting agents, solvents, solubilizers, suspending agents. , Isotonic agents, buffering agents, soothing agents and the like. In addition, formulation additives such as preservatives, antioxidants, colorants, sweeteners and the like can be used as necessary. In addition, substances other than the polynucleotide, polypeptide and the like of the present invention can be added to the composition of the present invention. Examples of parenteral routes of administration include, but are not limited to, intravenous, intramuscular, subcutaneous, intradermal, mucosal, intrarectal, intravaginal, topical, and dermal. When administered systemically, the medicament used in the present invention does not contain pyrogens It can be in the form of a pharmaceutically acceptable aqueous solution. It is within the skill of the artisan to consider pH, isotonicity, stability, etc. for the preparation of such pharmaceutically acceptable compositions.
[0082] 液状製剤における溶剤の好適な例としては、注射溶液、アルコール、プロピレンダリ コール、マクロゴール、ゴマ油およびトウモロコシ油等が挙げられる。  [0082] Preferable examples of the solvent in the liquid preparation include an injection solution, alcohol, propylene dallicol, macrogol, sesame oil, corn oil and the like.
[0083] 液状製剤における溶解補助剤の好適な例としては、ポリエチレングリコール、プロピ レングリコール、 D マン-トール、安息香酸ベンジル、エタノール、トリスァミノメタン 、コレステロール、トリエタノールァミン、炭酸ナトリウムおよびクェン酸ナトリウム等が 挙げられるがそれらに限定されない。  [0083] Preferable examples of the solubilizer in the liquid preparation include polyethylene glycol, propylene glycol, D-mannitol, benzyl benzoate, ethanol, trisaminomethane, cholesterol, triethanolamine, sodium carbonate, and ken Examples include, but are not limited to, sodium acid.
[0084] 液状製剤における懸濁化剤の好適な例としては、例えば、ステアリルトリエタノール ァミン、ラウリル硫酸ナトリウム、ラウリルアミノプロピオン酸、レシチン、塩化ベンザルコ ユウム、塩ィ匕べンゼトニゥム、モノステアリン酸グリセリン等の界面活性剤、例えば、ポ リビニルアルコール、ポリビュルピロリドン、カルボキシメチルセルロースナトリウム、メ チノレセノレロース、ヒドロキシメチノレセノレロース、ヒドロキシェチノレセノレロース、ヒドロキシ ェチルセルロース、ヒドロキシプロピルセルロース等の親水性高分子等が挙げられる  [0084] Preferable examples of the suspending agent in the liquid preparation include, for example, stearyltriethanolamine, sodium lauryl sulfate, laurylaminopropionic acid, lecithin, benzalkonium chloride, salted benzethonium, glyceryl monostearate, etc. Surfactants such as polyvinyl alcohol, polypyrrole pyrrolidone, sodium carboxymethylcellulose, methinoresenorelose, hydroxymethinoresenorelose, hydroxyethinoresenorelose, hydroxyethylcellulose, hydroxypropylcellulose, etc. Functional polymers, etc.
[0085] 液状製剤における等張化剤の好適な例としては、塩ィ匕ナトリウム、グリセリン、 D— マン-トール等が挙げられるがそれらに限定されない。 [0085] Preferable examples of the isotonic agent in the liquid preparation include, but are not limited to, sodium chloride salt, glycerin, D-manntol and the like.
[0086] 液状製剤における緩衝剤の好適な例としては、リン酸塩、酢酸塩、炭酸塩およびク ェン酸塩等が挙げられるがそれらに限定されない。  [0086] Preferable examples of the buffering agent in the liquid preparation include, but are not limited to, phosphate, acetate, carbonate, citrate and the like.
[0087] 液状製剤における無痛化剤の好適な例としては、ベンジルアルコール、塩化ベン ザルコ -ゥムおよび塩酸プロ力イン等が挙げられるがそれらに限定されない。 [0087] Preferable examples of the soothing agent in the liquid preparation include, but are not limited to, benzyl alcohol, benzalkonium chloride and pro-in hydrochloride.
[0088] 液状製剤における防腐剤の好適な例としては、パラォキシ安息香酸エステル類、ク ロロブタノール、ベンジルアルコール、 2—フエ-ルエチルアルコール、デヒドロ酢酸、 ソルビン酸等が挙げられるがそれらに限定されない。  [0088] Preferable examples of the preservative in the liquid preparation include, but are not limited to, paraoxybenzoates, chlorobutanol, benzyl alcohol, 2-phenylethyl alcohol, dehydroacetic acid, sorbic acid and the like. .
[0089] 液状製剤における抗酸化剤の好適な例としては、亜硫酸塩、ァスコルビン酸、 a トコフエロールおよびシスティン等が挙げられるがそれらに限定されない。  [0089] Preferable examples of the antioxidant in the liquid preparation include, but are not limited to, sulfite, ascorbic acid, a tocopherol and cysteine.
[0090] 注射剤として調製する際には、液剤および懸濁剤は殺菌され、かっ血液または他 の目的のための注射部位における溶媒と等張であることが好ましい。通常、これらは 、細菌保留フィルタ一等を用いるろ過、殺菌剤の配合または照射などによって無菌化 する。さらにこれらの処理後、凍結乾燥等の方法により固形物とし、使用直前に無菌 水または無菌の注射用希釈剤 (塩酸リドカイン水溶液、生理食塩水、ブドウ糖水溶液 、エタノールまたはこれらの混合溶液等)を添加してもよい。 [0090] When prepared as an injection, solutions and suspensions are sterilized and blood or other It is preferably isotonic with the solvent at the injection site for these purposes. Usually, these are sterilized by filtration using a bacteria retention filter or the like, blending with a bactericide, or irradiation. Furthermore, after these treatments, solidify by freeze-drying or other methods, and add sterile water or sterile diluent for injection (lidocaine hydrochloride aqueous solution, physiological saline, aqueous glucose solution, ethanol or a mixture of these) immediately before use. May be.
[0091] さらに、本発明の医薬組成物は、着色料、保存剤、香料、矯味矯臭剤、甘味料等、 ならびに他の薬剤を含んで 、てもよ 、。  [0091] Further, the pharmaceutical composition of the present invention may contain a coloring agent, a preservative, a fragrance, a flavoring agent, a sweetener and the like, and other agents.
[0092] 本発明の処置方法において使用される組成物の量は、使用目的、対象疾患 (種類 、重篤度など)、患者の年齢、体重、性別、既往歴、細胞の形態または種類などを考 慮して、当業者が容易に決定することができる。本発明の処置方法を被験体 (または 患者)に対して施す頻度もまた、使用目的、対象疾患 (種類、重篤度など)、患者の 年齢、体重、性別、既往歴、および治療経過などを考慮して、当業者が容易に決定 することができる。頻度としては、例えば、毎日 数ケ月に 1回(例えば、 1週間に 1回 1ヶ月に 1回)の投与が挙げられる。 1週間ー1ヶ月に 1回の投与を、経過を見なが ら施すことが好ましい。投与する量は、処置されるべき部位が必要とする量を見積も ること〖こよって確定することができる。  [0092] The amount of the composition used in the treatment method of the present invention depends on the purpose of use, the target disease (type, severity, etc.), the patient's age, weight, sex, medical history, cell morphology or type, etc. In view of this, it can be easily determined by those skilled in the art. The frequency with which the treatment method of the present invention is applied to a subject (or patient) also depends on the purpose of use, target disease (type, severity, etc.), patient age, weight, gender, medical history, treatment history, etc. In view of this, it can be easily determined by those skilled in the art. Examples of the frequency include administration once a few months every day (for example, once a week, once a month). It is preferable to administer once a week-once a month with the progress of the test. The amount to be administered can be determined by estimating the amount required by the site to be treated.
[0093] 本明細書において使用される場合、用語「指示書」とは、本発明の医薬を投与する 方法または診断する方法などを、本発明の医薬などの投与を行う人または投与を行 われる人、診断する人または診断される人 (例えば、医師、患者など)に対して記載し たものである。この指示書は、本発明の診断薬、医薬などを投与する適切な方法を 指示する文言が記載されている。この指示書は、本発明が実施される国の監督官庁 (例えば、 日本であれば厚生労働省、米国であれば食品医薬品局 (FDA)など)が規 定した様式に従って作成され、その監督官庁により承認を受けた旨が明記される。指 示書は、いわゆる添付文書 (package insert)であり、通常は紙媒体で提供される 力 それに限定されず、例えば、電子媒体 (例えば、インターネットで提供されるホー ムページ (ウェブサイト)、電子メールなど)のような形態でも提供され得る。  [0093] As used herein, the term "instruction" refers to a method for administering or diagnosing the medicament of the present invention, a person who administers the medicament of the present invention, or the like. It is written for a person, a person to be diagnosed, or a person to be diagnosed (eg, doctor, patient, etc.). This instruction describes a word indicating an appropriate method for administering the diagnostic agent or medicine of the present invention. These instructions are prepared according to the format prescribed by the national supervisory authority (for example, the Ministry of Health, Labor and Welfare in Japan and the Food and Drug Administration (FDA) in the United States). It will be clearly stated that it has been approved. Instructions are so-called package inserts, and are not normally limited to paper-delivered forces, such as electronic media (eg, home pages (websites) provided on the Internet, emails, etc. Etc.) can also be provided.
[0094] 本発明の方法による治療の終了の判断は、商業的に利用できるアツセィもしくは機 器使用による標準的な臨床検査の結果または意図された治療に関連する疾患 (例え ば、骨疾患、心臓疾患、神経疾患など)に特徴的な臨床症状の消滅あるいは美容状 態の回復 (例えば、視覚的な回復など)によって支持され得る。治療は、分化細胞の 欠損などに関連する疾患 (例えば、神経疾患)の再発または美容状態の損傷により 再開することができる。 [0094] The end of treatment according to the method of the present invention may be determined by the results of standard laboratory tests using commercially available accessories or instrumentation or diseases associated with the intended treatment (eg, For example, it can be supported by the disappearance of clinical symptoms characteristic of bone disease, heart disease, nerve disease, etc., or recovery of the cosmetic state (eg, visual recovery, etc.). Treatment can be resumed by a recurrence of a disease (eg, neurological disease) associated with a lack of differentiated cells or a cosmetic condition injury.
[0095] 本発明はまた、医薬組成物の 1つ以上の成分を満たした 1つ以上の容器を備える 薬学的パックまたはキットを提供する。医薬品または生物学的製品の製造、使用また は販売を規制する政府機関が定めた形式の通知が、このような容器に任意に付属し 得、この通知は、ヒトへの投与に対する製造、使用または販売に関する政府機関によ る承認を表す。このキットは、注入デバイスを備え得る。  [0095] The present invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more ingredients of a pharmaceutical composition. A notice of the form prescribed by the government that regulates the manufacture, use or sale of a medicinal product or biological product may optionally be attached to such containers, and this notice may be made, used or administered for human administration. Represents approval by a government agency regarding sales. The kit can include an infusion device.
[0096] 本発明の細胞等の毒性研究は、モデルを用いて行われ得る。例えば、毒性研究は 、以下のような適切な動物モデルにおいて行われ得る:(1)化合物がマウスに投与さ れる(未処置のコントロールマウスもまた、使用されるべきである);(2)各々の処置群 中の 1匹のマウス力も尾静脈を介して血液サンプルを周期的に得る;そして(3)上記 サンプルを、神経系の細胞等について分析する。各々の投薬レジメンについての結 果とコントロールとの比較は、毒性が存在するか否かを示す。  [0096] Toxicity studies such as cells of the present invention can be performed using a model. For example, toxicity studies can be performed in appropriate animal models such as: (1) compounds are administered to mice (untreated control mice should also be used); (2) each One mouse in the treatment group also obtains blood samples periodically via the tail vein; and (3) The samples are analyzed for cells of the nervous system and the like. Comparison of the results for each dosing regimen with controls shows whether toxicity is present.
[0097] 各々の毒性研究の終了の際に、動物を屠殺することによって、さらなる研究を行い 得る (好ましく ii、 American Veterinary Medical Association guidelines Report of the American Veterinary Medical Assoc. Panel on Eutha nasia, (1993)J. Am. Vet. Med. Assoc. 202 : 229— 249【こ従う)。次!ヽで、各 処置群からの代表的な動物が、転移、異常な病気または毒性の直接的な証拠のた めに全体的な検屍によって試験され得る。組織における全体の異常が記載され、組 織が組織学的に試験される。神経系の異常を惹起したり、体重の減少または血液成 分の減少を引き起こす医薬は、主要な臓器に対する有害作用を有する化合物と同 様に好ましくない。一般的に、有害作用が大きいほど、その化合物は好ましくない。  [0097] Further studies can be performed by sacrificing animals at the end of each toxicology study (preferably ii, American Veterinary Medical Association guidelines Report of the American Veterinary Medical Assoc. Panel on Eutha nasia, (1993) J. Am. Vet. Med. Assoc. 202: 229—249. Then, representative animals from each treatment group can be tested by global examination for direct evidence of metastasis, abnormal illness or toxicity. Overall abnormalities in the tissue are described and the tissue is examined histologically. Drugs that cause abnormalities in the nervous system or cause weight loss or blood component reduction are not preferred, as are compounds that have adverse effects on major organs. In general, the greater the adverse effect, the less preferred the compound.
[0098] (神経堤細胞の用途)  [0098] (Use of neural crest cells)
本発明の神経堤細胞は、種々の用途、例えば、神経系の処置に使用することがで きる。例えば、神経系の細胞が関与する任意の疾患、障害または異常状態などの処 置が挙げられる力 これらに限定されない。 (好ましい実施形態の説明) The neural crest cells of the present invention can be used for various purposes, for example, treatment of the nervous system. For example, forces including treatment of any disease, disorder or abnormal condition involving cells of the nervous system are not limited to these. (Description of Preferred Embodiment)
以下に本発明の好ましい実施形態を説明する。以下に提供される実施形態は、本 発明のよりよい理解のために提供されるものであり、本発明の範囲は以下の記載に 限定されるべきでない。従って、当業者は、本明細書中の記載を参酌して、本発明の 範囲内で適宜改変を行うことができることは明らかである。  Hereinafter, preferred embodiments of the present invention will be described. The embodiments provided below are provided for a better understanding of the present invention, and the scope of the present invention should not be limited to the following description. Therefore, it is obvious that those skilled in the art can make appropriate modifications within the scope of the present invention in consideration of the description in the present specification.
[0099] (神経堤細胞を調製するための方法)  [0099] (Method for preparing neural crest cells)
1つの局面において、本発明は、神経堤細胞を調製するための方法を提供する。こ の方法によって、神経堤細胞を一定量以上で提供することができる。この方法は、 A) 脂肪から幹細胞を得る工程;および B)該幹細胞を、神経堤細胞を誘導する条件に 供する工程を包含する。  In one aspect, the present invention provides a method for preparing neural crest cells. By this method, neural crest cells can be provided in a certain amount or more. This method comprises the steps of A) obtaining stem cells from fat; and B) subjecting the stem cells to conditions for inducing neural crest cells.
[0100] ここで、脂肪由来の幹細胞は、国際公開第 OOZ53795号パンフレット、同第 03Z 022988号パンフレット、同第 01Z62901号パンフレット、 Zuk, P. A.ら、 Tissue Engineering, Vol. 7, 211— 228、 2001、および Zuk, P. A.ら、 Molecular Bi ology of the Cell Vol. , 13, 4279—4295、 2002などに記載される方法また はその改変のように脂肪吸引による吸引物(吸引脂肪 (lipoaspirate) )の脂肪部分 力も分離することができる。具体的には、例えば、(1)吸引脂肪(suctioned fat)を 1リットル大の分液漏斗を用いて生理食塩水で十分に洗浄し;(2)上層に吸引脂肪、 下層に生理食塩水が十分に分離したのを確認し、下層を捨てる。肉眼で見て生理食 塩水がほぼ透明になるまでこれを繰り返し;(3)吸引脂肪と同量の 0. 075%コラゲナ ーゼ ZPBSをカ卩え、 37°Cでよく攪拌しながら 30分間インキュベートし;(4)上記の試 料に等量の 10%血清加 DMEMを加え; (5)上記の試料を 1200gで 10分間遠心分 離し;(6)得られたペレットに 0. 16M NH C1ZPBSをカ卩えて懸濁し、室温で 10分  [0100] Here, stem cells derived from adipose are disclosed in International Publication No. OOZ53795, No. 03Z 022988, No. 01Z62901, Zuk, PA et al., Tissue Engineering, Vol. 7, 211-228, 2001, And Zuk, PA, et al., Molecular Biology of the Cell Vol., 13, 4279-4295, 2002, etc., or modifications thereof, and the fat portion of aspirate (lipoaspirate) by liposuction Force can also be separated. Specifically, for example, (1) suctioned fat is thoroughly washed with physiological saline using a 1-liter separatory funnel; (2) suction fat is in the upper layer, and physiological saline is in the lower layer. Make sure it is well separated and discard the lower layer. Physiological diet with the naked eye Repeat this until the saline is almost clear; (3) Hold 0.075% collagenase ZPBS in the same amount as the aspirated fat and incubate for 30 minutes with good agitation at 37 ° C. (4) Add an equal volume of 10% serum-added DMEM to the above sample; (5) Centrifuge the above sample at 1200 g for 10 minutes; (6) Add 0.16M NH 4 C1ZPBS to the resulting pellet. Suspended and suspended for 10 minutes at room temperature
4  Four
間インキュベートし;(7)上記の試料を口径 100 mのメッシュを用いて吸引ろ過し; および (8)得られたろ過物を 1200gで 5分間遠心分離することによって分離すること ができる。ここで、調製量に応じて、上記プロトコールをスケールアップまたはスケー ルダウンすることは、当業者の技術範囲内である。  (7) The above sample can be filtered by suction using a 100 m mesh mesh; and (8) The resulting filtrate can be separated by centrifuging at 1200 g for 5 minutes. Here, it is within the technical scope of those skilled in the art to scale up or scale down the above protocol according to the preparation amount.
