WO2005040361A1 - Method of simply preparing stem cell and feeder cell to be used therein - Google Patents

Method of simply preparing stem cell and feeder cell to be used therein Download PDF

Info

Publication number
WO2005040361A1
WO2005040361A1 PCT/JP2004/015854 JP2004015854W WO2005040361A1 WO 2005040361 A1 WO2005040361 A1 WO 2005040361A1 JP 2004015854 W JP2004015854 W JP 2004015854W WO 2005040361 A1 WO2005040361 A1 WO 2005040361A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
cells
feeder
cell preparation
stem
Prior art date
Application number
PCT/JP2004/015854
Other languages
French (fr)
Japanese (ja)
Inventor
Norio Nakatsuji
Hirofumi Suemori
Original Assignee
Kyoto University
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 Kyoto University filed Critical Kyoto University
Priority to JP2005515006A priority Critical patent/JPWO2005040361A1/en
Publication of WO2005040361A1 publication Critical patent/WO2005040361A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3895Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells using specific culture conditions, e.g. stimulating differentiation of stem cells, pulsatile flow conditions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • A61L27/3804Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells characterised by specific cells or progenitors thereof, e.g. fibroblasts, connective tissue cells, kidney cells
    • A61L27/3834Cells able to produce different cell types, e.g. hematopoietic stem cells, mesenchymal stem cells, marrow stromal cells, embryonic stem cells
    • 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
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]

Definitions

  • the present invention is in the field of stem cells. More specifically, the present invention relates to a method for simple preparation and storage of stem cells (particularly, primate embryonic stem cells (hereinafter, also referred to as ES cells)).
  • stem cells particularly, primate embryonic stem cells (hereinafter, also referred to as ES cells)).
  • Non-Patent Document 1 Stem cells, especially ES cells, which are at the center of regenerative medicine, have recently gained importance due to their potential in medical applications.
  • Human ES cells are particularly important among them, but their establishment or stable subculture maintenance is difficult, and they hinder efficient use.
  • Primate ES cells, including humans, require a very small supply of blastocysts to establish them. A small number of blastocysts need to efficiently establish ES cell lines. According to the conventional method, the efficiency of the establishment from the blastocyst is at most about 10 to 30%.
  • ES cells are obtained by culturing cells of the inner cell mass (ICM), which is an undifferentiated cell group that will be future animal individuals and is present in embryos at the developmental stage, usually called blastocysts. The resulting cell line.
  • ICM inner cell mass
  • ES cells were established in 1981 by M. J. Evans and M. H. Kaufman (Non-Patent Document 2), followed by G. R. Martin (Non-Patent Document 3) as a cell line possibly having a dangling ability in mice.
  • Non-Patent Document 4 is a paper using fibroblasts of human skin as a feeder, and is found to be 1 The force that establishes one strain from two inner cell masses. Its efficiency is quite low.
  • Non-Patent Document 5 is the first paper on human ES cells. Its establishment efficiency is 5 cell lines inside Z14. In Non-Patent Document 6, human ES cells are established using STO cells. However, the efficiency of the establishment is still very low, such as the internal cell mass of three strains Z14. In this way, in terms of the establishment efficiency, it is expected that 50% will be a downsized force and 30% will be achieved.
  • Patent Document 1 the present inventors have studied the optimal conditions for a medium. However, optimizing the medium alone has a limitation in increasing the establishment efficiency, and differentiated cells are significantly present in established ES cells.
  • Patent Document 2 Non-Patent Documents 5 and 7
  • Non-Patent Document 7 only one kind of feeder cell is used, and differentiating cells are always contaminated.
  • Non-Patent Document 7 about 50% is a partially divided cell population.
  • Patent Document 1 JP 2003-116527A
  • Patent Document 2 US Patent No. 6200806
  • Non-patent document 1 Stem cell 'clone research protocol Nakatsuji ed., Yodosha (2001)
  • Non-patent document 2 M.J.Evans & M.H.Kaufman: Nature, 292, 15
  • Non-Patent Document 3 G.R.Martin: Proc. Natl. Acad. Sci. USA, 78, 7634, 1981
  • Non-Patent Document 4 Hovatta O., et al., Human Reproduction Vol. 18, No. 7 pp. 1404-1409, 2003
  • Non-Patent Document 5 Thomson A., et al., Science.282: 1145-1147, 1998.
  • Non-Patent Document 6 Park JH et al., Biology of Reproduction, 2003 onl ine Aug. 20.
  • Non-Patent Document 7 Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A, Nat Biotechnol. 2000 Apr; 18 (4): 399-404
  • the present invention provides a technique capable of establishing stem cells (especially ES cells of primates including humans) with an efficiency (particularly, reducing contamination of differentiated cells) that cannot be achieved by conventional techniques.
  • the task is to provide
  • the present invention provides an unexpectedly efficient use of a conventionally used feeder cell by mixing a plurality of feeder cells (eg, primary cultured fibroblast cells, fibroblast cell lines, etc.) and using them as a feeder cell.
  • feeder cells eg, primary cultured fibroblast cells, fibroblast cell lines, etc.
  • the present invention provides the following.
  • the present invention provides a feeder-one cell preparation for preparing a stem cell, comprising a normal cell and a cell line.
  • the normal cells include primary cultured cells.
  • the normal cells are non-immortalized cells.
  • the normal cells include fibroblasts.
  • the normal cells are primary cultured fibroblast cells.
  • the normal cells are derived from a mouse.
  • the normal cells are derived from a fetus.
  • the normal cells have a passage number of 5 or less.
  • the normal cells are fibroblasts obtained by enzymatic treatment of mouse 11-16 days old fetal power.
  • the normal cells are primary cultured cells having a passage number of 5 or less.
  • the cell line is immortalized. [0025] In one embodiment, the cell line is a fibroblast cell line.
  • the cell line has been subcultured for 2 months or less.
  • the cell line has been passaged for one month or less.
  • the cell line is derived from a mouse.
  • the cell line is derived from a fetus.
  • the cell line is an STO line.
  • the cell line is neo-resistant.
  • the cell line is the SL10 line.
  • the ratio of the normal cells to the cell line is about 1:10 to 10: 1.
  • the normal cells and the cell line are about 1: 3-3: 1.
  • the normal cells and the cell line are present in approximately equal amounts.
  • the stem cells are embryonic stem cells.
  • the stem cells are human embryonic stem cells.
  • the normal cells are mouse fibroblast primary culture cells, and the cell line is SL10 strain.
  • the present invention provides a method for preparing a stem cell, comprising a step of culturing the stem cell on a feeder-one-cell preparation containing a normal cell and a cell line. .
  • the feeder-one-cell preparation can take any form of the embodiment described above.
  • the culture dish on which the feeder-cell preparation is placed is gelatin-coated.
  • the feeder single cell preparations is plated at about 1 X 10 4 cells / cm 2 one about 1 X 10 5 cells / cm 2.
  • the feeder-one cell preparation is mixed and seeded. Used within 5 days.
  • the normal cells in the feeder-one-cell preparation are mouse fibroblast primary culture cells, and the cell line is SL10 strain.
  • the stem cells are cultured in a medium containing a knockout serum replacement additive (KSR).
  • KSR knockout serum replacement additive
  • the method further includes a sub-passing step, wherein collagenase is used in the sub-passing step.
  • the present invention provides a stem cell preparation prepared by the method of the present invention.
  • the present invention provides an ES cell preparation prepared by the method of the present invention.
  • the present invention provides a primate ES cell preparation prepared by the method of the present invention.
  • the present invention provides a human ES cell preparation prepared by the method of the present invention.
  • the present invention provides a method for preparing a transplant for regenerating an organ, tissue or cell, comprising: A) a stem cell capable of differentiating into a desired organ, tissue or cell. B) culturing the stem cells on a feeder cell containing normal cells and a cell line; and C) transforming the stem cells into a transplant for regenerating a desired organ, tissue or cell. Differentiating.
  • the feeder-cell preparation can take any form of the embodiments described in the feeder-cell preparation.
  • the desired organ, tissue or cell is a nerve, blood cell, bone, cartilage, heart, pericardium, blood vessel, muscle, eye, liver, spleen, intestine, stomach, Lung, trachea, hair and skin.
  • the desired organ, fibrous tissue or cell and the feeder cell are of the same species.
  • the desired organ, tissue or cell is a primate.
  • the feeder cells are derived from a mouse.
  • the culturing is performed ex vivo.
  • the stem cell is a force that is also obtained immediately after the removal of the test subject, or is a cryopreserved one.
  • the culturing is performed at 37 ° C in a saturated humidity under 5% CO.
  • the differentiation is performed by using a DNA demethylating agent, a histone deacetylating agent, a nuclear receptor ligand, a cell growth factor, a cytokinin, hexamethylene bisacetamide.
  • the present invention provides an organ, tissue or cell prepared by the method for preparing a transplant for regenerating an organ, tissue or cell of the present invention.
  • the present invention provides a system for regenerating an organ, tissue or cell, comprising: A) a container; and B) a normal cell and a cell line seeded on the container. And a feeder cell containing the same.
  • the feeder-cell preparation can take any form of the embodiment described in the feeder-cell preparation.
  • the present invention provides a mixture of a normal cell and a cell line.
  • the mixture comprises primary fibroblast cells and a fibroblast cell line.
  • the primary cultured cells are mouse primary cultured fibroblasts.
  • the fibroblast cell line is an STO line.
  • the normal cells are mouse fibroblast primary culture cells
  • the cell line is the SL10 strain.
  • the present invention relates to a method for regenerating an organ, fibrous tissue or cell, comprising: A) providing a stem cell capable of dividing into a desired organ, tissue or cell; B) a step of culturing the stem cells on a feeder cell containing normal cells and a cell line; and C) a step of transplanting the cultured stem cells to a site of the subject to be treated.
  • a method is provided.
  • the feeder-cell preparation can take any form of the embodiments described in the feeder-cell preparation.
  • the present invention further includes D) a step of differentiating the stem cells.
  • the present invention provides use of a cell mixture containing primary fibroblast cells and a fibroblast cell line as a feeder cell.
  • the present invention provides use of a cell mixture containing a primary fibroblast cell line and a fibroblast cell line for producing a medicament containing a feeder cell.
  • the present invention provides the use of a cell mixture containing normal cells and a cell line as a feeder-cell preparation.
  • the feeder-cell preparation can take any form of the embodiment described in the feeder-cell preparation.
  • the feeder cell preparation is treated so as not to proliferate.
  • the feeder cell preparation is treated with mitomycin C.
  • the present invention provides a feeder comprising a normal cell and a cell line in the manufacture of a medicament containing a stem cell for treating or preventing a disease, disorder or condition in which the use of the stem cell is appropriate.
  • a feeder comprising a normal cell and a cell line in the manufacture of a medicament containing a stem cell for treating or preventing a disease, disorder or condition in which the use of the stem cell is appropriate.
  • the use of a single cell preparation is provided.
  • the feeder-cell preparation can take any form of the embodiments described in the feeder-cell preparation.
  • the feeder-cell preparation is treated so as not to proliferate! It is.
  • the feeder-cell preparation is treated with mitomycin C.
  • the present invention provides a technique for efficiently establishing stem cells in particular.
  • stem cell preparation prepared using the feeder cell of the present invention contains less than 50% of differentiated cells, and can achieve a low rate of less than 10% or less than about 1%. Became.
  • the present invention provides a powerful and highly pure preparation of pluripotent cells that could not be achieved conventionally.
  • FIG. 1A is an explanatory photograph of a monkey ES cell line established by the method of the present invention. No. 1 indicates 100 / zm.
  • FIG. 1B is an explanatory photograph of a monkey ES cell line established by this method. Bars indicate 50 ⁇ m.
  • FIG. 2A is a photograph of human ES cells. Bars indicate 275 ⁇ m.
  • FIG. 2B is a photograph of human ES cells. The bar indicates 70 m.
  • FIG. 3 shows undifferentiated state-specific marker expression.
  • FIG. 4 is a diagram illustrating the division of nerve cells.
  • FIG. 5 is a proof of the supposedly cosmic ability by teratoma formation.
  • FIG. 6 shows an example of an ES cell establishment scheme.
  • cell is defined in the same broadest sense as used in the art, and is a constituent unit of a tissue of a multicellular organism, wrapped in a membrane structure that isolates the outside world. Rarely, it refers to an organism that has a self-renewal ability inside and has genetic information and its expression mechanism. In the present invention, any cell can be targeted.
  • the number of "cells” used in the present invention can be counted through a light microscope. When counting through an optical microscope, count by counting the number of nuclei.
  • the tissue is used as a slice of a tissue section, and extracellular matrix (for example, elastin or collagen) and nuclei derived from cells are stained with a dye by hematoxylin-eosin (HE) staining.
  • This tissue section can be examined under an optical microscope, and the number of nuclei per specific area (for example, 200 m ⁇ 200 m) can be counted by estimating the number of cells.