[0101] 他方、脂肪由来幹細胞は、例えば、以下のような脂肪吸引による吸引物の液体部 分 (液体吸引物)より単離され得る:(1)脂肪吸引による吸引物の液体部分が調製さ れる;(2)この液体部分は、細胞画分を得るために遠心分離される;(3)この細胞画 分は、密度勾配遠心分離に供され、そして細胞分離は、比重に基づいて実施される ;そして (4)細胞は、赤血球よりも低い比重を有する細胞層から回収される。吸引物 の液体部分は、生理食塩水またはリンガー注入液を用いて調製され得る。遠心分離 は、約 800 X g以下、または約 400 X g以上の速度で実施され得る。密度勾配遠心分 離は、約 370 X g〜l, 100 X gの速度で実施される。密度勾配遠心分離は、約 1. 0 76gZml〜l. 078gZmlの比重(20°C)を有する溶媒を用いて実施される。密度勾 配遠心分離において使用される溶媒は、 Ficoll™, Percoll™またはスクロースであり 得る。回収される細胞層の比重は、約 1. 050〜1. 075の範囲であり得る。細胞層は 、ピペットを用いて回収され得る。 [0101] On the other hand, adipose-derived stem cells can be isolated from, for example, a liquid part (liquid aspirate) of aspirate by liposuction as follows: (1) The liquid part of aspirate by liposuction is prepared. (2) The liquid portion is centrifuged to obtain a cell fraction; (3) The cell fraction is subjected to density gradient centrifugation, and the cell separation is performed based on specific gravity. And (4) the cells are recovered from a cell layer having a specific gravity lower than that of red blood cells. The liquid portion of the aspirate can be prepared using saline or Ringer's infusion. Centrifugation can be performed at a speed of about 800 Xg or less, or about 400 Xg or more. Density gradient centrifugation is performed at a rate of about 370 X g to l, 100 X g. Density gradient centrifugation is carried out using a solvent having a specific gravity (20 ° C.) of about 1.0 76 gZml to 1.07 gZml. The solvent used in density gradient centrifugation can be Ficoll ™, Percoll ™ or sucrose. The specific gravity of the recovered cell layer can range from about 1.050 to 1.075. The cell layer can be collected using a pipette.
[0102] 本発明において使用される脂肪由来幹細胞は、 CD13、 CD29、 CD34、 CD36、 CD44、 CD49d、 CD54、 CD58、 CD71、 CD73、 CD90、 CD105、 CD106、 CD 151、および SH3からなる群より選択される少なくとも 1つ(好ましくは、 2つ、 3つ、 . . . n個)のタンパク質を発現し得るものであり得る。さらに好ましくは、本発明において 用いられる脂肪由来幹細胞は、 CD13、 CD29、 CD34、 CD36、 CD44、 CD49d、 CD54、 CD58、 CD71、 CD73、 CD90、 CD105、 CD106、 CD151、および SH3 のすベてを発現し得るものである。  [0102] The adipose-derived stem cells used in the present invention are selected from the group consisting of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106, CD 151, and SH3. Can express at least one (preferably two, three,... N) proteins. More preferably, the adipose-derived stem cells used in the present invention express all of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106, CD151, and SH3. It is possible.
[0103] これらの CD抗原の判定は、当該分野において公知の方法 (例えば、抗体を使用す る免疫学的手法など)を用いて行うことができる。ここで、発現しているかしていないか は、免疫学的方法などによって、適宜選択することができる。  [0103] These CD antigens can be determined using a method known in the art (for example, an immunological technique using an antibody). Here, whether it is expressed or not can be appropriately selected by an immunological method or the like.
[0104] 好ましくは、本発明において用いられる脂肪由来幹細胞は、 CD3、 CD4、 CD14、 CD15、 CD16、 CD19、 CD33、 CD38、 CD56、 CD61、 CD62e、 CD62p、 CD6 9、 CD104、 CD135、および CD144の少なくとも 1つ(特に、 CD56)を発現しない。 そのような CD抗原は、分化細胞のマーカーであり、発現しないことが幹細胞の指標 であり得るからであるがそれらに限定されない。従って、好ましい実施形態では、本発 明において使用される脂肪由来幹細胞は、 CD3、 CD4、 CD14、 CD15、 CD16、 C D19、 CD33、 CD38、 CD56、 CD61、 CD62e、 CD62p、 CD69、 CD104、 CD1 35、および CD144の!、ずれも発現しな!、細胞であることが有利であり得る。 [0105] 別の実施形態では、簡便に、脂肪由来幹細胞として、 CD49dを発現し、 CD56を 発現しな 、細胞を選択してもよ ヽ。 [0104] Preferably, the adipose-derived stem cells used in the present invention are CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD135, and CD144. Does not express at least one (especially CD56). Such a CD antigen is a marker for differentiated cells, and since it can be an indicator of stem cells that it is not expressed, it is not limited thereto. Therefore, in a preferred embodiment, the adipose-derived stem cells used in the present invention are CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD1 35 , And CD144 !, no deviations expressed! It may be advantageous to be a cell. [0105] In another embodiment, cells that express CD49d and do not express CD56 may be conveniently selected as adipose-derived stem cells.
[0106] 本発明において用いられる脂肪由来幹細胞は、吸引脂肪に由来し得る。従来、吸 引脂肪は、捨てられていたが、本発明において、脂肪由来幹細胞から神経幹細胞( 好ましくは、神経堤細胞)が単離できるようになった。従って、実際に神経系疾患、障 害または状態の治療に用いることができる幹細胞の供給源として使用できることが明 らかになつた。従って、そのような吸引脂肪は、例えば、脂肪吸引による吸引物の液 体部分または脂肪部分であってもよ!/、。  [0106] The adipose-derived stem cells used in the present invention can be derived from aspirated fat. Conventionally, sucked fat has been discarded, but in the present invention, neural stem cells (preferably neural crest cells) can be isolated from fat-derived stem cells. Thus, it has become clear that it can be used as a source of stem cells that can actually be used to treat neurological diseases, disorders or conditions. Thus, such aspiration fat may be, for example, the liquid part or the fat part of the aspirate from liposuction! /.
[0107] 本発明において用いられる脂肪由来幹細胞は、分離されたものを使用しても良い 力 部分精製したものまたは完全に精製したものを使用しても良い。  [0107] As the adipose-derived stem cells used in the present invention, isolated ones may be used. Force-purified ones or completely purified ones may be used.
[0108] 好ましい実施形態では、本発明において使用される脂肪幹細胞は、現代人が不要 とする部分の脂肪 (例えば、腹部、胸部、臀部、大腿部、上腕部、顔部などの脂肪) 力も脂肪細胞より調製ができる。腹部、臀部などが好ましい。腹部、臀部などは、脂肪 がたまりやすい部位であり、除去することが所望されることが多いからである。  [0108] In a preferred embodiment, the adipose stem cells used in the present invention also have a portion of fat (for example, fat in the abdomen, chest, hips, thigh, upper arm, face, etc.) that is unnecessary for modern humans. Can be prepared from adipocytes. The abdomen, buttocks, etc. are preferred. This is because the abdomen, buttocks, etc. are sites where fat tends to accumulate and are often desired to be removed.
[0109] 本方法において、脂肪から得られた幹細胞を、従来神経系の組織力 分離した幹 細胞を-ユーロスフェアに分ィ匕させる条件として知られる条件に供することによって神 経堤細胞を調製することができる。そのような条件としては、それぞれ独立して、約 1 X 104細胞/ ml〜約 1 X 106細胞/ ml (例えば、約 1 X 105細胞/ ml)の密度、増殖 因子(例えば、 EGF、 bFGF)の添加、神経誘導因子(例えば、ピオチン、 L—カル- チン、コルチコステロン、エタノールァミン、 D ( + )—ガラタトース、グルタチオン(還元 型)、リノール酸、リノレン酸、プロゲステロン、レチュルアセテート、セレン、トリョード サイロ-ン(T3)、 DL— a—トコフエロール、 DL- a—トコフエロールアセテート、ァ ルブミン(ゥシ)、カタラーゼ、インスリン、スーパーォキシドジスムターゼ、トランスフェリ ンを含む)、ペニシリンおよびストレプトマイシン力もなる群より選択される少なくとも 1 つの添加、培地の選択(BME、 MEM、 DMEMまたは HAMF12培地、あるいはそ れらの混合培地(例えば、 DMEM/HAMF12 (1: 1) )、 2〜6日間(例えば、 4〜5 日間)に 1度の培地交換、 6〜10日間(例えば、 8日間)に 1度の継代、 20°C〜40°C の温度 (例えば、 37°C)、炭酸ガス 5%、 80%以上の湿度 (例えば、 100%)、および コートしていないディッシュの使用力も選択され得る。一例として、 1 X 105細胞/ ml の密度、 EGFの添加、 bFGFの添加、神経栄養因子(例えば、 B27 supplement ( GIBCO) )の添加、ペニシリンおよびストレプトマイシンの添加、 DMEM/HAMF1 2 (1: 1)の使用、 4〜5日間に 1度の培地交換、 8日間に 1度の継代、 37°C、炭酸ガス 5%、 80%以上の湿度、コートしていないディッシュの使用にて培養するという条件で あり得る。本明細書において使用され得る神経栄養因子は、神経の誘導を補助また は促進する作用を有しさえすればどのようなものであってもよい。 1つの実施形態に おいて、神経誘導因子は、ピオチン、 L—カル-チン、コルチコステロン、エタノール ァミン、 D ( + )—ガラクトース、ダルタチオン (還元型)、リノール酸、リノレン酸、プロゲ ステロン、レチュルアセテート、セレン、トリョードサイロニン(T3)、 DL— a—トコフエ ロール、 DL— a—トコフエロールアセテート、アルブミン(ゥシ)、カタラーゼ、インスリ ン、スーパーォキシドジスムターゼ、トランスフェリンを含む。 1つの実施形態において 、神経誘導因子は、 B27 supplement (GIBCO)であり得る。 [0109] In this method, a neural cell is prepared by subjecting a stem cell obtained from fat to a condition known as a condition for separating a stem cell separated from the tissue power of a conventional nervous system into -eurosphere. be able to. Such conditions include, independently, a density of about 1 × 10 4 cells / ml to about 1 × 10 6 cells / ml (eg, about 1 × 10 5 cells / ml), a growth factor (eg, EGF) BFGF), nerve inducer (eg, piotin, L-carcin, corticosterone, ethanolamine, D (+)-galatose, glutathione (reduced form), linoleic acid, linolenic acid, progesterone, (Including tuluacetate, selenium, tolydothyrone (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin, catalase, insulin, superoxide dismutase, transferrin), Add at least one selected from the group that also has penicillin and streptomycin activity, select medium (BME, MEM, DMEM or HAMF12 medium, or a mixed medium thereof) For example, DMEM / HAMF12 (1: 1)), medium change every 2-6 days (eg 4-5 days), passage once every 6-10 days (eg 8 days), 20 ° C-40 ° C temperature (e.g. 37 ° C), carbon dioxide 5%, humidity above 80% (e.g. 100%), and The power to use uncoated dishes can also be selected. For example, density of 1 × 10 5 cells / ml, addition of EGF, addition of bFGF, addition of neurotrophic factors (eg B27 supplement (GIBCO)), addition of penicillin and streptomycin, DMEM / HAMF1 2 (1: 1 ), Medium change once every 4-5 days, passage once every 8 days, 37 ° C, carbon dioxide gas 5%, humidity of 80% or higher, use uncoated dish It can be the condition. The neurotrophic factor that can be used herein may be any as long as it has an effect of assisting or promoting nerve induction. In one embodiment, the nerve-inducing factor is piotin, L-carthine, corticosterone, ethanolamine, D (+)-galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, Contains leturyl acetate, selenium, triodothyronine (T3), DL-a-tocopherol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin. In one embodiment, the neural inducer can be B27 supplement (GIBCO).
[0110] 上記の条件は、神経幹細胞 (例えば、神経堤細胞)を維持するために採用され得る がそれに限定されない。  [0110] The above conditions can be employed to maintain neural stem cells (eg, neural crest cells), but are not limited thereto.
[0111] 脂肪由来幹細胞から神経堤細胞を誘導するには、脂肪由来幹細胞から神経堤細 胞への誘導を促進する因子を含む任意の培養培地を用いることができる。そのような 培地としては、例えば、 DMEM/HAMF12 (1: 1)にヒト組換え EGF (20ngZml, PeproTech 、ヒト糸且換 basic— FGF (20ng/ ml, Kaken Pharmaceutical, Ja pan)、 2% B27 supplement (GIBCO)、 lOOUZmlペニシリンおよび 100 gZ mlストレプトマイシンをカ卩えた培地中での培養が挙げられ得るがそれらに限定されな い。この培地は、 37°C、酸素 20%、炭酸ガス 5%および 80%以上の湿度、で使用さ れ得る。本発明では、 EGFが誘導に重要であることが見出された。  [0111] In order to induce neural crest cells from adipose-derived stem cells, any culture medium containing a factor that promotes induction from adipose-derived stem cells to neural crest cells can be used. Examples of such medium include DMEM / HAMF12 (1: 1) and human recombinant EGF (20 ng Zml, PeproTech, human thread and basic-FGF (20 ng / ml, Kaken Pharmaceutical, Japan), 2% B27 supplement. (GIBCO), lOOUZml penicillin, and culture in a medium supplemented with 100 gZ ml streptomycin, including but not limited to: 37 ° C, 20% oxygen, 5% carbon dioxide and 80% In the present invention, it was found that EGF is important for induction.
[0112] 別の実施形態において、本方法は、幹細胞が神経堤細胞マーカー、および必要に 応じて神経幹細胞マーカーを発現しているかどうかを確認する工程をさらに包含し得 る。この神経堤細胞は、神経堤細胞マーカーを少なくとも 1つ発現する。この神経堤 細胞は、さらに、神経幹細胞マーカーを少なくとも 1つ発現し得る。例えば、神経堤細 胞の細胞マーカーとしては、例えば、 CRABP1 (NM004378 (ヒト) )、 AP2 (NM00 2097 (ヒ卜))、 Slug (NM003068 (ヒ卜))、 SoxlO (NM006941 (ヒ卜))、 Snail (NM 005985 (ヒト))、 Twist (NM000474 (ヒト))、 Pax3 (NM000438 (ヒト))、 Pax7 (N M002584 (ヒ卜))、 HNKl (NM004854 (ヒ卜))、 p75NTR (NM002507 (ヒ卜))、 TRP2 (NM006267 (ヒト))、 Wntl (NM005430 (ヒト))、 PO (NM002723 (ヒト)) 、 tPA(NM000930 (ヒト))などを挙げることができるがこれらに限定されない。例え ば、神経幹細胞マーカーは、 Nestin (NM006617 (ヒト))、 Musashi—1 (NM002 442 (ヒ卜))、 CD133 (NM0006017 (ヒ卜))、 notchl (NM017617 (ヒ卜))、 Hesl ( NM005524 (ヒ卜) )、 Mashl (NM004316 (ヒ卜) )、 Neurogenin (NM006161 (ヒ 卜))、 Pax6 (NM001604 (ヒ卜))、 CD15 (NM002033 (ヒ卜))、 PDGFR(NM0026 09 (ヒト))が挙げられるがこれらに限定されない。好ましくは、神経幹細胞マーカーは 、 Nestinおよび Musashi—lを含む。 [0112] In another embodiment, the method may further comprise the step of ascertaining whether the stem cell is expressing a neural crest cell marker, and optionally a neural stem cell marker. The neural crest cells express at least one neural crest cell marker. The neural crest cell may further express at least one neural stem cell marker. For example, cell markers of neural crest cells include, for example, CRABP1 (NM004378 (human)), AP2 (NM00 2097 (Human)), Slug (NM003068 (Human)), SoxlO (NM006941 (Human)), Snail (NM 005985 (Human)), Twist (NM000474 (Human)), Pax3 (NM000438 (Human)), Pax7 (N M002584 (chicken)), HNKl (NM004854 (chicken)), p75NTR (NM002507 (chicken)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002723 (human)) ), TPA (NM000930 (human)) and the like, but are not limited thereto. For example, neural stem cell markers include Nestin (NM006617 (human)), Musashi—1 (NM002 442 (blue)), CD133 (NM0006017 (blue)), notchl (NM017617 (blue)), Hesl (NM005524 (が)), Mashl (NM004316 (卜)), Neurogenin (NM006161 (ヒ)), Pax6 (NM001604 (卜)), CD15 (NM002033 (卜)), PDGFR (NM0026 09 (human)) Although it is mentioned, it is not limited to these. Preferably, neural stem cell markers include Nestin and Musashi-l.
[0113] 別の実施形態において、本方法は、幹細胞が脂肪細胞マーカーを発現のレベルを 確認する工程をさらに包含し得る。本方法にぉ 、て用いられる脂肪細胞マーカーは 、脂肪細胞を判定できるものであれば、どのようなものであってもよいが、好ましくは、 Leptin(NM000230 (ヒト))が使用され得る力 これに限定されない。従って、脂肪 細胞マーカーのレベルの確認は、 Leptin (NM000230 (ヒト))の減少によって確認 され得る。 1つの実施形態において、脂肪力も得られた幹細胞は、神経堤細胞を誘 導する条件に供する工程により、神経堤細胞と脂肪由来幹細胞とを含む脂肪由来神 経堤細胞前駆体となることを特徴し得る。  [0113] In another embodiment, the method may further comprise the step of confirming the level of expression of the adipocyte marker by the stem cell. The adipocyte marker used in the present method may be any as long as it can determine adipocytes, but preferably Leptin (NM000230 (human)) can be used. It is not limited to. Thus, confirmation of the level of adipocyte markers can be confirmed by a decrease in Leptin (NM000230 (human)). In one embodiment, the stem cells that have also obtained fat power become fat-derived neural crest cell precursors including neural crest cells and adipose-derived stem cells by the step of subjecting them to conditions for inducing neural crest cells. Can do.
[0114] 別の実施形態において、本方法は、幹細胞が神経堤細胞マーカーまたは神経幹 細胞マーカーを発現して!/ヽるかどうかを確認する工程、および前記幹細胞が脂肪細 胞マーカーを発現のレベルを確認する工程をさらに包含することができる。好ましく は、例えば、神経堤細胞の細胞マーカーとしては、例えば、 CRABP1 (NM004378 (ヒ卜))、 AP2 (NM002097 (ヒ卜))、 Slug (NM003068 (ヒ卜))、 SoxlO (NM0069 41 (ヒ卜))、 Snail (NM005985 (ヒ卜))、 Twist (NM000474 (ヒ卜))、 Pax3 (NM00 0438 (ヒ卜))、 Pax7 (NM002584 (ヒ卜))、 HNKl (NM004854 (ヒ卜))、 p75NTR ( NM002507 (ヒト))、 TRP2 (NM006267 (ヒト))、 Wntl (NM005430 (ヒト))、 P0 (NM002723 (ヒト) )、 tPA (NM000930 (ヒト) )などを挙げることができるがこれらに 限定されない。例えば、神経幹細胞マーカーは、 Nestin(NM006617 (tl ) )、Mu sashi- 1 (NM002442 (ヒ卜))、 CD133 (NM0006017 (ヒ卜))、 notchl (NM017 617 (ヒ卜))、 Hesl (NM005524 (ヒ卜))、 Mashl (NM004316 (ヒ卜))、 Neurogeni n (NM006161 (ヒ卜))、 Pax6 (NM001604 (ヒ卜))、 CD15 (NM002033 (ヒ卜))、 P DGFR (NM002609 (ヒト) )などが挙げられるがこれらに限定されな!、。好ましくは、 神経幹細胞マーカーは、 Nestinおよび Musashi—lを含む。この場合、本発明の神 経堤細胞は、 Nestinおよび Musashi— 1を発現し、かつ、脂肪細胞マーカーである Leptinの発現が脂肪由来幹細胞のものの発現レベルよりも低いことを特徴とし得る。 [0114] In another embodiment, the method wherein the stem cell expresses a neural crest cell marker or neural stem cell marker! The method may further comprise the step of confirming whether or not the stem cell is confirmed, and the step of confirming the level of expression of the fat cell marker by the stem cell. Preferably, for example, cell markers of neural crest cells include, for example, CRABP1 (NM004378 (Hin)), AP2 (NM002097 (Hin)), Slug (NM003068 (Hin)), SoxlO (NM0069 41 (Hin) )), Snail (NM005985 (Hui)), Twist (NM000474 (H2)), Pax3 (NM00 0438 (H2)), Pax7 (NM002584 (H2)), HNKl (NM004854 (H2)), p75NTR (NM002507 (human)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), P0 (NM002723 (human)), tPA (NM000930 (human)), and the like. It is not limited. For example, neural stem cell markers include Nestin (NM006617 (tl)), Mu sashi-1 (NM002442 (chicken)), CD133 (NM0006017 (chicken)), notchl (NM017 617 (chicken)), Hesl (NM005524 (卜)), Mashl (NM004316 (()), Neurogenin (NM006161 (卜)), Pax6 (NM001604 (卜)), CD15 (NM002033 (卜)), PDGFR (NM002609 (human)) But is not limited to these! Preferably, neural stem cell markers include Nestin and Musashi-l. In this case, the neural crest cells of the present invention may be characterized by expressing Nestin and Musashi-1 and expressing Leptin, which is an adipocyte marker, lower than the expression level of adipose-derived stem cells.
[0115] これらの神経堤細胞マーカー、神経幹細胞マーカー、または脂肪細胞マーカーに よる判定は、当該分野において公知の方法 (例えば、 RT— PCR、ノーザンブロット、 ウェスタンブロット、免疫染色、 FACSなど)を用いて行うことができる。ここで、発現し ているかまたはしていないかは、 RT—PCR、ノーザンブロット、ウェスタンブロット、免 疫染色、 FACSなどによって、適宜選択することができる。  [0115] The determination using these neural crest cell marker, neural stem cell marker, or adipocyte marker is performed using a method known in the art (eg, RT-PCR, Northern blot, Western blot, immunostaining, FACS, etc.). Can be done. Here, whether it is expressed or not can be appropriately selected by RT-PCR, Northern blot, Western blot, immunostaining, FACS and the like.
[0116] 別の実施形態において、本方法は、幹細胞が遊走能を有するかどうかを確認する 工程をさらに包含し得る。この遊走能は、胚細胞の頭部領域に移植し、その後、前頭 鼻隆起、上顎隆起、鰓弓の間充織に遊走することを観察することによって確認され得 る。  [0116] In another embodiment, the method may further comprise the step of determining whether the stem cell has the ability to migrate. This migratory ability can be confirmed by transplanting into the head region of the embryonic cell and then observing that it migrates to the frontal nasal protuberance, maxillary protuberance, and arch mesenchyme.
[0117] (脂肪由来の神経堤細胞)  [0117] (Adipose-derived neural crest cells)
本発明では、脂肪由来の神経堤細胞が提供される。このような神経堤細胞は、本 発明の上記方法などによって得ることができる。好ましくは、脂肪は、脂肪由来幹細 胞であり得るが、脂肪組織自体を使用してもよ ヽことが理解される。  In the present invention, a neural crest cell derived from fat is provided. Such neural crest cells can be obtained by the above-described method of the present invention. Preferably, the fat can be a fat-derived stem cell, but it will be understood that the adipose tissue itself may be used.
[0118] 本発明の神経堤細胞はまた、遊走能を有し得る。この遊走能は、胚の頭部領域に 目的の細胞を移植し、その後、前頭鼻隆起、上顎隆起、鰓弓の間充織に遊走するこ とを観察することによって確認され得る。  [0118] The neural crest cells of the present invention may also have migration ability. This migration ability can be confirmed by transplanting the cells of interest into the head region of the embryo and then observing that they migrate to the frontal nasal protuberance, maxillary protuberance, and arch mesenchyme.
[0119] 本発明の神経堤細胞はまた、神経堤細胞マーカー(例えば、 CRABP1 (NM004 378 (ヒ卜))、 AP2 (NM002097 (ヒ卜))、 Slug (NM003068 (ヒ卜))、 SoxlO (NM0 06941 (ヒ卜))、 Snail (NM005985 (ヒ卜))、 Twist (NM000474 (ヒ卜))、 Pax3 (N M000438 (ヒ卜))、 Pax7 (NM002584 (ヒ卜))、 HNKl (NM004854 (ヒ卜))、 p75 NTR (NM002507 (ヒト) )、 TRP2 (NM006267 (ヒト) )、 Wntl (NM005430 (ヒト) )、 PO (NM002723 (ヒト))ゝ tPA (NM000930 (ヒト)))を少なくとも 1つ発現し得る。 [0119] The neural crest cells of the present invention also have neural crest cell markers (for example, CRABP1 (NM004 378 (Hin)), AP2 (NM002097 (Hin)), Slug (NM003068 (Hin)), SoxlO (NM0). 06941 (Hui)), Snail (NM005985 (H 卜)), Twist (NM000474 (H 卜)), Pax3 (N M000438 (H 卜)), Pax7 (NM002584 (H 卜)), HNKl (NM004854 (H 卜) )), P75 NTR (NM002507 (human)), TRP2 (NM006267 (human)), Wntl (NM005430 (human)), PO (NM002723 (human)) tPA (NM000930 (human))) can be expressed.
[0120] 本発明の神経堤細胞はさらに、神経幹細胞マーカー(例えば、 Nestin (NM0066 17 (ヒ卜))、 Musashi— 1 (NM002442 (ヒ卜))、 CD133 (NM0006017 (ヒ卜))、 not chl (NM017617 (ヒ卜))、 Hesl (NM005524 (ヒ卜))、 Mashl (NM004316 (ヒ卜) )、Neurogenin (NM006161 (ヒト))、Pax6 (NM001604 (ヒト))、CD15 (NM00 2033 (ヒト))、 PDGFR (NM002609 (ヒト))を少なくとも 1つ発現し得る。好ましくは 、 Nestinおよび Musashi— 1を発現する細胞を同定することによって、本発明の神 経堤細胞を同定することができる。  [0120] The neural crest cells of the present invention further comprise neural stem cell markers (for example, Nestin (NM0066 17 (Hin)), Musashi-1 (NM002442 (Hin)), CD133 (NM0006017 (Hin)), not chl (NM017617 (Human)), Hesl (NM005524 (Human)), Mashl (NM004316 (Human)), Neurogenin (NM006161 (Human)), Pax6 (NM001604 (Human)), CD15 (NM00 2033 (Human)) PDGFR (NM002609 (human)) can be expressed, preferably the neural crest cells of the present invention can be identified by identifying cells that express Nestin and Musashi-1.
[0121] 本発明では、独自の遊走能および少なくとも 1つの神経堤細胞の細胞マーカーの 発現からなる群より選択される少なくとも 1つの特徴を有するか否かを確認すること〖こ より、本発明の脂肪由来の神経堤細胞であると同定することができる。好ましくは、本 発明の神経堤細胞は、独自の遊走能および少なくとも 1つの神経堤細胞の細胞マー カーの発現の両方の特徴を確認することによって同定される。この独特の遊走能は、 胚の頭部領域に移植し、その後前頭鼻隆起、上顎隆起、鰓弓の間充織に遊走する ことを観察すること〖こより確認することができる。  [0121] In the present invention, it is confirmed whether or not it has at least one characteristic selected from the group consisting of unique migration ability and expression of a cell marker of at least one neural crest cell. It can be identified as a neural crest cell derived from fat. Preferably, neural crest cells of the present invention are identified by confirming both the unique migration ability and the expression of cell markers of at least one neural crest cell. This unique migratory ability can be confirmed by transplanting into the embryonic head region and then observing that it migrates to the frontal nasal protuberance, maxillary protuberance, and meridian mesenchyme.
[0122] 1つの実施形態では、本発明の細胞は、 Leptin (NM000230 (ヒト))の発現が脂 肪由来幹細胞のものの発現レベルよりも低 、。 [0122] In one embodiment, in the cell of the present invention, Leptin (NM000230 (human)) expression is lower than that of a fat-derived stem cell.
[0123] 本発明の好まし 、実施形態では、本発明の細胞は、 Nestinおよび Musashi— 1を 発現し、かつ、 Leptinの発現が脂肪由来幹細胞のものの発現レベルよりも低いことを 特徴とする。 [0123] In a preferred embodiment of the present invention, the cell of the present invention is characterized in that it expresses Nestin and Musashi-1, and the expression level of Leptin is lower than that of an adipose-derived stem cell.
[0124] (混合物) [0124] (mixture)
別の局面において、本発明は、神経堤細胞と脂肪由来幹細胞とを含む、脂肪由来 神経堤細胞混合物を提供する。本発明の脂肪由来神経堤細胞混合物に含まれる神 経堤細胞は、本明細書において、「脂肪由来の神経堤細胞」および「神経堤細胞を 調製する方法」において説明したような任意の形態であり得る。本発明の脂肪由来神 経堤細胞混合物に含まれる脂肪由来幹細胞は、本明細書において、「脂肪由来幹 細胞」において説明したような任意の形態であり得る。本発明の混合物は、神経堤細 胞と脂肪由来肝細胞とを含んでさえいれば、どのような形態であってもよい。本発明 の混合物は、神経幹細胞も含み得る。 In another aspect, the present invention provides a fat-derived neural crest cell mixture comprising neural crest cells and adipose-derived stem cells. The neural crest cells contained in the fat-derived neural crest cell mixture of the present invention are in any form as described in the “adipose-derived neural crest cells” and “method for preparing neural crest cells” in the present specification. possible. The adipose-derived stem cells contained in the adipose-derived neural crest cell mixture of the present invention can be in any form as described in “Adipose-derived stem cells” in the present specification. The mixture of the present invention Any form is acceptable as long as it includes vesicles and adipose-derived hepatocytes. The mixture of the present invention may also contain neural stem cells.
[0125] (組成物)  [0125] (Composition)
別の局面において、本発明は、脂肪由来または脂肪由来幹細胞由来の神経堤細 胞を含む、神経系疾患、障害または状態の処置のための細胞移植のための組成物 を提供する。神経系疾患、障害または状態は、例えば、神経系の分化細胞の欠損に 起因する疾患、障害または状態であり得るがこれらに限定されない。この組成物は、 神経系疾患、障害または状態を処置または予防するためであれば、任意の目的で使 用することができる。使用される組成物中の神経堤細胞は、本明細書において、「脂 肪由来の神経堤細胞」および「神経堤細胞を調製するための方法」において説明し たような任意の形態であり得る。  In another aspect, the present invention provides a composition for cell transplantation for the treatment of nervous system diseases, disorders or conditions, comprising adipose-derived or adipose-derived stem cell-derived neural crest cell. The nervous system disease, disorder or condition can be, but is not limited to, a disease, disorder or condition resulting from, for example, a loss of differentiated cells of the nervous system. The composition can be used for any purpose as long as it is intended to treat or prevent a nervous system disease, disorder or condition. The neural crest cells in the composition used may be in any form as described herein in “Fat-derived neural crest cells” and “Methods for preparing neural crest cells”. .
[0126] ここで、神経堤細胞は、それぞれ独立して、移植されるべき宿主に対して、異種、同 種異系または同系であり得る。好ましくは、同種異系または同系であり、より好ましくは 同系であるが、それに限定されない。理論に束縛されないが、同系であれば、免疫 拒絶反応が抑制できるからである。しかし、拒絶反応が予測される場合は、拒絶反応 を回避する工程をさらに包含してもよい。拒絶反応を回避する手順は当該分野にお いて公知であり、例えば、新外科学体系、第 12卷、心臓移植 ·肺移植 技術的,倫理 的整備から実施に向けて (改訂第 3版)、中山書店を参照のこと。そのような方法とし ては、例えば、免疫抑制剤、ステロイド剤の使用などの方法が挙げられるが、それら に限定されない。拒絶反応を予防する免疫抑制剤は、例えば、現在、「シクロスポリン 」(サンデイミユン Zネオ一ラル)、「タクロリムス」(プログラフ)、「ァザチォプリン」(ィム ラン)、「ステロイドホルモン」(プレドニン、メチルプレドニン)、および「T細胞抗体」(Ο KT3、 ATGなど)があり、予防的免疫抑制療法として世界の多くの施設で行われて いる方法は、「シクロスポリン、ァザチォプリンおよびステロイドホルモン」の 3剤併用で ある。免疫抑制剤は、本発明の薬剤と同時期に投与されることが望ましいが、必ずし も必要ではない。従って、免疫抑制効果が達成される限り免疫抑制剤は本発明の再 生療法 ·治療の前または後にも投与され得る。  [0126] Here, the neural crest cells can be independently xenogeneic, allogeneic or syngeneic with respect to the host to be transplanted. Preferably, they are allogeneic or syngeneic, more preferably syngeneic, but not limited thereto. Without being bound by theory, it is possible to suppress immune rejection if it is syngeneic. However, if rejection is predicted, a step of avoiding rejection may be further included. Procedures to avoid rejection are well known in the art, for example, from the New Surgery System, Section 12, Heart / Lung Transplantation from technical and ethical arrangements to implementation (Revised 3rd Edition), See Nakayama Shoten. Examples of such methods include, but are not limited to, methods such as the use of immunosuppressants and steroids. Immunosuppressants that prevent rejection include, for example, “Cyclosporine” (Sunday Miyun Z Neoral), “Tacrolimus” (Prograf), “Azathioprine” (Imlan), “Steroid Hormone” (predonin, methylpredonin) ), And “T-cell antibodies” (ΟKT3, ATG, etc.), and the method used in many centers around the world as a preventive immunosuppressive therapy is a combination of three drugs “cyclosporine, azathioprine and steroid hormones”. is there. The immunosuppressive agent is desirably administered at the same time as the agent of the present invention, but it is not absolutely necessary. Therefore, as long as the immunosuppressive effect is achieved, the immunosuppressive agent can be administered before or after the regeneration therapy / treatment of the present invention.
[0127] このような組成物は、医薬として提供され得る。このような医薬は、神経系疾患、障 害または状態 (例えば、神経系の分ィ匕細胞の欠損に起因する疾患、障害または状態 )を処置または予防するために用いられる。本発明の医薬には、このような組成物の ほか、薬学的に受容可能なキャリアが含まれていてもよい。そのようなキャリアとしては[0127] Such a composition may be provided as a medicament. Such medicines are used for nervous system diseases, disorders It is used to treat or prevent a harm or condition (eg, a disease, disorder or condition resulting from a loss of neuronal dividing cells). The medicament of the present invention may contain a pharmaceutically acceptable carrier in addition to such a composition. As such a carrier
、その用途に応じて当業者によって、本明細書において記載される任意のキャリアが 選択され、そして用いることができる。 Depending on its application, any carrier described herein can be selected and used by one of ordinary skill in the art.
[0128] (神経系疾患、障害または状態の処置のための細胞移植の方法)  [0128] (Methods of cell transplantation for the treatment of nervous system diseases, disorders or conditions)
別の局面において、本発明は、神経系疾患、障害または状態の処置 (例えば、神 経系の分化細胞の欠損に起因する疾患、障害または状態)のための細胞移植のた めの方法を提供する。この方法は、脂肪由来または脂肪由来幹細胞由来の神経堤 細胞を投与する工程を包含する。ここで、移植に使用される神経堤細胞は、本明細 書において、「脂肪由来の神経堤細胞」および「神経堤細胞を調製するための方法」 において説明したような任意の形態であり得る。  In another aspect, the invention provides a method for cell transplantation for the treatment of a nervous system disease, disorder or condition (eg, a disease, disorder or condition resulting from a loss of differentiated cells of the nervous system). To do. This method includes the step of administering a neural crest cell derived from a fat or a fat-derived stem cell. Here, the neural crest cells used for transplantation can be in any form as described herein in “Adipose-derived neural crest cells” and “Methods for preparing neural crest cells”.
[0129] この神経堤細胞は、当該分野において公知の任意の方法によって投与することが できる。例えば、この神経堤細胞は、シリンジ、カテーテル、チューブなどを用いて注 入され得るがそれらに限定されない。好ましくは、例示的な投与形態としては、局所 注入 (皮下注入、筋肉や脂肪など臓器内注入)、静脈内注入、動脈内注入、組織上 投与などが挙げられる力 それらに限定されない。本発明のこの移植による処置また は予防方法力 Sもたらす、例えば、従来技術と比べて有利な点としては、例えば、(1) 生体外で再生組織を作る(ェキソビボ産生)必要がな 、; (2)より確実に大きな組織の 再生が可能である;(3)簡便かつ短時間の処理により産生が可能である;(4)皮膚な ど臓器を切開する手術を必要とせず、針を刺すことによって細胞および組織を投与( 移植)することが可能であることなどが挙げられるがそれらに限定されない。  [0129] The neural crest cells can be administered by any method known in the art. For example, the neural crest cells can be injected using a syringe, a catheter, a tube, and the like, but are not limited thereto. Preferably, exemplary dosage forms include, but are not limited to, local injection (subcutaneous injection, intramuscular or intramuscular injection), intravenous injection, intraarterial injection, or tissue administration. For example, as an advantage compared with the prior art, for example, (1) it is necessary to create a regenerated tissue ex vivo (ex vivo production); 2) Larger tissue regeneration is possible more reliably; (3) It can be produced by simple and short-time treatment; (4) Needle insertion without the need for surgery to open organs such as skin It is possible to administer (transplant) cells and tissues by, but not limited to.
[0130] (使用)  [0130] (Use)
別の局面において、本発明は、神経堤細胞の、神経系疾患、障害または状態を処 置または予防するための医薬の調製のための使用を提供する。ここで、医薬の調製 のために使用される神経堤細胞は、本明細書において、「脂肪由来の神経堤細胞」 および「神経堤細胞を調製する方法」において説明したような任意の形態であり得る [0131] 以下に、実施例に基づいて本発明を説明するが、以下の実施例は、例示の目的の みに提供される。従って、本発明の範囲は、上記実施形態にも下記実施例にも限定 されるものではなぐ添付の特許請求の範囲によってのみ限定される。 In another aspect, the present invention provides the use of neural crest cells for the preparation of a medicament for treating or preventing a nervous system disease, disorder or condition. Here, the neural crest cells used for the preparation of the medicament are in any form as described herein in “Adipose-derived neural crest cells” and “Method of preparing neural crest cells”. obtain [0131] Hereinafter, the present invention will be described based on examples. However, the following examples are provided for illustrative purposes only. Accordingly, the scope of the present invention is limited only by the appended claims, which are not limited to the above-described embodiment and the following examples.
実施例  Example
[0132] 以下に示した実施例において使用した試薬は、特に言及しない限り和光純薬、 Sig ma力ら得た。動物の飼育は、 National Society for Medical Researchg作成 した [Principles of Laboratory Animal CareJおよひnstitute of Laborat ory Animal Resource力 S作成、 National Institute of Healths、公表した「G uide for the Care and Use of Laboratory AnimalsJ (NIH Publicatio n, No. 86- 23, 1985,改訂)に遵つて、動物愛護精神に則って行った。ヒトを対象 とする場合は、事前に同意を得た上で実験を行った。  [0132] Reagents used in the following examples were obtained from Wako Pure Chemicals and Sigma force unless otherwise specified. Animal breeding was created by the National Society for Medical Researchg (Principles of Laboratory Animal CareJ and the Institute of Laboratory Animal Resource S, National Institute of Healths, published `` Guide for the Care and Use of Laboratory AnimalsJ ( NIH Public Ratio, No. 86-23, 1985, revised), and conducted in accordance with the spirit of animal welfare.