  • the cells used herein may be naturally occurring cells or artificially modified cells (eg, fusion cells, genetically modified cells).
  • the source of cells can be, for example, a single cell culture, or a source of cells from a normally grown transgenic animal embryo, blood, or body tissue, or a normally grown cell line. Such cell mixtures include, but are not limited to.
  • the cells used as the feeder cells used in the present invention include normal cells and cell lines, and preferably, cells derived from any organism as long as fibroblasts or their counterparts are present.
  • mammals e.g., monotremes, marsupials, oligodonts, skin wings, winged fins, carnivores, carnivores, long nose, odd ungulates, artiodactyla, tubulars, Cells derived from squamous species, marine species, cetaceans, whales, primates, rodents, apogonidae, etc.
  • mammals e.g., monotremes, marsupials, oligodonts, skin wings, winged fins, carnivores, carnivores, long nose, odd ungulates, artiodactyla, tubulars
  • cells from primates eg, chimpanzees, macaques, humans
  • Cells are used, but are not limited thereto.
  • the feeder cell is a rodent cell, and more preferably, a mouse cell is used as the feeder cell.
  • the cells used as stem cells used in the present invention include cells derived from any organism (eg, mammals (eg, monopores, marsupials, oligodentates, Skin wings, winged hands, carnivores, carnivores, longnoses, hoofed horses, artiodactyla, tube teeth, scales, squids, cetaceans, primates, primates, rodents, etc. ) -Derived cells) may be used. More preferably, cells derived from primates (for example, chimpanzees, Japanese macaques, and humans) are used as stem cells. Most preferably, human-derived cells are used as the stem cells.
  • mammals eg, monopores, marsupials, oligodentates, Skin wings, winged hands, carnivores, carnivores, longnoses, hoofed horses, artiodactyla, tube teeth, scales, squids, cetaceans, primates, prima
  • counterpart refers to a cell in a certain organism, and refers to a cell of another species having similar properties and Z or function. Such similar properties can be identified by secreted cytokins, growth factors, expressed cellular markers, and the like.
  • stem cell has a self-renewal ability and a pluripotency (that is,
  • Stem cells can usually regenerate tissue when it is damaged.
  • a stem cell can be, but is not limited to, an ES cell or a tissue stem cell (also a tissue stem cell, a tissue-specific stem cell, or a somatic stem cell).
  • artificially created cells eg, reprogrammed cells, etc.
  • embryonic stem cells and “ES cells” are used interchangeably and refer to any pluripotent stem cells derived from an early embryo. Generally, ES cells are considered to be totipotent or nearly totipotent.
  • a germline chimera (a chimeric mouse having functional germ cells derived from ES cells) having high chimera-forming ability was obtained. ) was obtained (A. Bradley et al .: Nature, 309, 255, 1984).
  • Various gene transfer methods for example, a calcium phosphate method, a retrovirus vector method, a ribosome method, an electoral poration method, etc.
  • a method to select cells into which genes have been incorporated We also devised a method to select cells into which genes have been incorporated.
  • the transgenic mouse production method using ES cells has many advantages over the transgenic animal production method using microinjection in that it makes it possible to obtain individuals in which only certain genes have been arbitrarily modified. Benefits are conceivable.
  • a knockout animal in which a specific gene has been inactivated can be produced, and the function of the gene can be elucidated, or only the exogenous gene can be expressed. Therefore, if the establishment of ES cells is facilitated, the effect is immeasurable.
  • tissue stem cell refers to a cell that has a limited direction of differentiation, unlike ES cells. Normally, tissue stem cells are present at specific positions in a tissue and have an undifferentiated intracellular structure. Thus, tissue stem cells have a low level of pluripotency. Tissue stem cells are poor in organelles with a high nuclear Z cytoplasmic ratio. Tissue stem cells generally have a probable ability to maintain their proliferative capacity over the life of an individual whose cell cycle is slow. As used herein, ES cells are preferably used as stem cells, but tissue stem cells may also be the object of maintenance.
  • ES cell lines are extremely important in producing chimeric mice, knockout mice, and the like, and the analysis of gene function has been dramatically advanced by these techniques.
  • the development of a system for inducing specific fibrous tissue from ES cells has been progressing, and the system has been applied to transplantation medicine.
  • preclinical research using ES cells from appropriate model animals such as monkeys is considered to be almost as important as essential. Standing is also important.
  • ES cells are established by culturing a blastocyst-separated inner cell mass (ICM) on a single feeder cell.
  • ICM inner cell mass
  • mouse fetal fibroblasts or cell line STO derived therefrom have conventionally been used alone.
  • this efficiency varies depending on the stage of the blastocyst used, the ICM separation method, and the like. Therefore, in the current system with low establishment efficiency, several trials and errors were necessary to succeed in establishment. Using the present invention, the efficiency is over 80%, the contamination of differentiated cells is drastically reduced, and the need for such trial and error is eliminated.
  • mice In terms of mice, embryos usually develop into blastocysts 3.5 days after fertilization.
  • the inner cell mass which is an undifferentiated stem cell forming the embryo body, is separated from the blastocyst by immunosurgery.
  • This ICM has the ability to differentiate into trophectoderm, a cell made by the placenta and the like. Therefore, if ICM is directly isolated from 3.5-day embryos, undifferentiated cell strength S is often lost due to the differentiation of ICM into trophectoderm. 3.
  • the blastocyst escapes the zona pellucida. At this stage, the ICM no longer differentiates into trophectoderm.
  • the ratio of undifferentiated cells can be increased. Therefore, even when stem cells are established in the present invention, undivided cells having such a number of days can be used.
  • the number of days can be used as a reference and used as appropriate. In the case of humans, those that are 5-8 days after fertilization (excluding the freezing period) are often used, but are not limited thereto.
  • blastocysts can be cultured directly on feeder cells, but in those cases, blastocysts adhere and nutritive ectoderm extends. Exposure of the ICM is also less favorable. A culture obtained by separating and culturing this may be used, but immunosurgery is preferably used.
  • Fig. 6 shows a schematic ES cell line establishment scheme. Briefly, ICM isolated by immunosurgery is cultured on a single feeder cell, and as the passage is continued, undifferentiated stem cell colonies appear in the mixed cells. By separating and subculturing this, an ES cell line that can be stably maintained can be established. According to the present invention, since the establishment rate exceeds 80%, the efficiency of obtaining ES cells that can be stably maintained is significantly improved.
  • the feeder cell of the present invention was used to prepare the cells of the feeder, PBS (containing no Ca and Mg), 0.25% trypsin 'ImM EDTA in PBS, a medium for ES cells (DMEM ( noisy course) 400ml, fetal calf serum 100ml, non-essential amino acid solution (Gibco) 4ml, Nucle Oside solution (nucleoside, guanosine, cytidine, peridine 3 mM each, thymidine ImM aqueous solution, dissolve by heating at 40 ° C, filter sterilize, and store at 20 ° C) 4 ml, 2-mercaptoethanol 4 1 (0.
  • DMEM Noigram course
  • ImM LIF 100 ⁇ l (2000 U / ml)), ⁇ 2 medium (available from Sigma etc.), M16 medium (available from Sigma etc.), acidic Tyrode's solution (available from Sigma etc.), anti-mouse serum (5 x 10 8 mouse spleen cells or lymphocytes are immunized intravenously three times every 2 weeks into a egret, and blood is collected 2 weeks after the final immunization. This is inactivated as an antiserum and stored at 80 ° C ), Guinea pig complement, and liquid paraffin (light; available as nacalitesk) are necessary.
  • CO ink As an experimental device, CO ink
  • Protocol for establishing ES cells is described below. This protocol is based on the protocol described in the Human ES Cell Line Establishment Program compiled at the Institute of Regenerative Medicine at Kyoto University. In this specification, the protocol is not limited to this particular protocol, but may be any protocol. Can also be used.
  • Cryopreserved human fertilized eggs or early embryos up to the blastocyst stage are sequentially thawed and cultured. Since the substance that can identify the donor is removed from each frozen embryo container, the origin of the human fertilized embryo used in each thawing and culture experiment cannot be identified. However, in order to prevent the handling of each human fertilized embryo from being neglected, the human fertilized embryos stored in one cryocontainer from the time of their acceptance as frozen embryos were identified as individual entities during the establishment research. And record what progress has been made.
  • Embryos that have reached the blastocyst stage are separated from the inner cell mass by immunosurgery with anti-human serum, and then cultured on the feeder cell layer I do.
  • the feeder cell of the present invention is used as the feeder cell. The remainder after collecting the inner cell mass is also handled with courtesy.
  • the cells grown on a single feeder cell are dissociated in a timely manner, divided and subcultured, and cell cultures that are likely to be ES cells are established while performing selective culture of cell colonies that are thought to be stem cells. During this time, We will conduct research aimed at improving the culture maintenance method and cell cryopreservation method.
  • stem cell markers alkaline phosphatase activity and specific antigens
  • Detection of stem cell markers is performed to confirm that the cells are ES cells.
  • karyotype analysis is performed to test whether the number and morphology of chromosomes are normal.
  • induction of cell differentiation by changing culture conditions and creation of cell aggregates and analysis of the ability to differentiate into various functional cells are performed.
  • transplantation into an immunodeficient mouse or the like is performed to analyze the tissue division ability by teratoma formation.
  • Temporary storage of human frozen embryos will use a dedicated liquid nitrogen tank to prevent contamination by viruses and microorganisms derived from cells and animal embryos other than those used for the establishment plan.
  • a dedicated carbon dioxide incubator for cell culture experiments, mixing with other types of cells is prevented, and the possibility of contamination by viruses and microorganisms is reduced.
  • the culture medium and culture equipment used for cell culture are subjected to autoclaving in a laboratory and then discarded. Strictly control the entry to the laboratory where human fertilized embryos are stored and cell lines are established.
  • pluripotency or "probably dandelion” is used interchangeably and refers to a property of a cell, and is divided into one or more, preferably two or more various tissues or organs. The ability to dagger. Therefore, “pluripotency” and “pluripotency” are used interchangeably with “undifferentiated” in this specification unless otherwise specified.
  • cell pluripotency is limited as development progresses, and in adults, the constituent cells of one tissue or organ rarely change into cells of another. Thus pluripotency is usually lost.
  • epithelial cells are unlikely to change into other epithelial cells. When this happens, it is usually a pathological condition and is called metaplasia.
  • ES cells have pluripotency.
  • Tissue Stem cells are pluripotent.
  • the ability to divide into all kinds of cells constituting a living body such as a fertilized egg is called totipotency, and pluripotency is totipotency. It can encompass concepts. Whether a certain cell has pluripotency includes, but is not limited to, for example, formation of an embryoid body (Embryoid Body) in an in vitro culture system, culture under conditions for inducing induction, and the like.
  • Assays for the presence or absence of pluripotency using living organisms include the formation of teratomas by transplantation into immunodeficient mice, the formation of chimeric embryos by injection into blastocysts, and the transfer into living tissues. And proliferation by infusion into ascites, but are not limited thereto.
  • pluripotency the ability to divide into all kinds of cells constituting a living body such as a fertilized egg
  • pluripotency is a concept of totipotency. May be included.
  • totipotency and pluripotency can be distinguished, the former refers to the ability to divide into any cell, and the latter has multiple directions, Has the ability to divide in all possible directions.
  • the ability to divide in only one direction is also referred to as singularity.
  • totipotency and pluripotency can be determined, for example, by the number of days after fertilization.
  • they can be distinguished on the basis of about 8 days after fertilization.
  • mice typically follow the following course after fertilization.
  • 6.5 days after fertilization also referred to as E6.5
  • the primitive streak also referred to as the original streak
  • the original streak indicates the future posterior end of the embryo, extending across the ectoderm to the distal end of the cylinder.
  • the original streak is the area where cell motility takes place, resulting in the formation of future endoderm and mesoderm.
  • E7.5 a head process appears in front of the nodule, which forms the notochord, surrounding it, the future endoderm in the lower layer and the neural plate in the upper layer.
  • the nodules appear around E6.5 and move backwards, forming an axial structure from front to back.
  • E8.5 By day 5, the embryos have grown somewhat longer, and large anterior folds are formed at their anterior ends, most of which also have anterior neural plate strength. The somites begin to evolve backwards at a rate of one every 1.5 hours at E8. Cells beyond this time will no longer exhibit totipotency and will not form an individual, even if they are returned to the placenta, unless dissociated.
  • the term "fetus” or “fetus” is used interchangeably and refers to a period in which the offspring of a mammal complete differentiation of each organ primordium and enter the anagen phase until power delivery. A certain organism.
  • an "established” or “established” cell refers to a cell that maintains a specific property (eg, pluripotency) and proliferates stably under culture conditions. It is the state that has started to continue. Therefore, established stem cells maintain pluripotency.