[0133] (実施例 1:コラゲナーゼを用いる脂肪由来幹細胞の調製)  (Example 1: Preparation of adipose-derived stem cells using collagenase)
本実施例では、まず、脂肪由来幹細胞を、本実験に対して同意を示したヒトから吸 引脂肪より調製した。詳細には、吸引脂肪を 1リットル大の分液漏斗を用いて生理食 塩水で十分に洗浄した。上層に吸引脂肪、下層に生理食塩水が十分に分離したの を確認し、下層を捨て、肉眼で見て生理食塩水がほぼ透明になるまでこれを繰り返し た。この実施例では、 5回行った。  In this example, first, adipose-derived stem cells were prepared from sucked fat from humans who gave consent to this experiment. Specifically, the aspirated fat was thoroughly washed with physiological saline using a 1-liter separatory funnel. After confirming that the aspirated fat was sufficiently separated in the upper layer and the physiological saline was sufficiently separated in the lower layer, the lower layer was discarded, and this was repeated until the physiological saline became almost transparent with the naked eye. In this example, it was performed five times.
[0134] 吸引脂肪を 10mlと同量の 0. 075%コラゲナーゼ ZPBSを加え、 37°Cでよく攪拌 しながら 30分間インキュベートした。この試料に、同量の 10%血清加 DMEMを加え 、 1200 X gで 10分間遠心分離した。  [0134] The aspirated fat was added in an amount equal to 10 ml of 0.075% collagenase ZPBS, and incubated at 37 ° C for 30 minutes with good agitation. To this sample, the same amount of 10% serum-added DMEM was added and centrifuged at 1200 × g for 10 minutes.
[0135] 遠心分離により得られたペレットに 0. 16M NH ClZPBSをカ卩えて懸濁し、室温  [0135] Add 0.16M NH 4 ClZPBS to the pellet obtained by centrifugation and suspend it at room temperature.
4  Four
で 10分間インキュベートした。この試料を口径 100 μ mのメッシュ(Whatman)を用 いて吸引ろ過した。この得られたろ過物を 1200 X gで 5分間遠心分離した。この細胞 調製物は、 PLAとも呼ぶ。幹細胞であることは、細胞マーカー(例えば、 CD4、 CD1 3、 CD34、 CD36、 CD49d、 CD71、 CD90、 CD105、 CD117、 CD151等)を用 いて確認する。  Incubated for 10 minutes. This sample was subjected to suction filtration using a mesh (Whatman) having a diameter of 100 μm. The obtained filtrate was centrifuged at 1200 × g for 5 minutes. This cell preparation is also called PLA. Stem cells are confirmed using cell markers (eg, CD4, CD13, CD34, CD36, CD49d, CD71, CD90, CD105, CD117, CD151, etc.).
[0136] (実施例 2 :脂肪吸引による吸引物の液体部分力 の、細胞懸濁液の調製)  [0136] (Example 2: Preparation of cell suspension of liquid partial force of aspirate by liposuction)
脂肪吸引による吸引物の液体部分について、以下の 2方法のいずれかを用いて処 理すること〖こよって、幹細胞懸濁液を調製した。以下の 2方法のいずれも、コラゲナー ゼなどの酵素を用いる処理が不要であるため、コラゲナーゼなどの酵素の混入がな い点において、従来法とは異なる。 Use one of the following two methods for the liquid part of the aspirate by liposuction. Thus, a stem cell suspension was prepared. Either of the following two methods is different from the conventional method in that no treatment with an enzyme such as collagenase is required, and therefore no enzyme such as collagenase is mixed.
[0137] (I)調製方法 1 [0137] (I) Preparation Method 1
1)脂肪吸引による吸引物の液体部分 (通常、 2〜4リットル程度)を 400 X g、 10分間 、遠心分離した。  1) The liquid part (usually about 2-4 liters) of the aspirate by liposuction was centrifuged at 400 X g for 10 minutes.
2)上清を捨てた。ただし、沈殿した細胞は浮遊しやすいため、ァスピレーターを用い て、細胞を損傷しないよう慎重に吸引した。  2) The supernatant was discarded. However, since the precipitated cells tend to float, they were carefully aspirated using an aspirator so as not to damage the cells.
3)沈殿した細胞(ほとんどが赤血球)を 50mlのポリプロピレン製チューブ数本に移し 、遠心分離 (400 X g、 5分間)した。  3) The precipitated cells (mostly red blood cells) were transferred to several 50 ml polypropylene tubes and centrifuged (400 X g, 5 minutes).
4)上清を吸引し、総量が 15〜20mlとなるように沈殿細胞を集めた。マトリクス成分が 多く含まれる場合は、マトリクス成分を、 100 /z mフィルターで濾過'除去した。その後 必要に応じて、遠心分離を行なった。  4) The supernatant was aspirated and the precipitated cells were collected so that the total volume was 15-20 ml. When many matrix components were included, the matrix components were filtered out with a 100 / zm filter. Thereafter, centrifugation was performed as necessary.
5) 50mlのチューブにフイコール(登録商標) 15mlを入れ、その上に層を作るように 極力ゆっくりと細胞液 15〜 20mlを加えた。  5) Put 15 ml of Ficoll (registered trademark) into a 50 ml tube, and slowly add 15-20 ml of cell solution as much as possible to form a layer on it.
6)チューブを 400 X g、 30分間、遠心分離した。(18〜20°C)  6) The tube was centrifuged at 400 xg for 30 minutes. (18-20 ° C)
7)遠心分離後、細胞溶液を 4層に分離した。上から (A層)無細胞層 (透明)、(B層) 単核球層(淡赤色)、(C層)フイコール層(透明)、(D層)赤血球層(濃赤色)で、幹細 胞を含む接着細胞群が、 B層および C層に含まれていた。 A層を吸引後、 B層および 約 3ml程度の C層を細胞懸濁液として回収し、 50mlのチューブに移した。  7) After centrifugation, the cell solution was separated into 4 layers. From the top (A layer) Cell-free layer (Transparent), (B layer) Mononuclear cell layer (light red), (C layer) Ficoll layer (transparent), (D layer) Red blood cell layer (dark red) Adherent cell groups including vesicles were included in B and C layers. After the A layer was aspirated, the B layer and about 3 ml of the C layer were collected as a cell suspension and transferred to a 50 ml tube.
8)回収した細胞懸濁液に、血清加 PBS (10%FBS、もしくは 10%ヒト血清をカ卩えた PBS)を加えて 50mlとし、ピペッティングによる混和後、遠心分離 (400 X g、 5分間) した。  8) Add serum-containing PBS (10% FBS or PBS containing 10% human serum) to the collected cell suspension to make 50 ml, mix by pipetting, and centrifuge (400 X g, 5 minutes) ) did.
9)上清を吸引し、再度血清加 PBSをカ卩えて 50mlとし、ピペッティングによる混和後、 遠心分離 (400 X g、 5分間)した。  9) The supernatant was aspirated, again added with serum-added PBS to 50 ml, mixed by pipetting, and then centrifuged (400 X g, 5 minutes).
10)上清を吸引し、沈殿した幹細胞を含む細胞群を回収した。  10) The supernatant was aspirated and a cell group containing the precipitated stem cells was collected.
[0138] (Π)調製方法 2 [0138] (Π) Preparation method 2
1)クリーンベンチ内で、吸引管を用いて脂肪吸引による吸引物の液体部分を吸引し 、フィルター(ポアサイズ; 120 /z m)付きリザーバーを通して、ろ過液を閉鎖分離バッ クに封人した。 1) In the clean bench, use the suction tube to suck the liquid part of the sucked material by liposuction. The filtrate was sealed in a closed separation bag through a reservoir with a filter (pore size; 120 / zm).
2)セルセパレーター(血液成分分離装置 ASTEC204、(株)アムコ、東京、 日本)で 遠心分離法にて 3回プロセシングを行い、比重の軽い血小板成分、比重の重い赤血 球、顆粒球成分を可及的に除去した。  2) Using a cell separator (blood component separator ASTEC204, Amco Corporation, Tokyo, Japan), centrifuge three times to obtain platelet components with low specific gravity, red blood cells with high specific gravity, and granulocyte components. Removed as much as possible.
3)幹細胞を高濃度に含む分画 (約 30〜40ml)を採取した。単離した細胞の比重は 、 1. 050〜1. 075の範囲であった。  3) A fraction (about 30 to 40 ml) containing a high concentration of stem cells was collected. The specific gravity of the isolated cells ranged from 1.050 to 1.075.
[0139] おおまかな細胞の比重は、パーコール TM、レディグラッド TMなどのような密度勾配遠 心分離媒体を塩ィ匕ナトリウム溶液またはスクロース溶液に配合し、収集した細胞およ びデンシティ一マーカービーズ(density marker beads)を混合物に加えて遠心 し、ビーズによって分けられた 5— 10層のうち、どの層に細胞がある力 (細胞を含む層 が細胞の比重を示す)を確認することで、調べることが可能である。 [0139] Rough cell specific gravity is determined by mixing density gradient centrifuge separation media such as Percoll , Readygrad ™, etc. in saline-sodium solution or sucrose solution, and collecting collected cells and density-one marker beads ( Add density marker beads) to the mixture, centrifuge, and check by checking the force of cells in which of the 5-10 layers divided by the beads (the layer containing the cells indicates the specific gravity of the cells) It is possible.
[0140] (実施例 3:回収した幹細胞の特徴付け)  [0140] (Example 3: Characterization of recovered stem cells)
実施例 2にお ヽて回収した幹細胞を、以下の手順で FACSを用いて特徴付けした  The stem cells collected in Example 2 were characterized using FACS according to the following procedure:
[0141] 約 5mlの細胞懸濁液を、染色培地(SM ;0. 5% ゥシ血清アルブミンおよび 0. 05 % NaN3を含む PBS)で 2回洗浄した。必要に応じて、細胞を計数した。 [0141] Approximately 5 ml of the cell suspension was washed twice with staining medium (SM; PBS containing 0.5% ushi serum albumin and 0.05% NaN3). Cells were counted as needed.
[0142] 約 1〜: LO X 106細胞 Zmlの細胞懸濁液に対して、最終濃度として 0. 001-0. 1 [0142] About 1 to: LO X 10 6 cells A final concentration of 0.001-0.
gZmlの標識ィ匕抗体 (標識には、フィコエリトリン (PE)、ァロフィコシァニン (APC) および Zまたはフルォレセインイソチオシァネート (FITC)を用いた)を添カ卩した。  A gZml labeled antibody (phycoerythrin (PE), allophycocyanin (APC) and Z or fluorescein isothiocyanate (FITC) was used for labeling) was added.
[0143] 氷上で 30分間ほど、混合物をインキュベーションした後、細胞を洗浄し、 SMを用 いて細胞浮遊液の濃度を 5 X 105細胞 Zml程度に調整した。 [0143] After incubating the mixture for about 30 minutes on ice, the cells were washed, and the concentration of the cell suspension was adjusted to about 5 X 10 5 cells Zml using SM.
[0144] FACS Vantage (Becton Dickinson社)を使用した。抗体の標識を指標として 、単離した幹細胞における各種 CDタンパク質の発現を解析した。その結果、脂肪吸 引による吸引物の液体部分由来の幹細胞は、表 4に示すように、 CD90および CD4 9dを発現することが判明した。  [0144] FACS Vantage (Becton Dickinson) was used. Using the antibody label as an indicator, the expression of various CD proteins in the isolated stem cells was analyzed. As a result, as shown in Table 4, stem cells derived from the liquid portion of the aspirate from fat suction were found to express CD90 and CD49d.
[0145] 単離した幹細胞を、 DMEM培地にお!、て 2回継代培養した。継代は、 80%のコン フルエンス時に行った。 2度の継代培養後の細胞を、上記と同様の手順で FACS〖こ よる分析を行った。その結果を以下の表 4に示す。 [0145] The isolated stem cells were subcultured twice in DMEM medium! Passaging was done at 80% confluence. Use the same procedure as above to incubate cells after the second passage. Analysis was performed. The results are shown in Table 4 below.
(表 4 : 2回継代培養した後の幹細胞における種々の CDの発現) CD 発現量  (Table 4: Expression of various CDs in stem cells after 2 passages) CD expression level
3  Three
4  Four
11c  11c
13 + +  13 + +
14  14
15  15
16  16
19  19
29 + +  29 + +
31 +  31 +
33  33
34 +  34 +
36 + +  36 + +
38  38
44 +  44 +
45 +  45 +
49d + +  49d ++
54 +  54 +
56  56
58 +  58 +
61  61
62E  62E
62P  62P
69  69
71 + + 73 + + 71 + + 73 + +
90 + +  90 + +
104  104
105 + +  105 + +
106  106
117 +  117 +
135  135
144  144
146 +  146 +
151 + +  151 + +
235a - SH3 +  235a-SH3 +
STRO- 1 +  STRO- 1 +
「一」 =発現検出されず、  “One” = expression not detected
「 +」 = 20%以下の細胞に検出される  "+" = Detected in 20% or less of cells
「+ +」= 20%以上の細胞に検出される  "++" = detected in more than 20% of cells
以上の結果から、脂肪吸引による吸引物の液体部分力 調製された幹細胞には、 間葉系幹細胞は含まれるものの、従来法によって調製される脂肪由来幹細胞群と異 なり、 CD31、 34陽性細胞が含まれた。従って、本発明の方法によって調製された幹 細胞は、血管内皮への分化 (血管新生)が容易かつ高効率で可能な細胞群であるこ とが理解できる。さらに、本明細書中で指標として用いた CD発現は、 2回継代培養し た後に確認されていることから、本発明の幹細胞は、 2回程度の継代培養後もその表 現型をほとんど変化しな 、ことが理解される。  From the above results, stem cells prepared by liquid partial force of aspirate by liposuction contain mesenchymal stem cells, but differ from the adipose-derived stem cell group prepared by conventional methods, and CD31, 34 positive cells are Included. Therefore, it can be understood that the stem cells prepared by the method of the present invention are a group of cells that can easily differentiate into vascular endothelium (angiogenesis) with high efficiency. Furthermore, since the CD expression used as an index in the present specification has been confirmed after being subcultured twice, the stem cell of the present invention shows its phenotype after about two subcultures. It is understood that there is little change.
[0147] (実施例 4 :複数の被検体力 脂肪吸引により得た吸引物の液体部分より回収した 幹細胞の特徴付け) [0147] (Example 4: Characterization of stem cells collected from the liquid portion of the aspirate obtained by liposuction of multiple subjects)
さらに、複数の被検体力 脂肪吸引により得た吸引物の液体部分より幹細胞を回 収し、その特徴付を行った。その結果を以下に示す。  Furthermore, stem cells were collected from the liquid portion of the aspirate obtained by multiple subject force liposuction and characterized. The results are shown below.
[0148] (表 5 :複数の被検体力 脂肪吸引により得た吸引物の液体部分より回収した幹細 胞の特徴付けの結果) [0148] (Table 5: Multiple subject forces Stem fines collected from the liquid portion of aspirate obtained by liposuction (Results of cell characterization)
[表 5] [Table 5]
Figure imgf000050_0001
数字は、細胞集団中で、各タンパク質を発現する幹細胞の割合 (%)を示す。
Figure imgf000050_0001
Numbers indicate the percentage of stem cells expressing each protein in the cell population.
「一」 =発現検出されず、「 +」=発現検出された、 N. T. =試験せず。  “One” = expression not detected, “+” = expression detected, N. T. = not tested.
収集された幹細胞は、その集団のほとんどの細胞が、 CD13、 CD29、 CD34、 CD 36、 CD44、 CD49d、 CD54、 CD58、 CD71、 CD73、 CD90、 CD105、 CD106 、 CD151、および SH3について陽性であった。従って、本発明の脂肪由来幹細胞 は、 CD13、 CD29、 CD34、 CD36、 CD44、 CD49d、 CD54、 CD58、 CD71、 C D73、 CD90、 CD105、 CD106、 CD151、および SH3力もなる群より選択される少 なくとも 1つのタンパク質を発現する細胞である。 CD106を発現する幹細胞であるこ とが、本発明において使用される脂肪由来幹細胞の特徴の 1つである。また、 CD31 、 CD45、 CD117、および CD146については、その幹細胞の集団の一部が陽性で あり、一部は陰'性であった。 The collected stem cells are most of the population of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106 Positive for CD151 and SH3. Therefore, the adipose-derived stem cells of the present invention are selected from the group consisting of CD13, CD29, CD34, CD36, CD44, CD49d, CD54, CD58, CD71, CD73, CD90, CD105, CD106, CD151, and SH3 force. Both are cells that express one protein. One feature of the adipose-derived stem cells used in the present invention is that they are stem cells that express CD106. Also, for CD31, CD45, CD117, and CD146, some of the stem cell populations were positive and some were negative.
[0151] その幹細胞の集団は、 CD3、 CD4、 CD14、 CD15、 CD16、 CD19、 CD33、 CD 38、 CD56、 CD61、 CD62e、 CD62p、 CD69、 CD104、 CD135、および CD144 については、陰性であった。従って、本発明の脂肪由来幹細胞は、 CD3、 CD4、 C D14、 CD15、 CD16、 CD19、 CD33、 CD38、 CD56、 CD61、 CD62e、 CD62p 、 CD69、 CD104、 CD135、および CD144の少なくとも 1つを発現しない細胞であ る。 [0151] The stem cell population was negative for CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD135, and CD144. Therefore, the adipose-derived stem cells of the present invention do not express at least one of CD3, CD4, CD14, CD15, CD16, CD19, CD33, CD38, CD56, CD61, CD62e, CD62p, CD69, CD104, CD135, and CD144 It is a cell.
[0152] この幹細胞の集団を、分化誘導培地で培養する場合、 2— 3週間で骨、軟骨、脂肪 など臓器特異的なタンパク質の発現が認められた。この幹細胞の集団は、ヒト真皮由 来培養線維芽細胞とは異なり、線維芽細胞の多くの細胞で発現する CD56を、発現 しな力つた。逆に、この幹細胞の集団が発現する CD105の発現は、線維芽細胞で は、通常は見られな力つた。また、この幹細胞の集団が発現する CD49dの発現は、 骨髄由来間葉系幹細胞では通常は見られな力 た。  [0152] When this stem cell population was cultured in a differentiation-inducing medium, expression of organ-specific proteins such as bone, cartilage, and fat was observed in 2 to 3 weeks. This stem cell population, unlike human fibroblast-derived cultured fibroblasts, did not express CD56, which is expressed in many fibroblast cells. Conversely, the expression of CD105 expressed by this population of stem cells was not normally seen in fibroblasts. In addition, the expression of CD49d expressed by this stem cell population was not normally seen in bone marrow-derived mesenchymal stem cells.
[0153] また、 CD31、 CD34、 CD36、 CD45、 CD106、および CD117は培養期間が長く なると発現が無くなる傾向が見られた。そのため、継代培養を続けた場合、継代培養 前に見られた CD106発現が見られなくなる場合がある。  [0153] In addition, CD31, CD34, CD36, CD45, CD106, and CD117 tended to disappear when the culture period was prolonged. Therefore, when subculture is continued, CD106 expression observed before subculture may not be observed.
[0154] (実施例 5 :ヒト脂肪由来幹細胞の単離およびニューロスフェアの細胞培養)  [Example 5: Isolation of human adipose-derived stem cells and cell culture of neurospheres]
(ヒト脂肪由来幹細胞の単離)  (Isolation of human adipose-derived stem cells)
脂肪由来幹細胞を、 Yoshimura Kら、 J Cell Physiol 2006 ; 208 : 64— 76で 報告したように、ヒト吸引脂肪物力も単離した。まとめると、吸引脂肪 (suctioned fat )を、 PBS中、 0. 075%コラゲナーゼを用いて 37°Cで 30分間、シエイカー上で消化 した。成熟した脂肪細胞および結合糸且織を、遠心分離により取り除いた。血球をまた 、赤血球溶解緩衝液で処理することによって取り除き、脂肪由来幹細胞ペレットを得 た。代替的に、脂肪由来幹細胞を、赤血球溶解緩衝液で処理し、 FicolKGE Heal thcare Bio - sciences, Piscataway, NJ)を用いて密度勾配遠心分離することに より脂肪吸引物 (liposuction aspirate)の流体部分力も単離した。得られた細胞が 脂肪由来幹細胞であることは、細胞を、脂肪、軟骨、骨への分化誘導を実施する方 法に供することにより確認した。 Adipose-derived stem cells were also isolated from human aspirated fat as reported by Yoshimura K et al., J Cell Physiol 2006; 208: 64-76. In summary, suctioned fat was digested on a shaker for 30 minutes at 37 ° C with 0.075% collagenase in PBS. Mature adipocytes and connective yarns and weaves were removed by centrifugation. Blood cells again The fat-derived stem cell pellet was obtained by treatment with an erythrocyte lysis buffer. Alternatively, the fluid partial force of liposuction aspirate can also be achieved by treating fat-derived stem cells with erythrocyte lysis buffer and density gradient centrifugation using FicolKGE Healthcare Bio-sciences, Piscataway, NJ). Isolated. It was confirmed that the obtained cells were adipose-derived stem cells by subjecting the cells to a method for inducing differentiation into fat, cartilage, and bone.
[0155] (ニューロスフェアの細胞培養) [0155] (Cell culture of neurosphere)
ニューロスフェア培養には、 Kanemura Yら、 J Neurosci Res 2002 ; 69 : 869 — 879に記載された方法をわずかに改変した方法を用いた。新鮮な単離した脂肪由 来幹細胞を、 10cmのコートしていないディッシュに 2 X 107細胞の密度(約 1 X 105細 胞 Zml)でプレートし、湿潤下にて、 5% COの環境下にて 37°Cで、ニューロスフエ For neurosphere culture, a method slightly modified from the method described in Kanemura Y et al., J Neurosci Res 2002; 69: 869-879 was used. Freshly isolated adipose-derived stem cells are plated in 10 cm uncoated dishes at a density of 2 x 10 7 cells (approximately 1 x 10 5 cells Zml) and in a 5% CO environment under humidity. Underneath at 37 ° C, Neuros Hue
2  2
ァ培養培地において培養した。ニューロスフェア培養培地は、ヒト組換え EGF (20ng / ml, PeproTech)、ヒト糸且換 basic— FGF (20ngZ ml, Kaken Pharmaceutic al, Japan)、 2% B27 supplement (GIBCO)、 lOOUZmlペニシリンおよび 100 μ gZmlストレプトマイシンを補充した基礎培地としての DMEMZHAMF12 (1: 1) であった。培地の半分を、 4〜5日目に新しい培地に交換し、継代を 8日目に実施し た。  Cultured in a culture medium. Neurosphere culture medium consists of human recombinant EGF (20ng / ml, PeproTech), human thread and basic-FGF (20ngZ ml, Kaken Pharmaceutic al, Japan), 2% B27 supplement (GIBCO), lOOUZml penicillin and 100 μgZml DMEMZHAMF12 (1: 1) as a basal medium supplemented with streptomycin. Half of the medium was replaced with fresh medium on days 4-5 and passage was performed on day 8.
[0156] (結果)  [0156] (Result)
脂肪由来幹細胞を、ニューロスフェア培養培地 (血清を含まず、 EGFおよび basic FGFを含む)において培養すると、新しく調製した脂肪由来幹細胞の培養 3日目 に、浮遊する脂肪由来幹細胞は小さな塊を形成し始めた (データ示さず。 ) oニューロ スフエア様細胞の凝集は、 5日目に明ら力に観察された(図 1Aを参照のこと。 ) 0この スフエアの数およびサイズは、 2日以内により大きくなつた(図 1Bおよび 1Cを参照の こと。)。 8日目に継代した。この継代の際、スフエアを解離させ、新しい培地に再懸濁 した。継代から 7日目に、球状の細胞塊を新たに形成した (データ示さず。 ) oこのこと は、ニューロスフェアの自己複製能を示唆している。 When adipose-derived stem cells are cultured in a neurosphere culture medium (without serum, and containing EGF and basic FGF), the floating adipose-derived stem cells form small clumps on the third day of culture of the newly prepared adipose-derived stem cells. began (data not shown.) aggregation of o neuro Sufuea like cells were observed in bright et force on day 5 (see FIG. 1A.) 0 the number and size of the Sufuea, due within 2 days (See Figures 1B and 1C.) Passaged on the 8th day. During this passage, the spheres were dissociated and resuspended in fresh media. On day 7 after passage, a new globular cell mass formed (data not shown.) O This suggests the neurosphere's ability to self-replicate.
[0157] (実施例 6:定量的リアル タイム RT— PCR (逆転写酵素ポリメラーゼ鎖反応) ) 脂肪由来幹細胞を、 M199培地および 10%ゥシ胎仔血清(fetal bovine serum : FBS)を含む標準培地で予備培養した。次いで、全 mRNAを、 RNeasy— miniキッ ト(Qiagen, Hilden Germany)を用いて、一継代目の脂肪由来幹細胞に由来する ニューロスフェアカゝら抽出した。この予備培養は、血球の混入を減少させるのに必要 であった。コントロール mRNAもまた、 M199および 10%FBS中で培養した一継代 目の分ィ匕していない脂肪由来幹細胞力 抽出した。 [0157] (Example 6: Quantitative real-time RT-PCR (reverse transcriptase polymerase chain reaction)) Adipose-derived stem cells were treated with M199 medium and 10% fetal bovine serum. : Pre-cultured in a standard medium containing FBS). Next, total mRNA was extracted from Neurospora derived from the first passage of adipose-derived stem cells using an RNeasy-mini kit (Qiagen, Hilden Germany). This preculture was necessary to reduce blood cell contamination. Control mRNA was also extracted from undissolved adipose-derived stem cells cultured in M199 and 10% FBS.
[0158] 未分化神経幹細胞マーカー遺伝子 Nestinおよび Musashi— 1の発現、ならびに 脂肪生成分化マーカー Leptinの発現を、 ABI PRISM 7700 (Applied Biosyst ems, Foster City, CA)を用いて、慣習的なリアル—タイム定量的 RT— PCRによ り分析した。標的配列の遺伝子発現を、ハウスキーピング遺伝子 j8—ァクチンの発現 につ 、て正規ィ匕した。コントロール (未分化脂肪由来幹細胞)群における転写レベル を 1として表した。設計プライマーおよびプローブセットによる TaqManィ匕学およびァ ッセィを、ヒト Nestin、 Musashi— 1、 Leptinおよび j8—actinについて使用した。す ベてのプライマーおよびプローブのセットは、 Applied Bio systemsから購入した。 使用したプライマーセットは、以下のとおりである。 TaqMan Gene Expression Assaysシステム(Applied Biosystems)、 βァクチン: Hs99999903一 ml;ネス チン: Hs00707120— si ;ムサシ: Hs00159291— ml;レプチン: Hs001748 77— ml。  [0158] Expression of the undifferentiated neural stem cell marker genes Nestin and Musashi-1 and the expression of the adipogenic differentiation marker Leptin were performed using conventional real-time data using ABI PRISM 7700 (Applied Biosyst ems, Foster City, CA). Analyzed by quantitative RT-PCR. The gene expression of the target sequence was normalized for the expression of the housekeeping gene j8-actin. The transcription level in the control (undifferentiated adipose-derived stem cells) group was expressed as 1. TaqMan theory and assays with designed primers and probe sets were used for human Nestin, Musashi-1, Leptin and j8-actin. All primer and probe sets were purchased from Applied Bio systems. The primer sets used are as follows. TaqMan Gene Expression Assays system (Applied Biosystems), β-actin: 1 ml of Hs99999903; Nestin: Hs00707120-si; Musashi: Hs00159291-ml; Leptin: Hs001748 77-ml.
[0159] (結果)  [0159] (Result)
RT— PCRを用いて、神経幹細胞マーカー Nestinおよび Musashi— 1遺伝子、な らびに脂肪生成マーカー Leptinの発現を調べた結果、 Nestinおよび Musashi— 1 遺伝子の発現は、ニューロスフェア培養培地で培養して!/、な 、コントロール脂肪由来 幹細胞と比較して、ニューロスフェアにおいて著しくアップレギュレートした(図 2Aお よび Bを参照のこと。 ) oこの結果より、ニューロスフェアが神経前駆細胞の特徴を有 することが示唆される。対照的に、 Leptinの発現は、ニューロスフェアにおいて劇的 に減少した(図 2Cを参照のこと。 ) 0この結果より、ニューロスフェアの脂肪生成能力 が低下したことが示される。 RT-PCR was used to examine the expression of the neural stem cell markers Nestin and Musashi-1 genes and the adipogenic marker Leptin. The Nestin and Musashi-1 gene expression was cultured in neurosphere culture media! /, Well upregulated in neurospheres compared to control adipose-derived stem cells (see Figure 2A and B). O This result indicates that neurospheres have characteristics of neural progenitor cells Is suggested. In contrast, Leptin expression was dramatically reduced in neurospheres (see Figure 2C). 0 This result indicates that neurospheres have reduced adipogenic capacity.
[0160] (まとめ) [0160] (Summary)
本実施例において、脂肪由来幹細胞力 -ユーロスフェアが得られた。この-ユー ロスフェアの細胞増殖は著しく迅速であり、他の種々の器官 (例えば、真皮および心 臓)力 得られる-ユーロスフエアよりも速 、と考えられる。この著しく速 、細胞増殖は 、再生医療における神経前駆細胞の供給源としての、脂肪由来幹細胞の利用性を 示唆する。本実施例において、ニューロスフェアを構成する細胞は、神経幹細胞マ 一力一遺伝子である Nestinおよび Musaxhi—lを発現した。これは、ニューロスフエ ァの神経前駆細胞への分化傾向を反映しているものと考えられる。この知見は、脂肪 生成分ィ匕および成熟についてのマーカーである Leptinの発現の低下によりさらに支 持される。 In this example, adipose-derived stem cell force-eurosphere was obtained. This-you The cell growth of the rothsphere is remarkably rapid and can be achieved with a variety of other organs (eg, dermis and heart)-faster than the eurosphere. This remarkably fast cell growth suggests the availability of adipose-derived stem cells as a source of neural progenitor cells in regenerative medicine. In this example, the cells constituting the neurosphere expressed Nestin and Musaxhi-l, which are the genes of neural stem cells. This is thought to reflect the tendency of neurospheres to differentiate into neural progenitor cells. This finding is further supported by reduced expression of Leptin, a marker for adipogenesis and maturation.
[0161] (実施例 7:マウス全胚培養および-ユーロスフェア様細胞の移植)  [0161] (Example 7: Whole mouse embryo culture and transplantation of Eurosphere-like cells)
ヒト脂肪由来幹細胞由来の-ユーロスフェアを、 Li HOら、 J Virol 2000 ; 74 : 6 564— 6569および Inoue Mら、 J Virol 2003 ; 77 : 3238— 3246に記載された ように、 Sendaiウィルスベクター(DNAVEC corp. Tsukuba, Japan)を用いて緑 色蛍光タンパク質(green fluorescent protein : GFP)でトランスフエタトした。元 々のベクター SeVZ は、感染細胞へのリボヌクレオチド複合体の侵入に必要な 融合タンパク質をコードする F遺伝子を欠損するので、このベクターは、伝染性でも 病原性でもない。改変 SevZ A Fベクターは、細胞毒性を減少させるさらなる変異を 有する。この実施例では、改変ベクターを使用した。ニューロスフェアを、 GFP遺伝 子を保有する改変 SeVZ (感染の多重度 250)を含む培地において、 1時間イン キュペートし、次 、でリン酸緩衝ィ匕生理食塩水でリンスした。  Eurospheres derived from human adipose-derived stem cells were isolated from the Sendai virus vector (as described in Li HO et al., J Virol 2000; 74: 6 564-6565 and Inoue M et al., J Virol 2003; 77: 3238-3246). DNAVEC corp. Tsukuba, Japan) was used to transfect with green fluorescent protein (GFP). The original vector SeVZ lacks the F gene encoding the fusion protein required for entry of the ribonucleotide complex into infected cells, so this vector is neither infectious nor pathogenic. The modified SevZ A F vector has additional mutations that reduce cytotoxicity. In this example, a modified vector was used. Neurospheres were incubated for 1 hour in a medium containing modified SeVZ (multiplicity of infection of 250) carrying the GFP gene, and then rinsed with phosphate buffered saline.
[0162] マウス全胚培養を、以下に記載されるとおりに実施した: Nagase Tら、 J Anat 2 003 ; 203 : 77-88, Nagase Tら、 J Craniofac Surg 2005 ; 16 : 80-88, Na gase Tら、 Genes Cells 2005 ; 10 : 595— 604および Yamada Yら、 J Cranio fac Surg 2005 ; 16 : 1055 - 1061。  [0162] Mouse whole embryo culture was performed as described below: Nagase T et al., J Anat 2 003; 203: 77-88, Nagase T et al., J Craniofac Surg 2005; 16: 80-88, Na gase T et al., Genes Cells 2005; 10: 595-604 and Yamada Y et al., J Cranio fac Surg 2005; 16: 1055-1061.
[0163] 9匹のマウスの胚 (8日胚 (E8) )を、卵黄嚢を破壊することなく取り出した。次いで、 マイクロピペットを用いて、 GFPでトランスフエタトした-ユーロスフェアの細胞を胚の 頭部領域に移植した。これらの胚を、約 40時間培養し、 GFP陽性移植細胞が存在 するカゝ否かを、蛍光顕微鏡下で調べた。  [0163] Nine mouse embryos (8-day embryos (E8)) were removed without destroying the yolk sac. A micropipette was then used to transfer GFP-transferred eurosphere cells into the head region of the embryo. These embryos were cultured for about 40 hours and examined for the presence of GFP-positive transplanted cells under a fluorescence microscope.
[0164] (結果) マウス胚に移植した GFP陽性細胞は、 9つの胚のうち 2っ胚において明確に観察さ れ、生存したようであった。この GFP陽性細胞は、これら 2つの胚の頭蓋顔面領域な らびに心臓および体幹に組み込まれた(図 3を参照のこと。 )0特に、移植した細胞は 、第二鰓弓に沿って列になって並んだ(図 3Bおよび Cの矢印を参照のこと。 )0これ は、第二鰓弓内を遊走する神経堤細胞のパターンと非常に似ていた。この結果は、 脂肪由来幹細胞から誘導されたニューロスフェアの細胞が神経堤細胞様特性を有す るという、興味深い可能性を示唆している。 [0164] (Result) GFP positive cells transplanted into mouse embryos were clearly observed in 2 out of 9 embryos and seemed to survive. The GFP-positive cells were incorporated into the craniofacial region and the heart and trunk of these two embryos (see Figure 3). 0 In particular, the transplanted cells lined up along the second arch. (See arrows in Figures 3B and C.) 0 This was very similar to the pattern of neural crest cells migrating in the second arch. This result suggests an interesting possibility that neurosphere cells derived from adipose-derived stem cells have neural crest cell-like properties.
(まとめ)  (Summary)
ニューロスフェアを培養したマウス胚に移植するという試みは、これらの細胞のいく つかが、第二鰓弓にそって遊走し、頭蓋顔面形態形成に寄与することを示した。この 細胞の遊走パターンは、本発明者らが以前に報告 (Nagase Tら、 J Anat 2003 ; 203: 77-88)したように、頭蓋神経堤細胞の遊走パターンと非常に類似して ヽた。 この神経堤細胞は、広範な遊走および独特な分化レパートリーにより特徴付けられる 胚細胞集団である。神経堤細胞は、末梢神経細胞およびシュワン細胞についての幹 細胞または前駆細胞として、しばしば考えられている。頭蓋顔面骨格間葉はまた、神 経堤を誘導する。従って、本実施例により、移植した-ユーロスフェアは、インビボで 神経前駆細胞である神経堤細胞と同じ遊走能を有することが確認できた。  Attempts to transfer neurospheres into cultured mouse embryos have shown that some of these cells migrate along the second arch and contribute to craniofacial morphogenesis. This cell migration pattern was very similar to that of the cranial crest cells, as previously reported by the inventors (Nagase T et al., J Anat 2003; 203: 77-88). This neural crest cell is an embryonic cell population characterized by extensive migration and a unique differentiation repertoire. Neural crest cells are often considered as stem or progenitor cells for peripheral and Schwann cells. The craniofacial skeleton mesenchyme also guides the neural dyke. Therefore, according to this example, it was confirmed that the transplanted-eurosphere had the same migration ability as the neural crest cell which is a neural progenitor cell in vivo.
[0165] 本発明により誘導された-ユーロスフェアは、神経幹細胞マーカー遺伝子である Ne stinおよび Musashi— 1を発現したこと(実施例 6を参照のこと)、ならびに神経堤細 胞と同じ遊走能を有したことから、この-ユーロスフェアには神経堤細胞が含まれてい るといえる。 [0165] The eurospheres induced by the present invention expressed the neural stem cell marker genes Ne stin and Musashi-1 (see Example 6), and had the same migration ability as neural crest cells. Therefore, it can be said that this -Eurosphere contains neural crest cells.
[0166] 通常、神経幹細胞力 -ユーロスフェアを形成するには、約 1ヶ月近くの期間を必要 とするが、本実施例により得られたヒト脂肪由来幹細胞は、約 1週間で-ユーロスフエ ァを形成することが観察された。さらに、本発明のヒト脂肪由来幹細胞から形成された ニューロスフ アは、神経堤細胞、脂肪由来幹細胞および神経幹細胞を含む細胞混 合物であることが確認された。本発明の細胞混合物はまた、継代を重ねても約 1週間 で-ユーロスフェアを形成することができた。  [0166] Normally, it takes a period of about one month to form neural stem cell force-eurosphere, but the human adipose-derived stem cells obtained by this example were able to produce -eurosphere in about one week. It was observed to form. Furthermore, it was confirmed that the neurosphere formed from the human adipose-derived stem cells of the present invention is a cell mixture containing neural crest cells, adipose-derived stem cells, and neural stem cells. The cell mixture of the present invention was also able to form eurospheres in about one week even after repeated passages.
[0167] 以上の結果より、本発明の細胞混合物は、顕著に速い速度で増殖し-ユーロスフエ ァを形成する能力を有し、かつ、神経堤細胞を含むことが確認できた。 [0167] From the above results, the cell mixture of the present invention proliferates at a remarkably fast rate. It was confirmed that it has the ability to form a and includes neural crest cells.
[0168] 従って、本発明の脂肪由来または脂肪由来幹細胞由来の神経堤細胞は、神経系 疾患、障害または状態を処置するための再生医療に応用可能であると考えられる。  Therefore, the neural crest cells derived from fat or adipose-derived stem cells of the present invention are considered to be applicable to regenerative medicine for treating nervous system diseases, disorders or conditions.
[0169] (実施例 8 :マウス脂肪由来幹細胞の単離およびニューロスフ アの細胞培養) [Example 8: Isolation of mouse adipose-derived stem cells and cell culture of neurospheres]
(マウス脂肪由来幹細胞の単離)  (Isolation of mouse adipose-derived stem cells)
検体としてヒトのかわりにマウスを用いること以外、実施例 5と同様の方法を用いて、 マウス脂肪由来幹細胞を単離する。このマウス脂肪由来幹細胞について、実施例 3と 同様の方法により、細胞の特徴づけを実施する。  Mouse adipose-derived stem cells are isolated using the same method as in Example 5 except that mice are used instead of humans as specimens. The mouse adipose-derived stem cells are characterized by the same method as in Example 3.
[0170] (ニューロスフェアの細胞培養) [0170] (Cell culture of neurosphere)
実施例 5と同様の方法を用いて、マウス脂肪由来幹細胞をニューロスフェア培養培 地において培養する。培地の半分を、 4〜5日目に新しい培地に交換し、継代を 8日 目に実施する。  Using the same method as in Example 5, mouse adipose-derived stem cells are cultured in a neurosphere culture medium. Half of the medium is replaced with fresh medium on days 4-5 and passage is performed on day 8.