  • Established differentiated cells have certain defined functions. Established differentiated cells are often cancerous, but are not limited thereto.
  • differentiation generally refers to the separation of one system into two or more qualitatively different systems. When used for, it means that the function and Z or form are special. With differentiation, pluripotency usually decreases or disappears.
  • the term "divided cells” refers to cells whose functions and morphology are specialized (for example, muscle cells, nerve cells, and the like). No or little potential.
  • the differentiated cells include, for example, epidermal cells, spleen parenchymal cells, splenic duct cells, hepatocytes, blood cells, cardiomyocytes, skeletal muscle cells, osteoblasts, skeletal myoblasts, nerve cells, vascular endothelial cells, and pigments Cells, smooth muscle cells, fat cells, bone cells, chondrocytes and the like.
  • the sorting cells may be in the form of a population or a tissue. Incorporation of shunt cells into a stem cell preparation is often detrimental in the use of stem cells. It has been difficult in the prior art to reduce such shunt cells.
  • somatic cells are cells other than germ cells such as eggs and sperm, and refer to all cells that do not directly transfer their DNA to the next generation. Somatic cells usually have a limited or pluripotent force or disappear. As used herein, somatic cells may be naturally occurring or genetically modified.
  • normal cells refers to any cells that are not in a pathological state. Therefore, such normal cells often have a finite number of divisions. Such finite split times The number is usually 50-80 times.
  • immortalization refers to the fact that cells do not die even after a finite number of divisions (for example, 50 to 80 times).
  • cell line refers to a cell that survives the above-mentioned finite number of divisions.
  • Such cell lines are obtained by sequentially subculturing passaged cells, such as primary cultured cells, and selecting cells that survive after passage beyond the number of passages at which normal cells usually die. Can be established. Such establishment methods are known in the art.
  • fibroblast refers to a cell that supplies a fiber component of a supporting tissue and forms an important component of a fibrous connective tissue.
  • Tissue section diagrams often have a flat, long outline and irregular projections.
  • the cytoplasm contains mitochondria, Golgi apparatus, centrosomes, small fat globules, etc., but does not show any special restriction.
  • the nucleus is elliptical and often lies close to collagen fibers.
  • feeder layer or “feeder cell” (feeder layer or feeder cell) is used interchangeably and is a cell provided on a culture substrate and which cannot be maintained alone by culture. Refers to a feeder layer with other cell types that allows the growth of the species and the expression of the Z or sliver trait. It is said that there are many types of tissue cells that cannot express the shunting phenotype or even proliferate under normal cell culture conditions. Such cells include, for example, stem cells (especially, ES cells, neural cells). Stem cells, mesenchymal stem cells, hematopoietic stem cells, etc.), embryonic germ cells (EG cells), germ stem cells (GS cells), and the like, but are not limited thereto.
  • stem cells especially, ES cells, neural cells.
  • Stem cells mesenchymal stem cells, hematopoietic stem cells, etc.
  • EG cells embryonic germ cells
  • GS cells germ stem cells
  • These cell types generally require specific auxotrophic growth and differentiation inducing factors.
  • a specific cell layer formed by supporting cells of the cell type or cells similar to the cell type in the living body as a culture substrate the factors required by the cell type to be cultured and Z Or, by being supplied with a nutrient source, they grow and divide.
  • the cell type to be used as a feeder cell is selected depending on the target cell type, but it is often used by suppressing cell proliferation by a method such as UV irradiation.
  • the ability to culture germ cells, ES cells, hematopoietic stem cells, etc. is largely dependent on the further utilization of feeders.
  • a feeder cell Mouse fibroblasts (primary cultured cells or cell lines) are used, and the efficiency of stem cell establishment is said to be about 10%.
  • a preparation containing such a feeder-cell is referred to as a “feeder-cell preparation”.
  • a single feeder cell mixture when a cell is a mixture of two or more cells and is used as a single feeder cell, it may be referred to as “a single feeder cell mixture” or “a single cell mixture”.
  • STO cell refers to an established mouse fibroblast cell line, and includes a cell deposited as CRL-1503 in the ATCC. STO cells are derived from the SIM fibroblast cell line, are resistant to 6-thioguanine and ⁇ abain, are HG PRT— (HPRT—), and are said to be sensitive to HAT. Conventionally, STO cells have been used as feeder cells, and their targets include the strength of ES cells, teratocarcinoma, and hybridomas.
  • any culture conditions as appropriate to maintain STO cells.
  • FCS 10% serum
  • the culture vessel may be coated with gelatin. It is recommended, but not limited to, passage at 1: 3—1: 10.
  • As the STO cells those deposited with the ATCC may be used, or irradiated STO cells (eg, deposited with ATCC as 56-X) may be used.
  • SL10 cell is a sub-established cell line of STO cell line, and its establishment method is described in Kawase E., et al., Int. J. Dev. Biol. 38: 385-390 (1994), preparation of feeder cells can be performed according to the method of Kawase et al. [Kawase et al., Experimental Medicine, Vol. 10. No. 13 (extra), 1575-1580, 1992]. . Robertson E. J. (ed) Teratocarsmomas and Emryonic Stem Cells, IRL Press, New York (1987), which is incorporated herein by reference. To do.
  • transfected Hue Tato the STO cells with the vector containing the neo 1 gene. Thereafter, the colonies that have become neomycin resistant are amplified, and their suitability as feeder cells is confirmed.
  • the ES cell line CCE can be used for the test, but is not limited thereto.
  • SL10 cells are selected for good maintenance of undifferentiated stem cell colonies of CCE. SL10 cells are said to be suitable as a feeder because of their good adhesion to gelatin.
  • primary cultured cell refers to a cell that has been cultured until the first passage, in which cells, tissues, organs, and the like separated from the body are implanted. It is understood in this document that passages performed up to about 5 times fall within the definition of primary cultured cells herein.
  • the primary culture cells it is usually preferable to use cells without any modification, but it is also possible to use cells inactivated with mitomycin C or X-rays.
  • any culture solution can be used as long as the cells are maintained or differentiated into desired differentiated cells.
  • Such cultures include adrenocortical steroids such as dexamethasone, insulin, glucose, indomethacin, isobutyl-methylxanthine (IBMX), ascorbate 2-phosphate, ascorbic acid and its derivatives, glycemic phosphate, Estrogen and its derivatives, progesterone and its derivatives, androgen and its derivatives, growth factors such as aFGF, bFGF, EGF, IGF, TGF jS, ECGF, BMP, PDGF, pituitary extract, pineal extract, retinoic acid, vitamin D , Thyroid hormone, calf serum, calf serum, horse serum, human serum, heparin, sodium bicarbonate, HEPES, albumin, transferrin, selenate (such as sodium selenite), linolenic acid, 3-isobutyl-1-methylxanthine Demethylating agents such as 5-azancytidine; Histone deacetylating agents such as
  • the term "diversion factor” refers to both a “differentiation promoting factor” and a factor known to promote diversion to differentiated cells (for example, a chemical substance, Temperature, etc.). Such factors include, for example, various environmental factors, such as temperature, humidity, pH, salt concentration, nutrition, metals, gases, organic solvents, pressure, and chemicals. (Eg, steroids, antibiotics, etc.) and any combination thereof, but are not limited thereto.
  • DNA demethylating agents such as 5-azacytidine
  • histone deacetylating agents such as trichostatin
  • nuclear receptor ligands such as retinoic acid (ATR A), vitamin D, T3, etc.
  • cell growth factors activin, IGF-1, FGF, PDGF, TG
  • cytosine arabinoside mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polypropylene, selenium, and the like.
  • diversion factor [0123] Specific examples of the diversion factor include the following.
  • VEGF vascular endothelial growth factor
  • EPO EGF
  • interleukins and interferons eg, interleukin 2, etc.
  • in vivo or “in vivo” (in vivo) refers to the inside of a living body.
  • in vivo refers to the location where the tissue or organ of interest is to be located.
  • in vitro refers to a state in which a part of a living body is extirpated or released (for example, in a test tube) for various research purposes. Say. A term that contrasts with in vivo.
  • ex vivo refers to the extraction of a target cell for gene transfer from a subject, the introduction of a therapeutic gene in vitro, and then the same subject again. When returning, a series of actions is called ex vivo.
  • self refers to an individual or a part thereof (eg, a cell, a tissue, an organ, etc.) derived from the individual.
  • self may broadly include a transplant from another genetically identical individual (eg, an identical twin).
  • the allogeneic refers to an individual who is the same species but is genetically different from another individual to be transplanted or a part thereof (eg, cells, tissues, organs, etc.). Say. Due to their genetic differences, allogenes can elicit an immune response in the transplanted individual (recipient). Examples of such cells include, but are not limited to, cells derived from the parent.
  • heterologous refers to a substance which is transplanted into a heterologous individual.
  • a transplant from a pig is referred to as a xenograft.
  • the term "recipient” refers to an individual that receives transplanted cells and the like, and is also referred to as a "host”. On the other hand, an individual that provides cells or the like to be transplanted is called a “donor” (donor). Recipient and donor may be the same or different
  • the cells used in the present invention may be derived from syngeneic (autologous (autologous)), allogeneic (from another individual (heterologous)), or heterologous. Autologous cells are preferred because rejection is likely, but if rejection is not an issue, it may be allogeneic.
  • transplantation means that the cells, compositions, medicaments, and the like of the present invention are transferred into the body alone or in combination with other therapeutic agents.
  • the present invention can use the following methods, forms and amounts of introduction into a treatment site (eg, bone, etc.): And then insert it.
  • Combinations of the adipose stem cells of the present invention and the sorting cells may be, for example, simultaneously as a mixture, separately but simultaneously or in parallel; or sequentially. May be administered. This includes presentation in which the combined drug power is administered together as a therapeutic mixture, and also includes procedures in which the combined drugs are administered separately but simultaneously (eg, a differentiation promoting factor).
  • “Combination" administration or transplantation further includes separately administering one of the cells, drugs, compounds or agents given first, followed by the second.
  • maintaining the cornea when used for a cell, tissue or organ, refers to substantially retaining its function and Z or morphology.
  • maintaining the cornea means having the function and Z or morphology of the cornea that the subject should normally have without substantially damaging the subject.
  • the function includes maintaining the visual acuity and the form includes maintaining the appearance. Listed, but not limited to.
  • regeneration refers to a phenomenon in which, when a part of an individual's tissue or organ is lost, the missing tissue is supplemented and restored. Depending on the species between animals or tissue in the same individual, the extent and mode of regeneration will vary. Many human tissues have a limited ability to regenerate, and complete loss cannot be expected if they are severely lost. In a major injury, incomplete regeneration can occur in which a highly proliferative tissue that is different from the lost tissue proliferates, incompletely regenerating the tissue and ending in a state in which function cannot be restored.
  • the proliferation power to the tissue defect is prevented, and the invasion of the tissue is prevented, thereby securing a space in which the original tissue can proliferate.
  • Regenerative medicine is being used to increase the ability of native tissues to regenerate by supplementing growth factors.
  • An example of this is regenerative medicine for cartilage, bone and peripheral nerves.
  • any tissue can be regenerated in principle, and the organs, tissues and cells of the present invention thus prepared can be regenerated.
  • Provided as an implant for Cells can be classified according to their origin into ectoderm, mesoderm and endoderm-derived stem cells.
  • the cells derived from the ectoderm mainly exist in the brain, and include neural stem cells and their cells.
  • Mesodermal-derived cells are mainly present in bone marrow, and include vascular stem cells and their differentiated cells, hematopoietic stem cells and their differentiated cells, mesenchymal stem cells and their cells, and the like.
  • Endoderm-derived cells are mainly present in organs, and include hepatic stem cells and their cells, knee stem cells and their cells. Honmei In the text, the cells can be from any germ layer.
  • stem cells can be genetically modified. Since stem cells are cultured cells, various gene transfer methods (eg, calcium phosphate method, ribosome method, microinjection method, electoral poration method, etc.) can be used just like other cultured cells.
  • the gene that can be introduced is not limited, and examples thereof include genes derived from bacterial, animal or human chromosomes, but are not limited thereto.
  • modification by homologous gene recombination of an endogenous gene using a targeting vector used as a method of gene targeting using an ES cell line is also possible.
  • the disease, disorder, or condition that can be targeted by the method of the present invention includes any disease, disorder, or condition in which regeneration of an organ or tissue is desired.
  • the present invention is particularly advantageous for diseases, disorders and conditions related to organs, fibrils or cells requiring stem cells which cannot be regenerated without feeder cells.
  • Such diseases and disorders that can be targeted by the present invention include: diseases or disorders of the circulatory system (such as blood cells); diseases or disorders of the nervous system; diseases or disorders of the immune system; Diseases or disorders of the skin; diseases or disorders of the skin; diseases or disorders of the endocrine system; diseases or disorders of the respiratory system; diseases or disorders of the digestive system; diseases or disorders of the urinary system; Obstacles, but are not limited to them.