[0171] 脂肪由来幹細胞を、ニューロスフェア培養培地 (血清を含まず、 EGFおよび basic  [0171] Adipose-derived stem cells were treated with neurosphere culture medium (serum-free, EGF and basic
FGFを含む)において培養すると、浮遊する脂肪由来幹細胞は小さな塊を形成し 始めることが確認される。ニューロスフェア様細胞の凝集が観察される。このスフエア の数およびサイズは、次第に大きくなる。継代後、球状の細胞塊を新たに形成するこ とが確認される。このことは、ニューロスフェアの自己複製能を示唆する。  When cultured in (including FGF), it is confirmed that floating adipose-derived stem cells begin to form small clumps. Aggregation of neurosphere-like cells is observed. The number and size of this sphere will gradually increase. After passage, it is confirmed that a new spherical cell mass is formed. This suggests the neurosphere's ability to self-replicate.
[0172] (実施例 9:定量的リアル タイム RT— PCR (逆転写酵素ポリメラーゼ鎖反応) ) 実施例 8において培養したマウス-ユーロスフェアを用いること以外、実施例 6と同 様の方法を用いて、未分化神経幹細胞マーカー遺伝子 Nestinおよび Musashi— 1 の発現、ならびに脂肪生成分ィ匕マーカー Leptinの発現を分析する。  (Example 9: Quantitative real-time RT—PCR (reverse transcriptase polymerase chain reaction)) Using the same method as in Example 6, except that the mouse-eurosphere cultured in Example 8 was used. Then, the expression of undifferentiated neural stem cell marker genes Nestin and Musashi-1 and the expression of the adipogenic marker Leptin are analyzed.
[0173] Nestinおよび Musashi—l遺伝子の発現は、ニューロスフェア培養培地で培養し て ヽな 、コントロール脂肪由来幹細胞と比較して、ニューロスフェアにぉ 、て著しくァ ップレギュレートすることを確認する。この結果より、ニューロスフェアが神経前駆細胞 の特徴を有することが示される。対照的に、 Leptinの発現は、ニューロスフェアにお いて劇的に減少することを確認する。この結果より、ニューロスフェアの脂肪生成能力 が低下したことが示される。  [0173] It is confirmed that the expression of the Nestin and Musashi-l genes is significantly up-regulated in the neurosphere compared with the control fat-derived stem cells cultured in the neurosphere culture medium. This result indicates that the neurosphere has the characteristics of neural progenitor cells. In contrast, we confirm that Leptin expression decreases dramatically in neurospheres. This result indicates that the neurosphere's ability to produce fat has decreased.
[0174] (実施例 10:マウス全胚培養およびマウス-ユーロスフェア様細胞の移植) 実施例 8において培養したマウス- ロスフェアを用いること以外、実施例 7と同 様の方法を用いる。 (Example 10: Mouse whole embryo culture and mouse-eurosphere-like cell transplantation) A method similar to that in Example 7 is used except that the mouse-spheres cultured in Example 8 are used.
[0175] マウス胚に移植したマウス GFP陽性細胞は、移植した胚のうちいくつかにおいて明 らかに観察され、そして生存したようである。この GFP陽性細胞は、胚の頭蓋顔面領 域ならびに心臓および体幹に組み込まれることが確認できる。特に、移植した細胞は 、第二鰓弓に沿って列になって並び、これは、第二鰓弓内を遊走する神経堤細胞の ターンと非常に似ていることが確認できる。この結果は、マウス脂肪由来幹細胞か ら誘導された- ロスフェアの細胞がマウス胚において神経堤細胞様特性を呈す るという、興味深い可能性を示唆する。  [0175] Mouse GFP positive cells transplanted into mouse embryos are clearly observed in some of the transplanted embryos and appear to survive. This GFP positive cell can be confirmed to be incorporated into the craniofacial region of the embryo and the heart and trunk. In particular, the transplanted cells are arranged in a row along the second arch, which can be confirmed to be very similar to the turn of the neural crest cells that migrate in the second arch. This result suggests an interesting possibility that the cells of murine adipose-derived stem cells exhibit neuronal crest-like properties in mouse embryos.
[0176] 通常、神経幹細胞力 - ロスフェアを形成するには、約 1ヶ月近くの期間を必要 とするが、本実施例により得られたマウス脂肪由来幹細胞は、約 1週間で- ロス フェアを形成することが観察される。さらに、本発明のマウス脂肪由来幹細胞力 形 成されたニューロスフ アは、神経堤細胞、脂肪由来幹細胞および神経幹細胞を含 む細胞混合物であることが確認される。本発明の細胞混合物はまた、継代を重ねて も約 1週間で- ロスフェアを形成することができる。  [0176] Normally, it takes a period of about one month to form neural stem cell force-loss spheres, but the mouse adipose-derived stem cells obtained in this example form the spheres in about one week. To be observed. Furthermore, it is confirmed that the neurosphere formed with the mouse adipose-derived stem cell force of the present invention is a cell mixture containing neural crest cells, adipose-derived stem cells and neural stem cells. The cell mixture of the present invention can also form a microsphere in about 1 week even after repeated passages.
[0177] 以上の結果より、本発明の細胞混合物は、顕著に速い速度で増殖し- ロスフエ ァを形成する能力を有し、かつ、神経堤細胞を含むことが確認できる。  [0177] From the above results, it can be confirmed that the cell mixture of the present invention has the ability to proliferate at a remarkably fast rate to form a rosphae and contains neural crest cells.
[0178] 従って、本発明のマウス脂肪由来幹細胞力 誘導される神経堤細胞は、同種であ るマウス胚への移植においても利用され得ることが理解される。  Accordingly, it is understood that the neural crest cells in which the mouse adipose-derived stem cell force of the present invention is induced can also be used for transplantation into mouse embryos of the same species.
[0179] (実施例 11 ユーロスフエアの細胞培養における培地の影響)  (Example 11: Effect of medium in cell culture of Eurosphere)
ニューロスフェア培養に用いる培地を、 DMEMZHAMF12 (1 : 1)を用いる代わり に、 BME MEM DMEMまたは HAMF12培地を用いて、培地が- ロスフエ ァの細胞培養に与える影響について検討する。それ以外の材料および方法は、実 施例 5と同じものを用いる。  Instead of using DMEMZHAMF12 (1: 1) as the medium for neurosphere culture, use BME MEM DMEM or HAMF12 medium to examine the effect of the medium on cell culture of rosophane. The other materials and methods are the same as in Example 5.
[0180] 脂肪由来幹細胞を、ニューロスフェア培養培地 (血清を含まず、 EGFおよび basic  [0180] Adipose-derived stem cells were treated with neurosphere culture medium (serum-free, EGF and basic
FGFを含む)において培養すると、浮遊する脂肪由来幹細胞は小さな塊を形成し 始めることが確認される。ニューロスフェア様細胞の凝集が観察される。このスフエア の数およびサイズは、次第に大きくなる。継代後、球状の細胞塊を新たに形成するこ とが確認される。このことは、ニューロスフェアの自己複製能を示唆する。 When cultured in (including FGF), it is confirmed that floating adipose-derived stem cells begin to form small clumps. Aggregation of neurosphere-like cells is observed. The number and size of this sphere will gradually increase. After passage, a new spherical cell mass is formed. Is confirmed. This suggests the neurosphere's ability to self-replicate.
[0181] (定量的リアル タイム RT— PCR (逆転写酵素ポリメラーゼ鎖反応))  [0181] (Quantitative real-time RT—PCR (reverse transcriptase polymerase chain reaction))
本実施例において培養したマウス-ユーロスフェアを用いること以外、実施例 6と同 様の方法を用いて、未分化神経幹細胞マーカー遺伝子 Nestinおよび Musashi— 1 の発現、ならびに脂肪生成分ィ匕マーカー Leptinの発現を分析する。  Except for using mouse-eurosphere cultured in this example, expression of undifferentiated neural stem cell marker genes Nestin and Musashi-1 and expression of adipogenic 分 marker Leptin were performed using the same method as in Example 6. Analyze expression.
[0182] Nestinおよび Musashi—l遺伝子の発現は、ニューロスフェア培養培地で培養し て ヽな 、コントロール脂肪由来幹細胞と比較して、ニューロスフェアにぉ 、て著しくァ ップレギュレートすることを確認する。この結果より、ニューロスフェアが神経前駆細胞 の特徴を有することが示される。対照的に、 Leptinの発現は、ニューロスフェアにお いて劇的に減少することを確認する。この結果より、ニューロスフェアの脂肪生成能力 が低下したことが示される。  [0182] It is confirmed that Nestin and Musashi-l gene expression is significantly up-regulated in neurospheres compared to control adipose-derived stem cells cultured in neurosphere culture medium. This result indicates that the neurosphere has the characteristics of neural progenitor cells. In contrast, we confirm that Leptin expression decreases dramatically in neurospheres. This result indicates that the neurosphere's ability to produce fat has decreased.
[0183] (マウス全胚培養および-ユーロスフェア様細胞の移植) 本実施例において培養 した-ユーロスフェアを用いること以外、実施例 7と同様の方法を用いる。  (Mouse whole embryo culture and transplantation of eurosphere-like cells) [0183] The same method as in Example 7 is used, except that the eurosphere cultured in this example is used.
[0184] マウス胚に移植した GFP陽性細胞は、移植した胚のうちいくつかにおいて明らかに 観察され、そして生存したようである。この GFP陽性細胞は、胚の頭蓋顔面領域なら びに心臓および体幹に組み込まれることが確認できる。特に、移植した細胞は、第二 鰓弓に沿って列になって並び、これは、第二鰓弓内を遊走する神経堤細胞のパター ンと非常に似ていることが確認できる。この結果は、脂肪由来幹細胞力 誘導された ニューロスフ アの細胞が神経堤細胞様特性を有すると 1ヽぅ、興味深!/ヽ可能性を示 唆する。  [0184] GFP positive cells transplanted into mouse embryos are clearly observed in some of the transplanted embryos and appear to survive. It can be confirmed that the GFP positive cells are incorporated into the craniofacial region of the embryo and the heart and trunk. In particular, the transplanted cells are arranged in a row along the second arch, which can be confirmed to be very similar to the pattern of neural crest cells that migrate in the second arch. This result suggests that the neurosphere cells induced by adipose-derived stem cell force have a neural crest cell-like property, which is interesting and possible.
[0185] (まとめ) [0185] (Summary)
以上の結果より、 BME、 MEM、 DMEMまたは HAMF12培地のいずれの培地で 培養した-ユーロスフェアを用いた場合でも、 DMEM/HAMF12 (1: 1)培地を用 V、た場合と同様の結果を示すことが確認できる。  Based on the above results, cultivated in any medium of BME, MEM, DMEM or HAMF12-Even when using Eurospheres, the results are the same as when using VMEM with DMEM / HAMF12 (1: 1) medium I can confirm that.
[0186] (実施例 12:維持培養した脂肪由来幹細胞の効果) (Example 12: Effect of maintenance-cultured adipose-derived stem cells)
実施例 5にお 、て単離したヒト脂肪由来幹細胞を DMEM培地中で 5回継代し、二 ユーロスフエアを調製し培養する。  In Example 5, the human adipose-derived stem cells isolated in the above are passaged 5 times in DMEM medium to prepare and culture eurosphere.
[0187] (定量的リアル タイム RT—PCR) 本実施例の- ロスフェアを用いること以外、実施例 6と同様の方法を用いて、神 経幹細胞マーカー Nestinおよび Musashi— 1遺伝子、ならびに脂肪生成マーカー Leptinの発現を分析する。 [0187] (Quantitative real-time RT—PCR) The expression of the neural stem cell marker Nestin and Musashi-1 gene and the adipogenic marker Leptin is analyzed using the same method as in Example 6 except that the spheres of this example are used.
[0188] Nestinおよび Musashi—l遺伝子の発現は、ニューロスフェア培養培地で培養し て ヽな 、コントロール脂肪由来幹細胞と比較して、ニューロスフェアにぉ 、て著しくァ ップレギュレートすることを確認する。この結果より、ニューロスフェアが神経前駆細胞 の特徴を有することが示される。対照的に、 Leptinの発現は、ニューロスフェアにお いて劇的に減少することを確認する。この結果より、ニューロスフェアの脂肪生成能力 が低下したことが示される。  [0188] It is confirmed that the expression of the Nestin and Musashi-l genes is significantly up-regulated in the neurosphere compared with the control fat-derived stem cells cultured in the neurosphere culture medium. This result indicates that the neurosphere has the characteristics of neural progenitor cells. In contrast, we confirm that Leptin expression decreases dramatically in neurospheres. This result indicates that the neurosphere's ability to produce fat has decreased.
[0189] (全胚培養およびマウス- ロスフェア様細胞の移植)  [0189] (Whole embryo culture and transplantation of mouse-rothsphere-like cells)
本実施例の- ロスフェアを用いること以外、実施例 7と同様の方法を用いる。  The same method as in Example 7 is used except that the spheres of this example are used.
[0190] マウス胚に移植したマウス GFP陽性細胞は、移植した胚のうちいくつかにおいて明 らかに観察され、そして生存したようである。この GFP陽性細胞は、胚の頭蓋顔面領 域ならびに心臓および体幹に組み込まれることが確認できる。特に、移植した細胞は 、第二鰓弓に沿って列になって並び、これは、第二鰓弓内を遊走する神経堤細胞の ターンと非常に似ていることが確認できる。この結果は、マウス脂肪由来幹細胞か ら誘導された- ロスフェアの細胞がマウス胚において神経堤細胞様特性を呈す るという、興味深い可能性を示唆する。  [0190] Mouse GFP positive cells transplanted into mouse embryos are clearly observed in some of the transplanted embryos and appear to survive. This GFP positive cell can be confirmed to be incorporated into the craniofacial region of the embryo and the heart and trunk. In particular, the transplanted cells are arranged in a row along the second arch, which can be confirmed to be very similar to the turn of the neural crest cells that migrate in the second arch. This result suggests an interesting possibility that the cells of murine adipose-derived stem cells exhibit neuronal crest-like properties in mouse embryos.
[0191] (まとめ)  [0191] (Summary)
以上の結果より、脂肪由来幹細胞は、取得後、培養し増殖させて維持したものであ つても用いることができることがわ力つた。さらに、培養し増殖させた脂肪由来幹細胞 からも- ロスフェアが誘導され、神経堤細胞を調製することが可能であることが確 認される。  From the above results, it was proved that adipose-derived stem cells can be used even if they are cultured and expanded and maintained after acquisition. Furthermore, it is confirmed that neurospheres can be prepared from adipose-derived stem cells cultured and proliferated, and neural crest cells can be prepared.
[0192] (実施例 13 : -ユーロスフエアにおける神経堤細胞マーカーの発現)  [Example 13: -Expression of neural crest cell marker in Eurosphere]
実施例 5 8および 11において調製した- ロスフェアを用いて、実施例 6と同様 の定量的リアル タイム RT— PCR法を使用して、神経堤細胞マーカー遺伝子 CRA BP1 (NM004378 (ヒト))、 AP2 (NM002097 (ヒト))、 Slug (NM003068 (ヒト))、 SoxlO (NM006941 (ヒ卜))、 Snail (NM005985 (ヒ卜))、 Twist (NM000474 (ヒ卜 ) )、 Pax3 (NM000438 (ヒト) )、 Pax7 (NM002584 (ヒト) )、 HNKl (NM004854 (ヒト) )、 p75NTR (NM002507 (ヒト) )、 TRP2 (NM006267 (ヒト) )、 Wntl (NM0 05430 (ヒト) )、 PO (NM002723 (ヒト) )、 tPA (NM000930 (ヒト) )の発現を分析す る。 Prepared in Examples 5 8 and 11-Using the spheres, the quantitative real-time RT-PCR method similar to that in Example 6 was used to determine the neural crest cell marker gene CRA BP1 (NM004378 (human)), AP2 ( NM002097 (human)), Slug (NM003068 (human)), SoxlO (NM006941 (chicken)), Snail (NM005985 (chicken)), Twist (NM000474 (chicken) )), Pax3 (NM000438 (human)), Pax7 (NM002584 (human)), HNKl (NM004854 (human)), p75NTR (NM002507 (human)), TRP2 (NM006267 (human)), Wntl (NM0 05430 (human)) ), PO (NM002723 (human)), tPA (NM000930 (human)) are analyzed.
[0193] CRABP1 (NM004378 (ヒト))、 AP2 (NM002097 (ヒト))、 Slug (NM003068 ( ヒ卜))、 SoxlO (NM006941 (ヒ卜))、 Snail(NM005985 (ヒ卜))、 Twist (NM0004 74 (ヒト))、 Pax3 (NM000438 (ヒト))、 Pax7 (NM002584 (ヒト))、 HNKl (NMO 04854 (ヒト))、 p75NTR (NM002507 (ヒト))、 TRP2 (NM006267 (ヒト))、 Wntl (NM005430 (ヒト) )、 PO (NM002723 (ヒト) )、 tPA (NM000930 (ヒト) )遺伝子の 発現は、ニューロスフェア培養培地で培養していないコントロール脂肪由来幹細胞と 比較して、ニューロスフェアにおいて著しくアップレギュレートすることを確認する。こ の結果より、ニューロスフェアが神経堤細胞マーカーを発現することが示される。  [0193] CRABP1 (NM004378 (human)), AP2 (NM002097 (human)), Slug (NM003068 (human)), SoxlO (NM006941 (human)), Snail (NM005985 (human)), Twist (NM0004 74) (Human)), Pax3 (NM000438 (human)), Pax7 (NM002584 (human)), HNKl (NMO 04854 (human)), p75NTR (NM002507 (human)), TRP2 (NM006267 (human)), Wntl (NM005430 ( (Human)), PO (NM002723 (Human)), tPA (NM000930 (Human)) gene expression is significantly up-regulated in neurospheres compared to control adipose-derived stem cells not cultured in neurosphere culture medium Make sure. This result indicates that neurospheres express neural crest cell markers.
[0194] 各マーカーに対して特異的な染色法、免疫組織化学的な手法、 in situハイブリダ ィゼーシヨン法、ウェスタンブロッテイング法によっても、ニューロスフェアが神経堤細 胞マーカーを発現することが確認できる。  [0194] It is also possible to confirm that neurospheres express neural crest cell markers by specific staining methods, immunohistochemical methods, in situ hybridization methods, and Western blotting methods for each marker.
[0195] 以上のように、本発明の好ましい実施形態を用いて本発明を例示してきた力 本発 明は、特許請求の範囲によってのみその範囲が解釈されるべきであることが理解され る。本明細書において引用した特許、特許出願および文献は、その内容自体が具体 的に本明細書に記載されているのと同様にその内容が本明細書に対する参考として 援用されるべきである。  [0195] As described above, it is understood that the scope of the present invention should be construed only by the scope of the claims by exemplifying the present invention using the preferred embodiments of the present invention. The patents, patent applications, and literature cited herein are to be incorporated by reference in their entirety, as if the contents themselves were specifically described herein.
産業上の利用可能性  Industrial applicability
[0196] 本発明は、簡便な方法で取得できる脂肪由来または脂肪由来幹細胞由来神経堤 細胞が、再生医療に応用できることを証明した。したがって、本発明の産業上の利用 は、医薬品業界において見出される。 [0196] The present invention proves that fat-derived or fat-derived stem cell-derived neural crest cells that can be obtained by a simple method can be applied to regenerative medicine. Therefore, the industrial use of the present invention is found in the pharmaceutical industry.

Claims

請求の範囲  The scope of the claims
[I] 脂肪由来の神経堤細胞。  [I] Neural crest cells derived from fat.
[2] 脂肪由来幹細胞力 誘導された、請求項 1に記載の神経堤細胞。  [2] The neural crest cell according to claim 1, induced by adipose-derived stem cell force.
[3] 前記神経堤細胞は、遊走能を有する、請求項 1に記載の神経堤細胞。  [3] The neural crest cell according to claim 1, wherein the neural crest cell has a migration ability.
[4] 前記遊走能は、胚の頭部領域に移植し、その後の前頭鼻隆起、上顎隆起または鰓 弓部の間充織への遊走能を観察することによって確認される、請求項 3に記載の神 経堤細胞。  [4] The method according to claim 3, wherein the migration ability is confirmed by transplanting to the head region of the embryo and then observing the migration ability to the mesenchyme of the frontal nasal bump, maxillary bump, or arch. The described neural dyke cell.
[5] 前記神経堤細胞は、神経幹細胞マーカーを少なくとも 1つ発現することを特徴とする [5] The neural crest cell expresses at least one neural stem cell marker
、請求項 1に記載の神経堤細胞。 The neural crest cell according to claim 1.
[6] 前記神経幹細胞マーカーは、 Nestin、 Musashi— 1、 CD133、 notch 1, Hesl、 M ashl、 Neurogenin、 Pax6、 CD15および PDGFRからなる群より選択される、請求 項 5に記載の神経堤細胞。 6. The neural crest cell according to claim 5, wherein the neural stem cell marker is selected from the group consisting of Nestin, Musashi-1, CD133, notch1, Hesl, Mashl, Neurogenin, Pax6, CD15 and PDGFR.
[7] 前記神経幹細胞マーカーは、 Nestinおよび Musashi— 1を含む、請求項 6に記載 の神経堤細胞。 [7] The neural crest cell according to claim 6, wherein the neural stem cell marker includes Nestin and Musashi-1.
[8] 前記神経堤細胞は、神経堤細胞マーカーを少なくとも 1つ発現することを特徴とする [8] The neural crest cell expresses at least one neural crest cell marker
、請求項 1に記載の神経堤細胞。 The neural crest cell according to claim 1.
[9] 前記神経堤細胞マーカーは、 CRABP1、 AP2、 Slug, SoxlO、 Snail, Twist, Pax[9] The neural crest cell marker is CRABP1, AP2, Slug, SoxlO, Snail, Twist, Pax
3、 Pax7、 HNK1、 p75NTR、 TRP2、 Wntl、 POおよび tP A力もなる群より選択さ れる、請求項 8に記載の神経堤細胞。 The neural crest cell according to claim 8, which is selected from the group consisting of 3, Pax7, HNK1, p75NTR, TRP2, Wntl, PO and tPA force.
[10] 前記神経堤細胞は、 Leptinの発現が脂肪由来幹細胞のものの発現レベルよりも低[10] In the neural crest cells, Leptin expression is lower than that of adipose-derived stem cells.
Vヽことを特徴とする、請求項 1に記載の神経堤細胞。 The neural crest cell according to claim 1, characterized in that it is V ヽ.
[II] 前記神経堤細胞は、 Nestinおよび Musashi— 1を発現し、かつ、 Leptinの発現が 脂肪由来前駆細胞のものの発現レベルよりも低いことを特徴とする、請求項 1に記載 の神経堤細胞。  [II] The neural crest cell according to claim 1, wherein the neural crest cell expresses Nestin and Musashi-1, and the expression level of Leptin is lower than that of an adipose-derived progenitor cell. .
[12] 前記脂肪は、哺乳動物由来である、請求項 1に記載の神経堤細胞。  12. The neural crest cell according to claim 1, wherein the fat is derived from a mammal.
[13] 前記脂肪は、ヒト由来である、請求項 12に記載の神経堤細胞。 13. The neural crest cell according to claim 12, wherein the fat is derived from a human.
[14] 神経堤細胞を調製するための方法であって、以下の工程: [14] A method for preparing neural crest cells comprising the following steps:
A)脂肪力も幹細胞を得る工程;および B)該幹細胞を、神経堤細胞を誘導する条件に供する工程 A) the step of obtaining fat stem cells as well B) Step of subjecting the stem cells to conditions for inducing neural crest cells
を包含する、方法。  Including the method.
[15] 前記前幹細胞は、脂肪由来幹細胞(adipose— derived stem or stromal cell) であることを特徴とする、請求項 14に記載の方法。  15. The method according to claim 14, wherein the anterior stem cell is an adipose-derived stem or stromal cell.
[16] 前記幹細胞を得る工程は、コラゲナーゼ処理および遠心分離処理を包含する、請求 項 14に記載の方法。 16. The method according to claim 14, wherein the step of obtaining stem cells includes collagenase treatment and centrifugation treatment.
[17] 前記幹細胞は、 PLA細胞および LAF細胞力 なる群より選択される、請求項 14に記 載の方法。  17. The method according to claim 14, wherein the stem cell is selected from the group consisting of PLA cell and LAF cell force.
[18] 前記 B)工程において、前記脂肪由来前駆細胞が、約 1 X 104細胞/ ml〜約 1 X 106 細胞 Zmlの密度で播種される、請求項 14に記載の方法。 [18] The method according to claim 14, wherein in step B), the adipose-derived progenitor cells are seeded at a density of about 1 × 10 4 cells / ml to about 1 × 10 6 cells Zml.
[19] 前記 B)工程において、前記脂肪由来幹細胞が、約 1 X 105細胞 Zmlの密度で播種 される、請求項 18に記載の方法。 [19] The method according to claim 18, wherein in the step B), the adipose-derived stem cells are seeded at a density of about 1 × 10 5 cells Zml.
[20] 前記 B)工程において、前記脂肪由来幹細胞が、少なくとも上皮増殖因子 (EGF)を 含む培地中で培養される、請求項 14に記載の方法。  [20] The method according to claim 14, wherein in step B), the adipose-derived stem cells are cultured in a medium containing at least epidermal growth factor (EGF).
[21] 前記 B)工程において、前記脂肪由来幹細胞が、補助成分として、上皮増殖因子、 塩基性線維芽細胞増殖因子、神経誘導因子、ペニシリンおよびストレプトマイシンを 含む培地中で培養される、請求項 14に記載の方法。  [21] The step (B), wherein the adipose-derived stem cells are cultured in a medium containing epidermal growth factor, basic fibroblast growth factor, nerve induction factor, penicillin and streptomycin as auxiliary components. The method described in 1.
[22] 前記神経誘導因子が、ピオチン、 L—カル-チン、コルチコステロン、エタノールアミ ン、 D ( + ) ガラクトース、ダルタチオン (還元型)、リノール酸、リノレン酸、プロゲステ ロン、レチ-ノレアセテート、セレン、トリョードサイロニン(T3)、 DL— a トコフェロー ル、 DL— a—トコフエロールアセテート、アルブミン(ゥシ)、カタラーゼ、インスリン、 スーパーォキシドジスムターゼ、トランスフェリンを含む、請求項 21に記載の方法。  [22] The nerve-inducing factor is piotin, L-carcin, corticosterone, ethanolamine, D (+) galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, retin-noreacetate , Selenium, triodothyronine (T3), DL-a tocopherol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin The method described.
[23] 前記 B)工程にぉ 、て、前記幹細胞を培養するために用いられる培地力 グルタミン 酸およびァスパラギン酸を含む、請求項 14に記載の方法。  [23] The method according to claim 14, wherein the step B) comprises medium strength glutamic acid and aspartic acid used for culturing the stem cells.
[24] 前記 B)工程にぉ 、て、前記幹細胞を培養するために用いられる培地力 イーグル基 礎培地(BME)、最小必須培地(MEM)、ダルベッコ改変イーグル培地(DMEM)ま たは HAMF12培地、あるいはそれらの混合培地である、請求項 14に記載の方法。  [24] Medium power used for culturing the stem cells after the step B) Eagle basic medium (BME), minimum essential medium (MEM), Dulbecco's modified Eagle medium (DMEM) or HAMF12 medium The method according to claim 14, or a mixed medium thereof.
[25] 前記 B)工程にぉ 、て、前記幹細胞を培養するために用いられる培地力 ダルベッコ 改変イーグル培地 (DMEM) /F12 (l: 1)である、請求項 14に記載の方法。 [25] Medium power Dulbecco used for culturing the stem cells in the step B) 15. The method according to claim 14, wherein the modified eagle medium (DMEM) / F12 (l: 1).
[26] 前記 B)工程は、 2〜6日目に培地を新しいものに交換することを包含することを特徴 とする、請求項 14に記載の方法。 [26] The method according to claim 14, wherein the step B) includes replacing the medium with a new medium on days 2 to 6.
[27] 前記 B)工程は、 4〜5日目に培地を新しいものに交換することを包含することを特徴 とする、請求項 14に記載の方法。 [27] The method according to claim 14, wherein the step B) comprises replacing the medium with a new one on the 4th to 5th days.
[28] 前記 B)工程は、 6〜10日目〖こ継代することを包含することを特徴とする、請求項 14 に記載の方法。 [28] The method according to claim 14, wherein the step B) includes passaging on day 6 to day 10.
[29] 前記脂肪は、哺乳動物由来である、請求項 14に記載の方法。  [29] The method of claim 14, wherein the fat is derived from a mammal.
[30] 前記脂肪は、ヒト由来である、請求項 29に記載の方法。 [30] The method of claim 29, wherein the fat is derived from a human.
[31] 前記 B)工程において、前記脂肪由来幹細胞は、ヒト由来であり、 [31] In the step B), the fat-derived stem cells are human-derived,
該脂肪由来幹細胞は、約 1 X 105細胞 Zmlの密度で播種され、 The adipose-derived stem cells are seeded at a density of about 1 × 10 5 cells Zml;
該脂肪由来幹細胞は、補助成分として、上皮増殖因子、塩基性線維芽細胞増殖 因子、神経誘導因子、ペニシリンおよびストレプトマイシンを含む培地中で培養され、 ここで、該神経誘導因子は、ピオチン、 L—カル-チン、コルチコステロン、エタノール ァミン、 D ( + )—ガラクトース、ダルタチオン (還元型)、リノール酸、リノレン酸、プロゲ ステロン、レチュルアセテート、セレン、トリョードサイロニン(T3)、 DL— a—トコフエ ロール、 DL— a—トコフエロールアセテート、ァルブミン(ゥシ)、カタラーゼ、インスリ ン、スーパーォキシドジスムターゼ、トランスフェリンを含み、そして  The adipose-derived stem cells are cultured in a medium containing epidermal growth factor, basic fibroblast growth factor, nerve inducer, penicillin and streptomycin as auxiliary components, wherein the nerve inducer comprises piotin, L— Carcin, corticosterone, ethanolamine, D (+) —galactose, dartathione (reduced form), linoleic acid, linolenic acid, progesterone, lettyl acetate, selenium, tolydothyronine (T3), DL— a-tocopherol, DL-a-tocopherol acetate, albumin (usi), catalase, insulin, superoxide dismutase, transferrin, and
該培地は、ダルベッコ改変イーグル培地(DMEM) ZF12 (1: 1)である、請求項 1 4に記載の方法。  15. The method according to claim 14, wherein the medium is Dulbecco's Modified Eagle Medium (DMEM) ZF12 (1: 1).
[32] 前記幹細胞が神経幹細胞マーカーを発現しているかどうかを確認する工程をさらに 包含する、請求項 14に記載の方法。  [32] The method according to claim 14, further comprising the step of confirming whether the stem cells express a neural stem cell marker.
[33] 前記神経幹細胞マーカーは、 Nestin、 Musashi— 1、 CD133、 notch 1, Hesl、 M ashl、 Neurogenin、 Pax6、 CD15および PDGFRからなる群より選択される、請求 項 32に記載の方法。 [33] The method according to claim 32, wherein the neural stem cell marker is selected from the group consisting of Nestin, Musashi-1, CD133, notch1, Hesl, Mashl, Neurogenin, Pax6, CD15, and PDGFR.
[34] 前記神経幹細胞マーカーは、 Nestinおよび Musashi— 1を含む、請求項 33に記載 の方法。  [34] The method according to claim 33, wherein the neural stem cell marker comprises Nestin and Musashi-1.
[35] 前記幹細胞が神経堤細胞マーカーを発現しているかどうかを確認する工程をさらに 包含する、請求項 14に記載の方法。 [35] The step of confirming whether the stem cell expresses a neural crest cell marker is further included. 15. The method of claim 14, comprising.
[36] 前記細胞は、神経堤細胞マーカーを少なくとも 1つ発現することを特徴とする、請求 項 14に記載の方法。 [36] The method according to claim 14, wherein the cell expresses at least one neural crest cell marker.
[37] 前記神経堤細胞マーカーは、 CRABP1、 AP2、 Slug, SoxlO、 Snail, Twist, Pax 3、 Pax7、 HNK1、 p75NTR、 TRP2、 Wntl、 POおよび tP A力もなる群より選択さ れる、請求項 36に記載の方法。  [37] The neural crest cell marker is selected from the group consisting of CRABP1, AP2, Slug, SoxlO, Snail, Twist, Pax3, Pax7, HNK1, p75NTR, TRP2, Wntl, PO, and tPA force. The method described in 1.
[38] 前記幹細胞が脂肪細胞マーカーを発現のレベルを確認する工程をさらに包含する、 請求項 14に記載の方法。 [38] The method according to claim 14, further comprising the step of confirming the level of expression of the adipocyte marker in the stem cells.
[39] 前記脂肪細胞マーカーのレベルの確認は、 Leptinの減少の確認である、請求項 38 に記載の方法。 [39] The method according to claim 38, wherein the confirmation of the level of the adipocyte marker is confirmation of a decrease in Leptin.
[40] 前記神経堤細胞は、 B)工程により、神経堤細胞と脂肪由来幹細胞とを含む脂肪由 来神経堤細胞前駆体となることを特徴とする、請求項 14に記載の方法。  [40] The method according to claim 14, wherein the neural crest cells become a fat-derived neural crest cell precursor containing neural crest cells and adipose-derived stem cells by the step B).
[41] 前記幹細胞が神経幹細胞マーカーおよび神経堤細胞マーカーを発現して 、るかど うかを確認する工程、および前記幹細胞が脂肪細胞マーカーを発現のレベルを確認 する工程をさらに包含する、請求項 14に記載の方法。 [41] The method further includes the step of confirming whether the stem cell expresses a neural stem cell marker and a neural crest cell marker, and the step of confirming the level of expression of the adipocyte marker by the stem cell. The method described in 1.
[42] 前記幹細胞が遊走能を有するかどうかを確認する工程をさらに包含する、請求項 14 に記載の方法。 [42] The method according to claim 14, further comprising the step of confirming whether or not the stem cells have migration ability.
[43] 前記遊走能は、胚の頭部領域に移植し、その後の遊走能を観察することによって確 認される、請求項 42に記載の方法。  [43] The method according to claim 42, wherein the migration ability is confirmed by transplanting to a head region of an embryo and observing the subsequent migration ability.
[44] 前記神経幹細胞マーカーは、 Nestinおよび Musashi— 1であり、かつ、前記脂肪細 胞マーカーは、 Leptinであることを特徴とする、請求項 41に記載の方法。 [44] The method according to claim 41, wherein the neural stem cell marker is Nestin and Musashi-1, and the fat cell marker is Leptin.
[45] 請求項 1に記載の細胞を含む、神経系疾患、障害または状態の処置のための細胞 移植のための組成物。 [45] A composition for cell transplantation for the treatment of a nervous system disease, disorder or condition, comprising the cell of claim 1.
[46] 前記神経系疾患、障害または状態は、神経系の分化細胞の欠損に起因する疾患、 障害または状態である、請求項 45に記載の組成物。  [46] The composition according to claim 45, wherein the nervous system disease, disorder or condition is a disease, disorder or condition caused by a defect in differentiated cells of the nervous system.
[47] 前記脂肪は、前記神経系疾患、障害または状態の対象の個体と同種異系の関係に ある個体由来である、請求項 45に記載の組成物。 [47] The composition according to claim 45, wherein the fat is derived from an individual having an allogeneic relationship with an individual subject to the nervous system disease, disorder or condition.
[48] 前記脂肪は、前記神経系疾患、障害または状態の対象の個体と異種の関係にある 個体由来である、請求項 45に記載の組成物。 [48] The fat is in a heterogeneous relationship with the individual subject to the nervous system disease, disorder or condition 46. The composition of claim 45, wherein the composition is derived from an individual.
[49] 前記脂肪は、前記神経系疾患、障害または状態の対象の個体と同種同系の関係に ある個体由来である、請求項 45に記載の組成物。 [49] The composition according to claim 45, wherein the fat is derived from an individual having an allogeneic relationship with an individual subject to the nervous system disease, disorder or condition.
[50] 請求項 1に記載の細胞を投与する工程を包含する、神経系疾患、障害または状態の 処置のための細胞移植のための方法。 [50] A method for cell transplantation for the treatment of a nervous system disease, disorder or condition comprising the step of administering the cell of claim 1.
[51] 前記神経系疾患、障害または状態は、神経系の分化細胞の欠損に起因する疾患、 障害または状態である、請求項 50に記載の方法。 [51] The method according to claim 50, wherein the nervous system disease, disorder or condition is a disease, disorder or condition caused by a defect in differentiated cells of the nervous system.
[52] 請求項 1に記載の細胞の、神経系疾患、障害または状態を処置または予防するため の医薬の調製のための使用。 [52] Use of the cell of claim 1 for the preparation of a medicament for treating or preventing a nervous system disease, disorder or condition.
[53] 前記神経系疾患、障害または状態は、神経系の分化細胞の欠損に起因する疾患、 障害または状態である、請求項 52に記載の使用。 [53] The use according to claim 52, wherein the nervous system disease, disorder or condition is a disease, disorder or condition caused by a defect in differentiated cells of the nervous system.
[54] 神経堤細胞と脂肪由来幹細胞とを含む、脂肪由来神経堤細胞混合物。 [54] A fat-derived neural crest cell mixture comprising neural crest cells and adipose-derived stem cells.
[55] 神経幹細胞をさらに含む、請求項 54に記載の脂肪由来神経堤細胞混合物。 [55] The fat-derived neural crest cell mixture according to claim 54, further comprising neural stem cells.
PCT/JP2007/051643 2006-08-09 2007-01-31 Fat-derived neural crest cells WO2008018190A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2006-217286 2006-08-09
JP2006217286 2006-08-09