  • the animal targeted by the present invention is any animal that has stem cells (for example, metal eel, scallop, cartilaginous fish, teleost, amphibian, reptile, bird, mammal, etc.). It may be.
  • stem cells for example, metal eel, scallop, cartilaginous fish, teleost, amphibian, reptile, bird, mammal, etc.
  • Such an animal is a mammal (e.g., monotremes, marsupials, oligodentates, dermis, winged fins, carnivores, carnivores, longnoses, snouts, even Hoofs, tube rodents, squamata, sponges, cetaceans, primates, rodents, egrets).
  • Exemplary subjects include, but are not limited to, animals such as, for example, horses, pigs, horses, chickens, cats, dogs, and the like. More preferably, primates (eg, chimpanzees, power-quisars, civil monkeys, humans) are used. Most preferably, it is intended for humans.
  • animals such as, for example, horses, pigs, horses, chickens, cats, dogs, and the like. More preferably, primates (eg, chimpanzees, power-quisars, civil monkeys, humans) are used. Most preferably, it is intended for humans.
  • such a medicament may further include a pharmaceutically acceptable carrier and the like.
  • a pharmaceutically acceptable carrier contained in the medicament of the present invention For example, any substance known in the art can be used.
  • Such suitable formulation materials or pharmaceutically acceptable carriers include antioxidants, preservatives, colorants, flavors, diluents, emulsifiers, suspending agents, solvents, fillers, bulking agents , Buffering agents, delivery vehicles, diluents, excipients and / or pharmaceutical adjuvants.
  • the medicament of the invention will be administered in the form of a composition comprising the cells of the invention together with one or more physiologically acceptable carriers, excipients or diluents.
  • suitable vehicles may be water for injection, physiological solutions, or artificial cerebrospinal fluid, which may be supplemented with other materials common in compositions for parenteral delivery. is there.
  • an acceptable carrier, excipient, or stabilizer is non-toxic to the recipient, and is preferably inert at the dosages and concentrations employed.
  • Eg phosphate, citrate or other organic acids; ascorbic acid, a-tocopherol; low molecular weight polypeptides; proteins (eg, serum albumin, gelatin or immunoglobulin); hydrophilic polymers (eg, polybutyl) Amino acids (eg, glycine, glutamine, asparagine, arginine or lysine); monosaccharides, disaccharides and other carbohydrates (including glucose, mannose, or dextrin); chelating agents (eg, EDTA); sugar alcohols (eg, EDTA); For example, mannitol or sorbitol); salt-forming counterion (Eg, sodium); and Z or non-ionic surfactants (eg, Tween, pluronic or polyethylene glycol (PEG)), and the like.
  • Eg phosphate,
  • Suitable carriers include neutral buffered saline or saline mixed with serum albumin.
  • the product is formulated as a lyophilizate using suitable excipients (eg, sucrose).
  • suitable excipients eg, sucrose
  • Other standard carriers, diluents and excipients may be included as desired.
  • the medicament 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 drug used in the treatment method of the present invention is determined in consideration of the purpose of use, the target disease (type, severity, etc.), the age, weight, sex, medical history, cell morphology or type of the patient, etc. Then, those skilled in the art can easily determine.
  • the frequency of applying the treatment method of the present invention to a subject (or patient) also depends on the purpose of use, the target disease (type, severity, etc.), the age, weight, sex, medical history, and course of treatment of the patient. A person skilled in the art can easily determine this in consideration of such factors.
  • the frequency includes, for example, administration once every few months (for example, once a week, once a month). It is preferable to administer once a week once a month while observing the progress.
  • the amount to be administered can be determined by estimating the amount required by the site to be treated.
  • the "instruction” refers to a method of administering or diagnosing the medicament or the like of the present invention for a physician, a patient or the like who administers or diagnoses (possibly a patient). It is described.
  • This instruction describes a word indicating a procedure for administering the diagnostic agent, the medicine and the like of the present invention.
  • This instruction is prepared in accordance with the format prescribed by the competent authority of the country where the present invention is implemented (for example, the Ministry of Health, Labor and Welfare in Japan and the Food and Drug Administration (FDA) in the United States) and is issued by the competent authority. The approval is clearly stated. Instructions are so-called package inserts, which are usually provided on paper media, but are not limited to, for example, electronic media (eg, homepages (websites) provided on the Internet, emails, etc.). ) Can also be provided.
  • Judgment of the termination of treatment by the method of the present invention may be based on the results of standard clinical tests using commercially available atseys or devices or the above-mentioned diseases (eg, neurological diseases, cardiac diseases, etc.). Can be supported by the disappearance of various clinical symptoms. Treatment can be resumed upon recurrence of the disease (eg, neurological disease, heart disease, etc.).
  • diseases eg, neurological diseases, cardiac diseases, etc.
  • the present invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more components (eg, stem cells such as ES cells) of the medicament of the present invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more components (eg, stem cells such as ES cells) of the medicament of the present invention.
  • the stem cells of the present invention are desirably distributed according to ethical rules set by the authorities.
  • Such rules include, but are not limited to, those stipulated by the Japanese government. The following is an example of the rules.
  • the distributor is responsible for the distribution. Distributors should have clean rooms and equipment necessary for cell culture and a liquid nitrogen tank for cell cryopreservation. It is used by the human ES cell line establishment team, and its staffing can be, for example, five researchers (two of whom are preferably doctor's license holders). At the start of cell distribution, about two additional assistants for experimentation and one assistant clerk for office work may be assigned to work. It is preferable to increase the number of personnel and facilities in response to the increasing demand for cell distribution and training.
  • Examples of the distribution method and conditions include the following: The priority should be given to securing a cell stock at an institution required for maintaining and growing the ES cell line.
  • the fertility treatment physician is responsible for neutralizing frozen embryos that have been decided to be discarded. Ask the candidate to receive an explanation of ES cell research while keeping the field.
  • the providing candidate responds to the head of the providing medical institution whether or not he / she agrees with the providing. Consent requires the signing of informed 'consent' documents, which will be kept securely by the provider. The results of consent or disagreement by a particular candidate will not be communicated to the relevant parties.
  • kits refers to a unit to which a part to be provided (eg, a reagent, a particle, or the like) is provided, usually divided into two or more sections.
  • This kit form is preferred when it is intended to provide a composition such that it should preferably be mixed and used shortly before use, which should be provided as a mixture.
  • kits will preferably include instructions describing how to process the provided components (eg, reagents, particles, etc.).
  • Such instructions may be in any medium. Examples of such medium include, but are not limited to, a paper medium, a transmission medium, and a recording medium. Transmission media include, for example, the Internet, intranets, , LAN, etc., but are not limited to them. Examples of the recording medium include, but are not limited to, CD-ROM, CD-R, flexible disk, DVD-ROM, MD, mini disk, MO, and memory stick.
  • the stem cells of the present invention can be used to generate transgenic mice.
  • General techniques for producing transgenic mice are described in International Publication WO 01Z13150 (Ludwig Inst. Cancer Res.).
  • U.S. Pat.No. 4,873,191 (Wagner et al.) Teaches mammals having exogenous DNA obtained by microinjection of DNA into mammalian zygotes; The description is incorporated herein. .
  • the force or the loss of function by replacing or destroying the endogenous target gene The method of producing a transgenic (target transgenic) mouse having an altered mutation is useful for analyzing the function of the gene because the mutation is introduced only into the targeted gene.
  • the obtained recombinant ES cells are mixed with normal embryos by a scutellum injection method or an assembly chimera method to produce a chimeric mouse of ES cells and a host embryo.
  • a scutellum injection method or an assembly chimera method to produce a chimeric mouse of ES cells and a host embryo.
  • blastocyst injection ES cells are injected into blastocysts with a glass pipette.
  • the assembly chimera method a mass of ES cells is adhered to an 8-cell stage embryo from which the zona pellucida has been removed.
  • the blastocyst into which the ES cells have been introduced is transplanted into the uterus of a surrogate mother who has been pseudopregnant to obtain a chimeric mouse.
  • ES cells Since ES cells have totipotency, they can be divided into all kinds of cells including germ cells in vivo.
  • a chimeric mouse having germ cells derived from ES cells is crossed with a normal mouse, a mouse having a heterologous ES cell chromosome is obtained. A mouse is obtained.
  • transgenic mice homozygous for the modified chromosome from the resulting chimeric mice male chimeric mice and female wild-type mice are bred to produce F1 generation heterozygous mice. And breeding female heterozygous mice to select F2 generation homozygous mice.
  • Whether or not the desired gene mutation has been introduced into each generation of F1 and F2 can be determined by methods commonly used in the art, such as Southern blotting, PCR, and decoding of the base sequence, as in the case of recombinant ES cells. Can be analyzed using
  • Cre- ⁇ As a next-generation technology that overcomes the inability to selectively analyze various gene functions, a technology that combines cell type-specific expression of Cre recombinase and site-specific recombination of Cre- Cre ⁇ has attracted attention. Being done.
  • a transgenic mouse using Cre- ⁇ inserts a neomycin resistance gene at a position that does not inhibit the expression of the target gene, and inserts a targeting vector that inserts the ⁇ ⁇ ⁇ sequence with an exon to be deleted later. The cells are introduced, and then the homologous recombinant is isolated. A chimeric mouse is obtained from the isolated clone, and a genetically modified mouse is produced.
  • Cre a site-specific recombination enzyme derived from the P1 phage of Escherichia coli, the gene is disrupted only in the tissue that expresses Cre.
  • the ⁇ sequence 34bp
  • Cre can be expressed in adults using the ability to breed with a transgenic mouse having a Cre gene linked to an organ-specific promoter, or a virus vector having a Cre gene.
  • a gene trap (gene trap) method As a method for analyzing a specific gene, a gene trap (gene trap) method has attracted attention.
  • a reporter gene without a promoter is introduced into a cell, and when that gene is accidentally inserted into the genome, the novel gene is isolated by utilizing the expression of the reporter gene ( Trap).
  • the gene trap method is a method for efficient insertion mutation and identification of unknown genes based on mouse early embryo manipulation, ES cell culture, and gene targeting by homologous recombination (Stanford WL., Et al.). al., Nature Genetics 2: 756—768 (2001)).
  • introduction of a gene, selection of an inserted mutant, and phenotyping thereof are relatively easy.
  • a gene trap vector in which ⁇ geo, a fusion gene of lacZ and neo, is linked between a splicing Z receptor sequence and a poly-A-added signal is introduced into ES cells.
  • G4108 When selecting with G418, only clones that accidentally trap the gene expressed in ES cells are selected.
  • the present invention can be used for production of various transgenic organisms.
  • the present invention provides a feeder-one cell preparation for preparing a stem cell, including a normal cell and a cell line.
  • a mixture of feeder cells having such a plurality of properties has been used as a feeder cell preparation.
  • Furthermore conventionally, several cell types that can be used as feeder cells have been known. No substantial difference was expected between the two, and it was anticipated that combining cells would significantly improve feeder-cell function.
  • the mechanism by which the combination of cells as in the present invention greatly improves the efficiency of stem cell line establishment and maintenance! /, And the functions of feeder cells are elucidated at the molecular level!
  • the invention can employ a mixture of any plurality of feeder-cells.
  • the feeder cell used in the present specification can be any cell that is conventionally used as a feeder cell.
  • normal cell used in the present specification can be used as long as the cell is appropriately used as a feeder cell.
  • normal cells include, but are not limited to, mouse primary cultured fibroblasts, rat primary cultured fibroblasts, monkey primary cultured fibroblasts, and human primary cultured fibroblasts. No.
  • a method for preparing such primary cultured fibroblasts for example, a fetus from which a head, internal organs, and the like are removed from an 11-16-day-old fetus is shredded, and the fibroblast is mainly extracted by enzymatic treatment. Then, it can be performed by using a method.
  • t, Fibroblasts can be prepared according to the method.
  • the normal cells are not immortalized. While not wishing to be bound by theory, it is believed that non-immortalization provides some of the factors necessary to maintain stem cells without differentiation.
  • the normal cells used in the present invention may be fibroblasts, but need not be.
  • fibroblasts are used. While not wishing to be bound by theory, it is important to note that fibroblasts, when given some of the factors necessary to maintain stem cells without differentiation, often do not confer suitability as feeder cells It is a conceivable force.
  • the normal cells used in the present invention are fibroblast primary culture cells. More preferably, the normal cells used in the present invention are mouse-derived. While not wishing to be bound by theory, a mouse is preferred because of its ease of preparation. Therefore, any mammal can be used, and if another simple preparation method exists, it can be preferably used by using cells of another mammal.
  • the normal cells used in the present invention may be derived from a fetus (fetus). While not wishing to be bound by theory, it is believed that cells from the fetus provide some of the factors necessary to maintain stem cells without differentiation and are more suitable as feeder cells. is there.
  • the cell line used in the present invention is considered to have the most different properties from the viewpoint of cytology, and therefore, cannot be provided by a cell line. This is because it is considered to have the ability to supply factors necessary for the maintenance of cells.
  • any passage number of cells can be used as long as the ability as a feeder cell can be provided, but it is preferable that primary culture cells have a low passage number. Cells (about 4 or 5 times) are recommended, and cells with 5 or less passages, 4 or less, 3 or less, 2 or less, 1 passage, or no passage are used.
  • the normal cells used in the present invention are fibroblasts obtained by enzymatic treatment from a mouse 11-16 day old fetus.
  • any cell line can be used as long as it has the ability as a feeder cell as used herein.
  • Such cell lines are typically immortalized.
  • immortalized cells does not provide for normal cells such as primary cell lines, but does not provide other factors necessary to maintain stem cells without fragmentation. It is considered that some of this will be supplied.
  • the cell line used in the present invention is a fibroblast cell line.
  • the cell line used in the present invention may have any passage period.
  • the passage period is 2 months or less, and more preferably, the passage period is not more than 2 months. It may be advantageous to use cells that have been for a period of less than one month. Without wishing to be bound by theory, it is possible to stably supply cells secreting the initial factor of interest by using such a cell line with a short passage time, and to obtain stem cells without differentiation. It is not necessary that the passage period be short, because some of the other factors necessary to maintain the force will be supplied adequately.
  • the cell line used in the present invention may be derived from a mouse, but is not limited thereto. While not wishing to be bound by theory, a mouse is preferred because of its ease of preparation. Therefore, any mammal can be used, and if another simple preparation method exists, it can be preferably used by using cells of another mammal. While not wishing to be bound by theory, it has been empirically shown that the use of allogeneic fibroblast cell lines in ES-establishment studies in Escherichia coli and egrets does not appear to be as effective as in mice. Generally, mouse cells are considered to be good.
  • the cell line used in the present invention may be derived from a fetus (fetus). While not wishing to be bound by theory, it is believed that fetal-derived cells provide some of the factors needed to maintain stem cells without differentiation and are more suitable as feeder cells It is.
  • the normal cells used in the present invention are considered to have the most different properties from a cytological point of view, and therefore have the ability to supply factors necessary for maintaining stem cells that normal cells cannot provide. Because it is possible.
  • the cell line used is an STO line.
  • this cell line has the properties of neomycin resistance (neo resistance).
  • the cell line used in the present invention is the SL10 strain.
  • Normal cells for example, primary fibroblast cells
  • cell lines for example, fibroblast cell lines contained in the feeder-cell preparation of the present invention are typically about 1: 10- Can exist at 10: 1.
  • 10: 1 a cell line contained in the feeder-cell preparation of the present invention.
  • normal cells eg., primary fibroblast cells
  • cell lines e.g., cell lines
  • a fibroblast cell line can be about 1: 4 to 4: 1. While not wishing to be bound by theory, the use of a feeder-one cell preparation containing multiple types of cells significantly improves the effects of maintaining stem cells and possibly maintaining dangling ability. It is.
  • a more preferred cell ratio may be a ratio of about 1: 3 to about 3: 1 normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts).
  • the ratio of normal cells (eg, primary fibroblasts) to cell lines (eg, fibroblasts) is approximately equal. Without wishing to be bound by theory, it is because using approximately equal amounts of multiple types of cells results in sufficient components of each other to maintain stem cells to complement each other. Therefore, those skilled in the art understand that, when referring to substantially equivalent amounts in this specification, means any ratio that is close to equivalent amounts but is optimal as a feeder cell.
  • the cell of interest in the feeder-one-cell preparation of the present invention is an embryonic stem cell, more preferably a human embryonic stem cell.
  • Cells like human embryonic stem cells It can be grown while maintaining pluripotency and has less than 50% differentiated cell contamination (preferably less than 30%, more preferably less than 10%, and most preferably less than 1%). It can be said that this is a powerful effect that cannot be achieved with the technology described above.
  • the present invention provides a process for culturing a stem cell on a feeder-one cell preparation containing a normal cell (eg, a primary fibroblast cell) and a cell line (eg, a fibroblast cell line).
  • a normal cell eg, a primary fibroblast cell
  • a cell line eg, a fibroblast cell line
  • a method for preparing a stem cell comprising: Normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts) are each normal cells (eg, primary cells) and, as long as they are established, Anything can be used.
  • the feeder cell any cell may be used as long as it contains these two types of cells, but it is advantageous that the cells are uniformly mixed.
  • Such mixing ratios are preferably about 10: 1 to 1:10, about 9: 1 to 1: 9, about 8: 1 to 1: 8, about 7: 1 to 1: 7, about 6: 1: 1: 6, approx. 5: 1—1: 5, approx. 4: 1: 1-1: 4, approx. 3: 1-1—1: 3, approx. 2: 1-1—1: 2, approx. 1: 1 (almost equivalent And the like).
  • approximately equal amounts are mixed.
  • normal cells eg, primary fibroblasts
  • cell lines eg, fibroblasts
  • stem cell maintenance are factors that have mutually synergistic effects (eg, stem cell maintenance). It is considered that the unexpected effect of the present invention has been achieved by complementarily secreting (required).
  • the normal cells (eg, primary fibroblasts) used in the feeder cell of the present invention are advantageously derived from a mouse, but are not limited thereto. Therefore, any animal can be used in the present invention. Mice are technically well established experimental animals, and methods for obtaining normal cells such as primary cultured cells are also well known to those skilled in the art.
  • the normal cells (eg, primary cultured fibroblast cells) used in the present invention are advantageously derived from a fetus, but are not limited thereto.
  • primary fibroblast cells have been used as feeder cells, but those derived from the fetus are mainly used.
  • the efficiency of stem cell establishment is not so high. About 10-30%.
  • the feeder-cell preparations and cell mixtures of the present invention the establishment efficiency is well over 50%, and in some cases as high as 80%. Therefore, it can be said that such a high rate of establishment was beyond imagination from the prior art.
  • a normal cell is preferably a primary cultured cell in a strict sense, but even if it is a subcultured cell, if it has a small number of passages, it has properties similar to those of the primary cultured cell. Since it can have, it can be used advantageously in the present invention.
  • a passage number include, but are not limited to, about 5 passages or less, 4 passages or less, 3 passages or less, 2 passages or less, 1 passage, and the like.
  • the cell line (eg, fibroblast cell line) contained in the feeder cell of the present invention is advantageously an STO line.
  • the STO strain has been used as a feeder cell for stem cells, but the establishment efficiency of stem cells is still as low as 30% or less, and the cell mixture and the cell preparation of the present invention can be prepared using a feeder cell.
  • the efficiency of stem cell establishment is well over 50% and can be as high as 80%. Therefore, it can be said that such a high rate of establishment was beyond imagination from the prior art.
  • a more preferable cell line can be established by using a method for separating cells having a high ability to support an ES cell in an undivided state from STO cells. The following is a brief description of such a method.
  • One feeder cell is used using the cells of each subclone, and ES cells are cultured using the cells to select a subline having a high ability to support an undivided state.
  • the STO strain used in the present invention is advantageously neo-resistant.
  • such neo-resistant Choosing a cell strain also has the power to make it easier to select one with the highest feeder effect.
  • neo 1 gene any known gene transfer techniques in the art (e.g., transformation, full Ekushi Yung, etc.) you to achieved by introducing with the leave in.
  • the cell line used in the present invention is advantageously the SL10 strain.
  • the SL10 strain is a cell strain isolated based on the above method. It is understood that the SL10 strain is available in Meiji Dairy Power, distributed at Kyoto University's Institute of Regenerative Medicine, and readily available to third parties.
  • the stem cells supported by the present invention include any stem cells, and include, but are not limited to, ES cells, tissue stem cells, and the like.
  • the stem cells supported by the present invention are ES cells.
  • ES cells are a feeder cell with high establishment efficiency The present invention is noted for achieving an establishment efficiency of 80%, far exceeding the previously achievable upper limit of 30% for ES cells Should.
  • the stem cells supported by the present invention are tissue stem cells.
  • tissue stem cells include, but are not limited to, hematopoietic stem cells, neural stem cells, mesenchymal stem cells, and the like.
  • Conventional techniques, which also require a single feeder cell for tissue stem cells do not have a technology that achieves so much satisfaction and a particularly high establishment efficiency. Therefore, it is understood that the present invention is also applicable to tissue stem cells.
  • the stem cells supported by the present invention are primate cells, more preferably human stem cells.
  • Primate cells, including humans, have only recently been developed as ES cells, and research on feeder cells is still under development. In particular, it is not an exaggeration to say that feeder cells that provide satisfactory establishment efficiency do not exist at all with respect to primate stem cells, more preferably human stem cells.
  • the height is particularly high for primate stem cells, more preferably human stem cells, and the significance is high.
  • the culture dish in which the feeder cells of the present invention are arranged is coated with gelatin.
  • the coating is not limited thereto, and any coating may be applied as long as it enhances the culture efficiency.
  • Such coatings include, but are not limited to, for example, any extracellular matrix (eg, fibronectin, collagen, vitronectin, etc.).
  • the culture dish may be made of any material.For example, it is desirable that the material of the container is biocompatible, but unless a material that gives toxicity to the living body is used. It is understood that anything can be used.
  • Examples of the material of such a container include glass, polyethylene, ethylene, polypropylene, polyisobutylene, polyethylene terephthalate, unsaturated polyester, fluorine-containing resin, polyvinyl chloride, polyvinyl chloride, polyvinyl acetate, and polyvinyl acetate.
  • the feeder single cell of the present invention about 1 X 10 4 cells Z cm 2 - seeded at approximately 1 X 10 5 cells / cm 2, preferably, 2-5 X 10 5 It is seeded in a cell / cm 2.
  • the feeder single cell of the present invention about 1 X 10 4 cells Z cm 2 - seeded at approximately 1 X 10 5 cells / cm 2, preferably, 2-5 X 10 5 It is seeded in a cell / cm 2.
  • the feeder-cell of the present invention is usually used within 5 days after being mixed and inoculated, preferably within 3 days, more preferably within 2 days, and still more preferably. Used within one day.
  • a certain period of time eg, 1 hour is a force that establishes sufficient conditions for the components necessary for stem cell maintenance to emerge.
  • the stem cells used in the present invention are advantageously cultured in a medium containing knockout serum replacement supplement (KSR).
  • KSR is also available in power, such as Invitrogen.
  • the ability to generally show a tendency for cell growth to be slow can be used, as long as the object of the present invention is satisfied.
  • the stem cells used in the present invention are established in a liquid medium containing fetal bovine serum. It is understood that fetal serum can be used without any problem as long as ES cells can be stably maintained for a long period of time.
  • the method of the present invention further includes a sub-passing step, in which collagenase is used. It is thought that the use of collagenase facilitates cell recovery and contributes to improving the establishment efficiency.
  • the present invention provides a stem cell preparation prepared by the method for preparing a stem cell of the present invention.
  • stem cells have an advantageous effect different from stem cells prepared by the conventional method in that they are considered to remain in a more undivided state than stem cells obtained by the conventional method. It is thought that.
  • the cells are preferably ES cells, more preferably primate ES cells, and even more preferably human ES cells.
  • ES cells particularly primates such as humans
  • the stem cell preparation of the present invention can: 1) maintain a more undivided state than stem cells obtained by conventional methods; 2) reduce stem cells that are more normal and support tissue. 3) It is found that it has advantages such as being closer to a normal tissue, having properties, and being more normal.
  • the method of the present invention allows the culture to be performed more easily and stably while maintaining the undivided state. It has been shown that compared to the contamination with cells, the method of the present invention shows almost no shunt cells.
  • the present invention provides a transplant for regenerating an organ, tissue or cell.
  • a method for preparing is provided. The method comprises the steps of: A) providing a stem cell capable of differentiating into a desired organ, tissue or cell; B) converting the stem cell into a normal cell (eg, a primary fibroblast cell) and a cell line (eg, a fiber Culturing on a feeder-cell preparation containing the blast cell line); and C) separating the stem cells into a transplant for regenerating a desired organ, tissue or cell.
  • a normal cell eg, a primary fibroblast cell
  • a cell line eg, a fiber Culturing on a feeder-cell preparation containing the blast cell line
  • C separating the stem cells into a transplant for regenerating a desired organ, tissue or cell.
  • the feeder-cell any of the forms described in detail in the “Stem cell preparation method” and “Feeder-cell preparation” section can be used.
  • Any stem cells can be used as long as they can be separated into desired organs, tissues or cells. Whether it can be differentiated into a desired organ, tissue or cell can be confirmed using a technique known in the art. As such a technique, after exposing the stem cells to shading conditions and then culturing for a certain period of time, it is checked whether or not the shading is performed as desired, for example, with the naked eye or by observing with a microscope. Alternatively, examples include, but are not limited to, observations by molecular biology and identification of cell surface markers.