Publications (1)

Publication Number Publication Date
WO2008018190A1 true WO2008018190A1 (en) 2008-02-14

Family

ID=39032730

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/051643 WO2008018190A1 (en) 2006-08-09 2007-01-31 Fat-derived neural crest cells

Country Status (1)

Country Link
WO (1) WO2008018190A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008150001A1 (en) * 2007-06-08 2008-12-11 Biomaster, Inc. Adipocluster
WO2010150650A1 (en) * 2009-06-23 2010-12-29 学校法人日本大学 Novel method for maintaining stem cells in an undifferentiated state
US20110110900A1 (en) * 2008-06-13 2011-05-12 Philip Stephens Novel adult progenitor cell
JP2011520434A (en) * 2008-05-07 2011-07-21 ボーン セラピューティクス エス.アー. Novel mesenchymal stem cells and osteogenic cells
JP2013063088A (en) * 2012-12-14 2013-04-11 Nippon Institute For Biological Science Method for inducing differentiation to neurocyte from adipose tissue stromal cells
JP2013532961A (en) * 2010-05-25 2013-08-22 メモリアル スローン−ケタリング キャンサー センター Methods and uses thereof for nociceptor differentiation of human embryonic stem cells
CN103751845A (en) * 2014-01-30 2014-04-30 中国人民解放军海军总医院 Tissue engineering biological material for transplanting and restoring peripheral nerve defect
JP5700301B2 (en) * 2009-06-03 2015-04-15 国立大学法人大阪大学 Method for inducing differentiation of neural crest cells from pluripotent stem cells
EP2952579A4 (en) * 2013-01-31 2016-07-20 Ajinomoto Kk Culture method for stable undifferentiated proliferation of pluripotent stem cells
US10260041B2 (en) 2009-02-17 2019-04-16 Memorial Sloan Kettering Cancer Center Methods for neural conversion of human embryonic stem cells
US10280398B2 (en) 2011-11-04 2019-05-07 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (DA) neurons for engraftment
CN111304167A (en) * 2018-12-12 2020-06-19 上海泉眼生物科技有限公司 Neuron precursor cell derived from human adipose-derived stem cell and preparation method and application thereof
US11649431B2 (en) 2013-04-26 2023-05-16 Memorial Sloan-Kettering Cancer Center Cortical interneurons and other neuronal cells produced by the directed differentiation of pluripotent and multipotent cells
US11959100B2 (en) 2017-11-30 2024-04-16 Kyoto University Method for culture of cells
US11970712B2 (en) 2020-06-03 2024-04-30 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (DA) neurons for engraftment

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
NAGASE T. ET AL.: "Neurosphere from human adipose tissue transplanted into cultured mouse embryos can contribute to craniofacial morphogenesis: a preliminary report", J. CRANIOFAC. SURG., vol. 18, no. 1, January 2007 (2007-01-01), pages 49 - 53, XP003020988 *
PIERRET C. ET AL.: "Neural crest at the source of adults stem cells", STEM CELLS DEV., vol. 15, no. 2, April 2006 (2006-04-01), pages 286 - 291, XP003020987 *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008150001A1 (en) * 2007-06-08 2008-12-11 Biomaster, Inc. Adipocluster
JP5388297B2 (en) * 2007-06-08 2014-01-15 株式会社バイオマスター Adipo cluster
JP2011520434A (en) * 2008-05-07 2011-07-21 ボーン セラピューティクス エス.アー. Novel mesenchymal stem cells and osteogenic cells
US9371515B2 (en) 2008-05-07 2016-06-21 Bone Therapeutics S.A. Mesenchymal stem cells and bone-forming cells
JP2017099386A (en) * 2008-05-07 2017-06-08 ボーン セラピューティクス エス.アー. Novel mesenchymal stem cells and bone-forming cells
US20110110900A1 (en) * 2008-06-13 2011-05-12 Philip Stephens Novel adult progenitor cell
US11560546B2 (en) 2009-02-17 2023-01-24 Memorial Sloan Kettering Cancer Center Methods for neural conversion of human embryonic stem cells
US10287546B2 (en) 2009-02-17 2019-05-14 Memorial Sloan Kettering Cancer Center Kits for neural conversion of human embryonic stem cells
US10260041B2 (en) 2009-02-17 2019-04-16 Memorial Sloan Kettering Cancer Center Methods for neural conversion of human embryonic stem cells
JP5700301B2 (en) * 2009-06-03 2015-04-15 国立大学法人大阪大学 Method for inducing differentiation of neural crest cells from pluripotent stem cells
WO2010150650A1 (en) * 2009-06-23 2010-12-29 学校法人日本大学 Novel method for maintaining stem cells in an undifferentiated state
JP2011004607A (en) * 2009-06-23 2011-01-13 Nihon Univ New method for maintaining stem cell in undifferentiated state
US9453198B2 (en) 2010-05-25 2016-09-27 Memorial Sloan Kettering Cancer Center Method of nociceptor differentiation of human embryonic stem cells and uses thereof
JP2013532961A (en) * 2010-05-25 2013-08-22 メモリアル スローン−ケタリング キャンサー センター Methods and uses thereof for nociceptor differentiation of human embryonic stem cells
US10711243B2 (en) 2011-11-04 2020-07-14 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (DA) neurons for engraftment
US10280398B2 (en) 2011-11-04 2019-05-07 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (DA) neurons for engraftment
JP2013063088A (en) * 2012-12-14 2013-04-11 Nippon Institute For Biological Science Method for inducing differentiation to neurocyte from adipose tissue stromal cells
US10662411B2 (en) 2013-01-31 2020-05-26 Ajinomoto Co., Inc. Culture method for stable proliferation of pluripotent stem cell while maintaining undifferentiated state
EP2952579A4 (en) * 2013-01-31 2016-07-20 Ajinomoto Kk Culture method for stable undifferentiated proliferation of pluripotent stem cells
US10745669B2 (en) 2013-01-31 2020-08-18 Ajinomoto Co., Ltd. Culture method for stable proliferation of pluripotent stem cell while maintaining undifferentiated state
US11649431B2 (en) 2013-04-26 2023-05-16 Memorial Sloan-Kettering Cancer Center Cortical interneurons and other neuronal cells produced by the directed differentiation of pluripotent and multipotent cells
CN103751845A (en) * 2014-01-30 2014-04-30 中国人民解放军海军总医院 Tissue engineering biological material for transplanting and restoring peripheral nerve defect
US11959100B2 (en) 2017-11-30 2024-04-16 Kyoto University Method for culture of cells
CN111304167A (en) * 2018-12-12 2020-06-19 上海泉眼生物科技有限公司 Neuron precursor cell derived from human adipose-derived stem cell and preparation method and application thereof
CN111304167B (en) * 2018-12-12 2024-03-26 上海泉眼生物科技有限公司 Neuron precursor cells derived from human adipose-derived stem cells, and preparation method and application thereof
US11970712B2 (en) 2020-06-03 2024-04-30 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (DA) neurons for engraftment

Similar Documents

Publication Publication Date Title
WO2008018190A1 (en) Fat-derived neural crest cells
JP4749331B2 (en) Cell differentiation of adipose-derived progenitor cells
CN101748096B (en) Sub totipotential stem cell and preparation method and application thereof
JP6478243B2 (en) Method for culturing mesenchymal stem cells
US20190367883A1 (en) Regulating stem cells
CA2667959C (en) Lithium stimulation of cord blood stem cell proliferation and growth factor production
BRPI0709349A2 (en) methods for cell expansion and uses of cells and conditioned media produced through them for therapy
JP2007509601A (en) Method and system for preparing stem cells from adipose tissue
US20080286246A1 (en) Internally administered therapeutic agents for cranial nerve diseases comprising mesenchymal cells as an active ingredient
WO2012133948A1 (en) Composition for allotransplantation cell therapy, said composition containing ssea-3 positive pluripotent stem cell capable of being isolated from body tissue
US20140105871A1 (en) Use Of Mesenchymal Stem Cells For The Improvement Of Affective And Cognitive Function
JP5388297B2 (en) Adipo cluster
JP2009536163A (en) Immunological tolerance and regulatory progenitor cells
KR101920277B1 (en) Method of treating the effects of stroke
US9650604B2 (en) Equine amniotic membrane-derived mesenchymal stem cells
CN102703380B (en) Sub-totipotent stem cell, preparation method and application thereof
US20140286910A1 (en) Stem cells and methods incorporating environmental factors as a means for enhancing stem cell proliferation and plasticity
JP2022120698A (en) Pharmaceutical composition for regeneration of soft tissue that contains cell population containing mesenchymal stem cell
AU2013203149B2 (en) Lithium stimulation of cord blood stem cell proliferation and growth factor production
KR20210046196A (en) Mesenchymal stem cell originated from equine amniotic membrane and its use
Marshall II Neurospheres and multipotent astrocytic stem cells: Neural progenitor cells rather than neural stem cells
Howell Identification of a common pluripotent stem cell population derived from multiple murine tissues

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: 07707829

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: RU

NENP Non-entry into the national phase

Ref country code: JP

122 Ep: pct application non-entry in european phase

Ref document number: 07707829

Country of ref document: EP

Kind code of ref document: A1