  • the dangling factor can be removed for culturing in the method of the present invention.
  • differentiation factors include, for example, DNA demethylating agents (eg, 5-azacytidine), histone deacetylating agents (eg, trichostatin), nuclear receptor ligands (eg, retinoic acid (ATRA), vitamin D
  • T3 cell growth factors (activin, IGF-1, FGF, PDGF, TGF-j8, BMP2 / 4, etc.), cytodynamics (LIF, IL-2, IL-6, etc.), hexamethylene bisacetate Amide, dimethyl acetoamide, dibutyl cAMP, dimethyl sulfoxide, ododeoxyperidine, hydroxyl urea, cytosine arabinoside, mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polyprene, selenium And the like, but are not limited thereto.
  • NGF and FGF can be added.
  • interleukin and EPO can be added.
  • Desired organs, tissues or cells for the purpose of preparing the regenerative transplant of the present invention include nerves, blood cells, bones, cartilage, heart, pericardium, blood vessels, muscles, eyes, liver, spleen, intestine, Stomach, lungs, trachea, Hair, skin, and the like, but are not limited thereto.
  • the desired organ, fibrous tissue or cell and the feeder cell are of the same species. By being the same species, it is also possible to minimize the immune response, which is preferable in terms of compatibility.
  • the above-mentioned desired organ, tissue or cell is a primate, and the feeder cell is preferably derived from a mouse.
  • This combination is a powerful factor in achieving a high rate of stem cell establishment efficiency of 80% or more.
  • combinations of species other than this combination may also be preferred in the present invention. Therefore, it is understood that feeder cells derived from primates such as humans may be used.
  • the culture of the present invention is performed ex vivo (ie, for the purpose of returning to self).
  • the stem cells of the present invention may be those that have just been removed from a subject or those that have been cryopreserved. It is preferable to use it as soon as it is removed. This is because the establishment efficiency is high.
  • the cultivation in the method of the present invention may be performed at 37 ° C under a saturated humidity of 5% CO under a 5% CO concentration.
  • Tilating agents eg, 5-azacytidine
  • histone deacetylating agents eg, trichostatin
  • nuclear receptor ligands eg, retinoic acid (ATRA)
  • cell growth factors activin, IGF-1, FGF, PDGF, TGF-j8, BMP2Z4, etc.
  • cytodynamics LIF, IL 2, IL-6, etc.
  • hexamethylene bisacetamide Dimethylacetamide, dibutyl cAMP, dimethyl sulfoxide, eododexperidine, hydroxyl urea, cytosine arabinoside, mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polyprene, selenium, etc.
  • one or more are included.
  • the present invention provides an organ, tissue or cell prepared by the method for preparing a regenerative transplant of the present invention.
  • an organ, tissue or cell is similar to a normal tissue and has properties in that it can be a regenerated organ or tissue prepared by a conventional method. It is thought to have a different beneficial effect than tissue or cells.
  • the present invention provides a system for regenerating an organ, tissue or cell.
  • the system comprises: A) a container; and B) feeder cells seeded on the container, comprising normal cells (eg, primary fibroblasts) and a cell line (eg, a fibroblast cell line).
  • normal cells eg, primary fibroblasts
  • a cell line eg, a fibroblast cell line.
  • the feeder-one-cell preparation can take any form described in the above “feeder-one-cell preparation” and “stem cell preparation method”.
  • the container can also utilize any material, for example, take any of the forms described in the section "Preparing a Regenerated Implant".
  • the present invention provides a mixture comprising normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts).
  • This mixture can be used as a stem cell feeder cell, as demonstrated in the present invention.
  • Normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts) can be any form as described in “Stem cell preparation method” and “Feeder-one cell preparation”. It is understood that it can be taken. Thus, it can be appreciated that substantially the cell mixture may be the same as the feeder-cell preparation.
  • the primary cultured cells in the mixture of the present invention are advantageously mouse fetal primary cultured fibroblasts.
  • This form is advantageous because stem cells can be established with unprecedented efficiency by supplying factors such as STO cells and other factors that cannot be obtained, and stem cells supporting more normal tissues can be established. This is because it is possible to do so.
  • the fibroblast cell line in the mixture of the present invention is advantageously an STO line.
  • the advantage of this form is that the factor supplied from mouse fetal fibroblasts can be obtained, so that stem cells can be established with unprecedented efficiency, and that stem cells that support more normal tissues can be established. It is a force that can be considered as an effect that can be done.
  • the present invention provides a method for regenerating an organ, tissue or cell.
  • This method comprises the steps of: A) providing a stem cell capable of dividing into a desired organ, tissue or cell; B) converting the stem cell into a normal cell (eg, a primary fibroblast cell) and a cell line (eg, , A fibroblast cell line) and C) transplanting the cultured stem cells to a site to be treated in a subject.
  • the feeder cell used herein can take any of the forms described in the above “feeder cell preparation” and “stem cell preparation method”.
  • the stem cells can also take any of the forms described in the “Feeder Cell Preparations” and “Stem Cell Preparation Methods” above.
  • Transplantation techniques can also be administered using any technique known in the art. Such techniques include, but are not limited to, stem cell transplantation into the myocardium using a catheter, for example.
  • the method of the present invention further comprises D) a step of differentiating the stem cell.
  • a method known in the art can be applied. It is understood that such conditions can vary depending on the target organ, tissue or cell. It is understood that those skilled in the art can, in consideration of such variations, appropriately determine and select appropriate fragmentation conditions.
  • the invention provides the use of a cell mixture comprising normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts) as a feeder-cell preparation.
  • a cell mixture comprising primary fibroblast cells and a fibroblast cell line as feeder cells.
  • Use of the preparation is provided.
  • feeder cell used herein can take any of the forms described in the above “stem cell preparation method” and “feeder cell preparation”.
  • Fee Dar cells are intended to support any cells, preferably stem cells.
  • stem cells can also take any of the forms described in the above “feeder-one cell preparation” and “stem cell preparation method”.
  • the present invention relates to a normal cell (for example, a primary fibroblast cell) and a cell line (for example, a fibroblast cell line) for producing a medicament containing a feeder cell.
  • a cell mixture comprising:
  • the feeder cells used herein can take any of the forms described in the “Stem cell preparation method” and “Feeder cell preparation” above.
  • the feeder cell can support any cells, but preferably aims to support stem cells.
  • the stem cells can also take any of the forms described in the above “Stem cell preparation method” and “Feeder-one cell preparation”.
  • a technique for producing a medicament a technique known in the art can be used.
  • a pharmaceutically acceptable carrier can be appropriately added to the medicament.
  • the feeder-cell preparation is treated so as not to proliferate (for example, with mitomycin C). This allows the feeder-cell preparation to be sold in a package.
  • Example 1 Preparation of primary cultured cells (MEF) of mouse fetal fibroblasts
  • Primary embryonic fibroblast cells also referred to as MEFs
  • mouse fetal power was prepared for mouse fetal power. Specifically, it is as follows.
  • a part of the fetus was removed from a pregnant mouse (ICR, CLEA Japan), the cells were separated, and seeded in a medium called DMEM + 10% fetal serum (FBS). The removed cells were maintained in DMEM + 10% FBS.
  • the MEF cells prepared in Example 1 were pretreated. The following were used for the pretreatment.
  • Mitomycin C was added to the confluent cells at a concentration of 10 ⁇ g / ml in the medium, and the cells were cultured for 1.5 to 12 hours.
  • the MEF cells thus prepared were used alone or in a mixture with other cells as feeder cells in the following experiments.
  • SL10 cells As STO cells, subclone SL10 cells of STO cells were used. SL10 cells were obtained from Meiji Dairies (Tokyo, Japan). In the case of SL10, 2 weeks is one feeder cell It is the limit of use, and it was subcultured in 7-10 days. DMEM + 10% FCS was used as the medium.
  • the STO cells were pretreated for use as feeder cells.
  • the procedure is as follows. The following were used for the pretreatment.
  • Mitomycin C was added to the confluent cells to a medium at 10 gZml, and the cells were cultured for 1.5 to 2 hours.
  • the cells were suspended at a concentration of 1.5 ⁇ 10 5 / ml.
  • STO cell strength also subclones suitable clones.
  • the procedure is as follows.
  • a subline of STO cells with similar properties can be isolated as follows.
  • One feeder cell is used by using the cells of each subclone, and ES cells are cultured using the cells to select a subline having a high ability to support an undivided state.
  • the cell line subcloned in this manner can also be used as a feeder cell.
  • one feeder cell was prepared.
  • a single feeder cell of the present invention a case where a fibroblast primary culture cell and a fibroblast cell line were used alone and a case where they were mixed were prepared. Preparation of feeder cells followed a similar protocol as shown below.
  • a culture dish for preparing a feeder was gelatin-coated in advance.
  • the gelatin coating was performed by covering the surface of the culture dish with a gelatin solution and incubating at 37 ° C for 1 hour or more.
  • Pretreated STO cells and pretreated MEF cells were used alone or as a 1: 1 mixture of suspensions.
  • the prepared feeder cells were used within 4 days.
  • stem cells were established using the feeder cells prepared in Example 5.
  • the culture solutions used are as follows.
  • Non-essential amino acids (Gibc0) 0.8 m
  • Human LIF (10, ug / 1) 00 ⁇ I (1 ng / ml) Basic fibroblast growth factor (1 mg / m1) ⁇ .4 / i1 (4 ng / ml).
  • the culture dish for making the feeder should be gelatin-coated in advance.
  • 0.1 Cover with 1% gelatin solution swine skin, Type A: Sigma) and incubate at 37 ° C for 1 hour or more. Remove the gelatin solution from the culture dish and add the cell suspension. After several hours, it can be used as a feeder cell.
  • ES cells were established using a single feeder cell as follows.
  • the zona pellucida was removed by enzyme treatment or the like, and the cells were incubated in an antibody solution that reacts with the mouse surface antigen. The outermost cells, vegetative ectoderm, were then removed by incubation in complement solution, and the inner cell mass was separated.
  • ES cell-like cells came off the culture dish.
  • the cells were picked up with a thinly drawn glass tube, raised and removed several times to divide the cell mass into several pieces. .
  • Example 6 the undifferentiated state of the cells established in Example 6 was examined by staining with a specific marker.
  • Embryonic stem cells were fixed with 4% PFA and immunostained in standard cultured cells (Willingham, MC et.al., 1985.An Atlas of Immunofluorescence in Cultured Cell, Academic Press, Orlando, FL, pp. 1-13. Staining was performed. Blocking was performed using 0.1% Triton X / PBS / 2% skim milk for 1 hour at room temperature, and washing was performed 4 times using 0.1% Triton XZPBS for 5 minutes at room temperature.
  • the primary antibody was a 200 ⁇ g / ml (CL ONTECH) 1Z100 diluted monoclonal antibody of SSEA-4 and TRA-1-60, respectively, and the secondary antibody was FITC-labeled goat anti-mouse IgG (H + L ) (ZYMED LABORATORIES, INC) diluted 1Z200 was used. After the reaction with the secondary antibody, staining was performed in the order of rhodamine phalloidin (Molecular Probe) and DAPI (SIGMA) to detect a signal.
  • Molecular Probe rhodamine phalloidin
  • SIGMA DAPI
  • ALP alkaline phosphatase histologic staining was performed as follows. The culture tissue was washed twice with Dulbecco's PBS (-), fixed with cold 95% ethanol at 4 ° C for 30 minutes or more, and then dehydrated with absolute ethanol at 4 ° C for 30 minutes or more. After removing the fixative, the dishes were washed three times for 5 minutes at room temperature with 0.1 M Tris-HCl (pH 9.0-9.5), and then 1.5--2 ml of the staining solution (Naphthol AS-BI phosphate ( Sigma, Catalog No. N-2125) 2.
  • FIG. Fig. 3 shows ALP, SSEA-4, and TRA-1-60 from left power.
  • ES cells prepared in the above Examples of the present invention were washed three times with a serum-free medium to completely remove serum. Undifferentiated cells were cultured on PA6 feeder cells for 8 to 11 days using ES cell culture medium containing KSR (Knockout Serum Replacement; GIBCO / Invitrogen Cat. No. 10828-028) (Kawasaki et al., 2000, Neur on 28; 31-40; see Tada et al., 2003; Dev. Dyn., 227; 504-510.
  • KSR Knockout Serum Replacement
  • SCID mouse (Clear Japan, Approximately 10 7 ES cells of the present invention were transplanted subcutaneously and intraperitoneally in (Tokyo) and observed for 13 months.
  • ES cells were differentiated into mesodermal cells. Undifferentiated ES cells are suspended in the above-mentioned differentiation-inducing culture medium, and seeded on a type IV collagen-coated 6 plate at a density of 1 ⁇ 10 4 Z-well. Incubate this at 37 ° C, 5% CO for 4 days.
  • the cells were suspended in 10 ml of HBSSZBSA, and centrifuged at 1,200 rpm for 5 minutes to remove the supernatant.
  • the cells were suspended in 0.1 ml of normal mouse serum per 1 ⁇ 10 7 cells and allowed to stand for 10 minutes. Appropriate amounts of E-force doherin antibody and anti-FLK1 antibody were placed on ice water for 20 minutes. After washing the cells twice with HBSSZBSA, the FLK1 + E-forcedherin monomesoderm cells are sorted by FACS.
  • the mesodermal cells selected here are suspended in a culture medium for inducing differentiation, and seeded on a type IV collagen-coated plate at a density of 3 x 10 5 .
  • the culture solution is 3ml per liter, 37 ° C, 5%
  • VE-doherin + vascular endothelial cells were sorted by FACS Prepare endothelial cells.
  • the ES cells of the present invention are also divided into hematopoietic cells.
  • Embryoid bodies are formed by suspending and culturing the ES cells prepared in the above Examples in a basic medium (eg, DMEM) containing KSR. Four to seven days after the formation of embryoid bodies, the embryoid bodies are collected and seeded on a culture dish under gelatin coating. 1 in a basic medium containing KSR
  • the cardiomyocytes are divided.
  • tissue stem cells neural stem cells, mesenchymal stem cells, etc.
  • tissue stem cells nerve stem cells, mesenchymal stem cells, etc.
  • ES cells are collected from the force-quisar, and the same treatment as in Example 9 is performed to divert the cells to nerve cells. Transplanting this transplant of nerve cells into a force-quisar subject with neurological disease shows that the transplant can function.
  • ES cells were collected from humans with consent from the patient, as described in Example 6.
  • Example 14 The processing is performed according to the same procedure as in Example 9 as described in Example 14. It can be seen that transplantation of this transplant of nerve cells into a human subject having a neurological disease after obtaining consent in advance has enabled the transplant to function.
  • Example 1 the primary cultured cells prepared in Example 1 and the cell line prepared in Example 3-4 were appropriately mixed to prepare a feeder-cell preparation, and whether or not a difference in the feeder effect was exhibited. Verify that
  • Example 16 An experiment similar to that in Example 16 is performed using force-quisar as a stem cell.
  • Example 16 As a result, as in Example 16, the effect improved from 10:90 to 90:10 as compared to the case where the feeder cell was used alone was shown. At 5:95 and 95: 5, the same degree of effect is observed as when used alone. In particular, at 20: 80—80: 20, contaminating differentiated cells When observed at about the same amount, it is observed that the number of mixed differentiated cells is reduced to about 1%.
  • Example 16 an experiment similar to that in Example 16 is performed using human cells as stem cells.
  • the protocol is according to Example 16. However, human ES cells are used with the consent of the provider as described above.
  • Example 16 As a result, as in Example 16, up to 10:90-90:10, an improved effect was obtained as compared to the case where the single feeder cell was used alone. At 5:95 and 95: 5, the same degree of effect is observed as when used alone. In particular, at 20: 80-80: 20, the amount of contaminated differentiated cells is reduced to less than about 30%, and when observed in almost equal amounts, the amount of contaminated differentiated cells is reduced to about 1%.
  • Example 16 the same experiment as in Example 16 was performed using human cells as stem cells, and as primary feeder cells, mouse fibroblast primary cultured cells and human epidermal fibroblasts (Clonetics, a division of BioWhittaker® USA; Cambrex, USA) using SL10 and MRC5 (ATCC number CCL-171) as cell lines.
  • the protocol is according to Example 16. However, human ES cells are used with the consent of the provider as described above.
  • Example 16 As a result, as in Example 16, up to 10:90-90:10, an improved effect was exhibited as compared to the case where the single feeder cell was used alone. At 5:95 and 95: 5, the same degree of effect is observed as when used alone. In particular, at 20: 80—80: 20, contaminating differentiated cells When observed at about the same amount, it is observed that the number of mixed differentiated cells is reduced to about 1%.
  • the preparation to be used as a feeder cell is treated with mitomycin C.
  • Mitomycin C is available from Sigma. By treating with mitomycin C, feeder cells do not proliferate, can be stored for a long time, and can be sold.
  • mitomycin C-treated cells can be cryopreserved and sold.
  • cryopreservation the same method as used in conventional primary culture cells can be used. For example, such a method is described in “Genetic Manipulation of ES Cells” Molecular Biology Protocol (Nan-Edo) p 471 — It is described in 479 (In addition, refer to Non-Patent Document 1.) o
  • processing can be performed as follows.
  • the package at the time of sale of the feeder cells prepared in this example is provided, for example, in the form of a cryotube.
  • the processing is as follows.
  • the medium is removed from the culture dish treated with mitomycin C, washed three times with PBS, the medium is cultivated, and cultured for several hours to overnight.
  • the cells are suspended in, for example, 5 ⁇ 10 6 Zml in a cell freezing medium (90% FCS + 10% DMSO). [0339] Add the cell suspension to a cryotube (NUNC 377224) and place the lid tightly.
  • a cell freezing medium 90% FCS + 10% DMSO.
  • the feeder cell preparation for sale prepared in this manner is provided to the seller, preferably in a frozen state.
  • the cells can be thawed in the same manner as normal frozen cells and used as a single feeder cell.
  • the present invention has tremendous utility in regenerative medicine.
  • the usefulness of the preferred embodiment is particularly high, since stem cells (especially ES cells of primates including humans) can be established with a high efficiency of establishment that was previously impossible. Therefore, the present invention has applicability in the field of manufacturing regenerative medicine and pharmaceuticals for treating it.

Abstract

It is aimed at providing a technique by which a stem cell (in particular, a primate ES cell) can be established at such a high efficiency that cannot be achieved by the existing techniques. Namely, it is intended to provide a feeder cell preparation for preparing stem cells containing a normal cell and a cell line. It is also intended to provide a method of preparing a stem cell which comprises the step of culturing a stem cell on a feeder cell preparation containing a normal cell and a cell line. Moreover, it is intended to provide a method of preparing an implant for the regeneration of an organ, a tissue or a cell which comprises: A) the step of providing a stem cell capable of differentiating into a desired organ, tissue or cell; B) the step of culturing the stem cell on a feeder cell preparation for preparing stem cells containing a normal cell and a cell line; and C) the step of differentiating the stem cells into an implant for the regeneration of the desired organ, tissue or cell.

Description

明 細 書  Specification
幹細胞の簡易調製法およびそれに使用するフィーダ一細胞  Simple method for preparing stem cells and feeder-one cells used for them
技術分野  Technical field
[0001] 本発明は、幹細胞の分野にある。より詳細には、本発明は、幹細胞 (特に、霊長類 の胚性幹細胞 (本明細書にぉ 、て以下 ES細胞とも 、う) )の簡易な調製および保存 のための方法に関する。  [0001] The present invention is in the field of stem cells. More specifically, the present invention relates to a method for simple preparation and storage of stem cells (particularly, primate embryonic stem cells (hereinafter, also referred to as ES cells)).
背景技術  Background art
[0002] 再生医療による疾患治療が最近注目を浴びている。再生医療は、臓器移植のほか 、医療機器での補助システムの利用などに代わる治療法としての役割が期待されて いる。しかし、これを臓器または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 ヽな 、。  [0002] Disease treatment by regenerative medicine has recently attracted attention. Regenerative medicine is expected to play a role as an alternative to organ transplantation and the use of auxiliary systems in medical devices. However, this has not been routinely applied to many patients with organ or tissue dysfunction.
[0003] 再生医療の中心にある幹細胞、特に ES細胞は、その医療応用面での可能性から 近年その重要性を増している(非特許文献 1)。ヒト ES細胞はその中でも特に重要で あるがその榭立または安定した継代維持が難しく効率的な利用を阻害している。ヒト を含めた霊長類 ES細胞は、その榭立必要とされる胚盤胞の供給がきわめて少なぐ 少数の胚盤胞力 効率よく ES細胞株の榭立を行う必要がある。従来の方法では胚 盤胞からの榭立の効率は多くてもせ ヽぜぃ 10から 30%程度であると 、われて 、る。  [0003] Stem cells, especially ES cells, which are at the center of regenerative medicine, have recently gained importance due to their potential in medical applications (Non-Patent Document 1). Human ES cells are particularly important among them, but their establishment or stable subculture maintenance is difficult, and they hinder efficient use. Primate ES cells, including humans, require a very small supply of blastocysts to establish them. A small number of blastocysts need to efficiently establish ES cell lines. According to the conventional method, the efficiency of the establishment from the blastocyst is at most about 10 to 30%.
[0004] トランスジェニック動物の作出力 ES細胞を使用することで可能となっている。 ES 細胞とは、通常胚盤胞と呼ばれる発生段階の胚に存在する将来動物個体となる未分 化な細胞群である内部細胞塊 (Inner cell mass, ICM)の細胞を培養することによ つて得られた細胞株である。 ES細胞は 1981年に M. J. Evans と M. H. Kaufma n (非特許文献 2)に続いて、 G. R. Martin (非特許文献 3)によりマウスで多分ィ匕能 を有する細胞株として樹立された。  [0004] Production output of transgenic animals This is made possible by using ES cells. ES cells are obtained by culturing cells of the inner cell mass (ICM), which is an undifferentiated cell group that will be future animal individuals and is present in embryos at the developmental stage, usually called blastocysts. The resulting cell line. ES cells were established in 1981 by M. J. Evans and M. H. Kaufman (Non-Patent Document 2), followed by G. R. Martin (Non-Patent Document 3) as a cell line possibly having a dangling ability in mice.
[0005] ES細胞が、霊長類で榭立され、その応用に対する期待がますます高まる中、これ らの問題を克服するために幹細胞治療とその応用を中心とした再生医学に対する期 待がますます高まっている。  [0005] With the emergence of ES cells in primates and increasing expectations for their applications, there is increasing hope for regenerative medicine, especially stem cell therapy and its applications, to overcome these problems. Is growing.
[0006] 非特許文献 4は、ヒトの皮膚の繊維芽細胞をフィーダ一に利用する論文で榭立は 1 つの内部細胞塊から 1株榭立している力 その効率はまったく低いものである。非特 許文献 5は、ヒト ES細胞の最初の論文である力 その榭立効率は、 5株 Z14内部細 胞塊である。非特許文献 6では STO細胞を用いてヒト ES細胞の榭立を行って 、るが 、その榭立効率は、 3株 Z14内部細胞塊と、まったく低いものにとどまつているのが 現状である。このように、榭立効率において、 50%はおろ力、 30%を達成するのも覚 束な ヽと 、うのが現状の技術水準であると!/、える。 [0006] Non-Patent Document 4 is a paper using fibroblasts of human skin as a feeder, and is found to be 1 The force that establishes one strain from two inner cell masses. Its efficiency is quite low. Non-Patent Document 5 is the first paper on human ES cells. Its establishment efficiency is 5 cell lines inside Z14. In Non-Patent Document 6, human ES cells are established using STO cells. However, the efficiency of the establishment is still very low, such as the internal cell mass of three strains Z14. In this way, in terms of the establishment efficiency, it is expected that 50% will be a downsized force and 30% will be achieved.
[0007] 本発明者らは、特許文献 1にお 、て、培地の最適条件を検討した。しかし、培地を 最適化するだけでは、榭立効率を上げるには限界があり、榭立された ES細胞には分 化細胞が有意に存在する。  [0007] In Patent Document 1, the present inventors have studied the optimal conditions for a medium. However, optimizing the medium alone has a limitation in increasing the establishment efficiency, and differentiated cells are significantly present in established ES cells.
[0008] 他のグループもまた、幹細胞の維持のための方法を開発して 、る(例えば、特許文 献 2、非特許文献 5および 7)。しかし、これらの方法では、フィーダ一細胞は単種類 使用されるのみであり、常に分化細胞が混入する。特に、非特許文献 7では、 50%ほ どが部分的に分ィ匕した細胞集団となっている。  [0008] Other groups have also developed methods for stem cell maintenance (eg, Patent Document 2, Non-Patent Documents 5 and 7). However, in these methods, only one kind of feeder cell is used, and differentiating cells are always contaminated. In particular, in Non-Patent Document 7, about 50% is a partially divided cell population.
[0009] 従って、より分化細胞の混入が低減し、未分化状態の維持がより亢進するために使 用される技術に対する要望が強まっている。  [0009] Therefore, there is an increasing demand for a technique used to reduce the contamination of differentiated cells and further enhance the maintenance of an undifferentiated state.
[0010] (先行技術文献)  [0010] (Prior art documents)
特許文献 1:特開 2003— 116527号  Patent Document 1: JP 2003-116527A
特許文献 2:米国特許第 6200806号  Patent Document 2: US Patent No. 6200806
非特許文献 1 :幹細胞'クローン 研究プロトコール 中辻編、羊土社 (2001) 非特許文献 2 : M. J. Evans & M. H. Kaufman: Nature, 292, 15 Non-patent document 1: Stem cell 'clone research protocol Nakatsuji ed., Yodosha (2001) Non-patent document 2: M.J.Evans & M.H.Kaufman: Nature, 292, 15
4, 1981 4, 1981
非特許文献 3 : G. R. Martin: Proc. Natl. Acad. Sci. USA, 78, 7634, 1981  Non-Patent Document 3: G.R.Martin: Proc. Natl. Acad. Sci. USA, 78, 7634, 1981
非特許文献 4:Hovatta O. , et al. , Human Reproduction Vol. 18, No. 7 pp. 1404-1409, 2003  Non-Patent Document 4: Hovatta O., et al., Human Reproduction Vol. 18, No. 7 pp. 1404-1409, 2003
非特許文献 5 : Thomson A. , et al. , Science. 282 : 1145-1147, 1998.  Non-Patent Document 5: Thomson A., et al., Science.282: 1145-1147, 1998.
非特許文献 6 : Park J. H. et al. , Biology of Reproduction, 2003 onl ine Aug. 20. Non-Patent Document 6: Park JH et al., Biology of Reproduction, 2003 onl ine Aug. 20.
非特許文献 7 :Reubinoff BE, Pera MF, Fong CY, Trounson A, Bo ngso A, Nat Biotechnol. 2000 Apr; 18 (4) : 399-404  Non-Patent Document 7: Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A, Nat Biotechnol. 2000 Apr; 18 (4): 399-404
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
[0011] 本発明は、従来技術では達成し得ない程度の効率 (特に、分化細胞の混入を低減 する)で幹細胞 (特に、ヒトを含む霊長類の ES細胞)を榭立することができる技術を提 供することを課題とする。 [0011] The present invention provides a technique capable of establishing stem cells (especially ES cells of primates including humans) with an efficiency (particularly, reducing contamination of differentiated cells) that cannot be achieved by conventional techniques. The task is to provide
課題を解決するための手段  Means for solving the problem
[0012] 本発明は、従来使用されている、フィーダ一細胞を複数 (例えば、繊維芽細胞初代 培養細胞、繊維芽細胞株など)を混合してフィーダ一細胞として用いると、予想外に 効率よく幹細胞を榭立することができることを見出したことによって上記課題を解決し た。 [0012] The present invention provides an unexpectedly efficient use of a conventionally used feeder cell by mixing a plurality of feeder cells (eg, primary cultured fibroblast cells, fibroblast cell lines, etc.) and using them as a feeder cell. The above problem was solved by finding that stem cells could be established.
[0013] 従って、本発明は、以下を提供する。  Therefore, the present invention provides the following.
[0014] 一つの局面において、本発明は、正常細胞と、細胞株とを含む、幹細胞を調製する ためのフィーダ一細胞調製物を提供する。  [0014] In one aspect, the present invention provides a feeder-one cell preparation for preparing a stem cell, comprising a normal cell and a cell line.
[0015] 一つの実施形態において、上記正常細胞は、初代培養細胞を含む。 [0015] In one embodiment, the normal cells include primary cultured cells.
[0016] 一つの実施形態において、上記正常細胞は、不死化していない細胞である。 [0016] In one embodiment, the normal cells are non-immortalized cells.
[0017] 一つの実施形態にお!、て、上記正常細胞は、繊維芽細胞を含む。 [0017] In one embodiment, the normal cells include fibroblasts.
[0018] 一つの実施形態にお!、て、上記正常細胞は、繊維芽細胞初代培養細胞である。 [0018] In one embodiment, the normal cells are primary cultured fibroblast cells.
[0019] 一つの実施形態において、上記正常細胞は、マウス由来である。 [0019] In one embodiment, the normal cells are derived from a mouse.
[0020] 一つの実施形態において、上記正常細胞は、胎児由来である。 [0020] In one embodiment, the normal cells are derived from a fetus.
[0021] 一つの実施形態において、上記正常細胞は、継代数が 5回以下である。 [0021] In one embodiment, the normal cells have a passage number of 5 or less.
[0022] 一つの実施形態において、上記正常細胞は、マウス 11一 16日齢胎児力 酵素処 理により得られた繊維芽細胞である。 [0022] In one embodiment, the normal cells are fibroblasts obtained by enzymatic treatment of mouse 11-16 days old fetal power.
[0023] 一つの実施形態にお!、て、上記正常細胞は、継代数が 5回以下の初代培養細胞 である。 In one embodiment, the normal cells are primary cultured cells having a passage number of 5 or less.
[0024] 一つの実施形態において、上記細胞株は、不死化している。 [0025] 一つの実施形態にお!ヽて、上記細胞株は、繊維芽細胞株である。 [0024] In one embodiment, the cell line is immortalized. [0025] In one embodiment, the cell line is a fibroblast cell line.
[0026] 一つの実施形態にお!、て、上記細胞株は、継代された期間が 2ヶ月以下である。 [0026] In one embodiment, the cell line has been subcultured for 2 months or less.
[0027] 一つの実施形態において、上記細胞株は、継代された期間が 1ヶ月以下である。 [0027] In one embodiment, the cell line has been passaged for one month or less.
[0028] 一つの実施形態において、上記細胞株は、マウス由来である。 [0028] In one embodiment, the cell line is derived from a mouse.
[0029] 一つの実施形態において、上記細胞株は、胎児由来である。 [0029] In one embodiment, the cell line is derived from a fetus.
[0030] 一つの実施形態において、上記細胞株は、 STO株である。 [0030] In one embodiment, the cell line is an STO line.
[0031] 一つの実施形態において、上記細胞株は、 neo抵抗性である。 [0031] In one embodiment, the cell line is neo-resistant.
[0032] 一つの実施形態において、上記細胞株は、 SL10株である。 [0032] In one embodiment, the cell line is the SL10 line.
[0033] 一つの実施形態において、上記正常細胞と、上記細胞株とは、約 1: 10— 10 : 1で める。 [0033] In one embodiment, the ratio of the normal cells to the cell line is about 1:10 to 10: 1.
[0034] 一つの実施形態において、上記正常細胞と、上記細胞株とは、約 1 : 3— 3 : 1である  [0034] In one embodiment, the normal cells and the cell line are about 1: 3-3: 1.
[0035] 一つの実施形態において、上記正常細胞と、上記細胞株とは、ほぼ等量で存在す る。 [0035] In one embodiment, the normal cells and the cell line are present in approximately equal amounts.
[0036] 一つの実施形態にお!、て、上記幹細胞は、胚性幹細胞である。  [0036] In one embodiment, the stem cells are embryonic stem cells.
[0037] 一つの実施形態において、上記幹細胞は、ヒト胚性幹細胞である。  [0037] In one embodiment, the stem cells are human embryonic stem cells.
[0038] 一つの実施形態において、上記正常細胞は、マウス繊維芽細胞初代培養細胞で あり、上記細胞株は、 SL10株である。  [0038] In one embodiment, the normal cells are mouse fibroblast primary culture cells, and the cell line is SL10 strain.
[0039] 一つの局面において、本発明は、正常細胞と、細胞株とを含むフィーダ一細胞調 製物の上で、幹細胞を培養する工程を包含する、幹細胞を調製するための方法を提 供する。 [0039] In one aspect, the present invention provides a method for preparing a stem cell, comprising a step of culturing the stem cell on a feeder-one-cell preparation containing a normal cell and a cell line. .
[0040] 一つの実施形態において、上記フィーダ一細胞調製物は、上記記載の実施形態 の任意の形態を採り得る。  [0040] In one embodiment, the feeder-one-cell preparation can take any form of the embodiment described above.
[0041] 一つの実施形態において、上記フィーダ一細胞調製物が配置される培養ディッシ ュはゼラチンコーティングされている。 [0041] In one embodiment, the culture dish on which the feeder-cell preparation is placed is gelatin-coated.
[0042] 一つの実施形態にお!、て、上記フィーダ一細胞調製物は、約 1 X 104細胞 /cm2 一約 1 X 105細胞 /cm2で播種される。 [0042] Te it!, In one embodiment, the feeder single cell preparations is plated at about 1 X 10 4 cells / cm 2 one about 1 X 10 5 cells / cm 2.
[0043] 一つの実施形態において、上記フィーダ一細胞調製物は、混合して播種された後 5日以内に使用される。 [0043] In one embodiment, the feeder-one cell preparation is mixed and seeded. Used within 5 days.
[0044] 一つの実施形態において、上記フィーダ一細胞調製物中の上記正常細胞は、マウ ス繊維芽細胞初代培養細胞であり、上記細胞株は、 SL10株である。  [0044] In one embodiment, the normal cells in the feeder-one-cell preparation are mouse fibroblast primary culture cells, and the cell line is SL10 strain.
[0045] 一つの実施形態にお!、て、上記幹細胞は、ノックアウト血清代替添加物 (KSR)を 含む培地中で培養される。 [0045] In one embodiment, the stem cells are cultured in a medium containing a knockout serum replacement additive (KSR).
[0046] 一つの実施形態にお!、て、上記方法は、さらに、継代する工程を包含し、上記継代 工程において、コラゲナーゼが使用される。 [0046] In one embodiment, the method further includes a sub-passing step, wherein collagenase is used in the sub-passing step.
[0047] 別の局面において、本発明は、本発明の方法によって調製される、幹細胞調製物 を提供する。 [0047] In another aspect, the present invention provides a stem cell preparation prepared by the method of the present invention.
[0048] 別の局面において、本発明は、本発明の方法によって調製される、 ES細胞調製物 を提供する。  [0048] In another aspect, the present invention provides an ES cell preparation prepared by the method of the present invention.
[0049] 別の局面において、本発明は、本発明の方法によって調製される、霊長類 ES細胞 調製物を提供する。  [0049] In another aspect, the present invention provides a primate ES cell preparation prepared by the method of the present invention.
[0050] 別の局面において、本発明は、本発明の方法によって調製される、ヒト ES細胞調 製物を提供する。  [0050] In another aspect, the present invention provides a human ES cell preparation prepared by the method of the present invention.
[0051] 別の局面において、本発明は、臓器、組織または細胞を再生するための移植物を 調製するための方法であって: A)所望の臓器、組織または細胞に分化し得る幹細胞 を提供する工程; B)該幹細胞を、正常細胞と、細胞株とを含むフィーダ一細胞の上 で培養する工程;および C)該幹細胞を所望の臓器、組織または細胞を再生するため の移植物へと分化させる工程、を包含する、方法を提供する。  [0051] In another aspect, the present invention provides a method for preparing a transplant for regenerating an organ, tissue or cell, comprising: A) a stem cell capable of differentiating into a desired organ, tissue or cell. B) culturing the stem cells on a feeder cell containing normal cells and a cell line; and C) transforming the stem cells into a transplant for regenerating a desired organ, tissue or cell. Differentiating.
[0052] 一つの実施形態において、上記フィーダ一細胞調製物は、上記フィーダ一細胞調 製物記載の実施形態の任意の形態を採り得る。  [0052] In one embodiment, the feeder-cell preparation can take any form of the embodiments described in the feeder-cell preparation.
[0053] 一つの実施形態にお!、て、前記所望の臓器、組織または細胞は、神経、血球、骨 、軟骨、心臓、心膜、血管、筋肉、眼、肝臓、脾臓、腸、胃、肺、気管、毛および皮膚 力 なる群より選択される。  [0053] In one embodiment, the desired organ, tissue or cell is a nerve, blood cell, bone, cartilage, heart, pericardium, blood vessel, muscle, eye, liver, spleen, intestine, stomach, Lung, trachea, hair and skin.
[0054] 一つの実施形態にお!、て、上記所望の臓器、糸且織または細胞と上記フィーダ一細 胞とは、同じ種である。  [0054] In one embodiment, the desired organ, fibrous tissue or cell and the feeder cell are of the same species.
[0055] 一つの実施形態にお!、て、上記所望の臓器、組織または細胞は霊長類のものであ り、上記フィーダ一細胞はマウス由来である。 [0055] In one embodiment, the desired organ, tissue or cell is a primate. In addition, the feeder cells are derived from a mouse.
[0056] 一つの実施形態において、上記培養は、ェキソビボで行われる。  [0056] In one embodiment, the culturing is performed ex vivo.
[0057] 一つの実施形態にお!、て、上記幹細胞は、被検体力も摘出されてすぐのものであ る力、または凍結保存されたものである。  [0057] In one embodiment, the stem cell is a force that is also obtained immediately after the removal of the test subject, or is a cryopreserved one.
[0058] 一つの実施形態において、上記培養は、 37°C、飽和湿度中で 5%CO下で行わ [0058] In one embodiment, the culturing is performed at 37 ° C in a saturated humidity under 5% CO.
2 れる。  2
[0059] 一つの実施形態にお!ヽて、上記分化は、 DNA脱メチル化剤、ヒストン脱ァセチル ィ匕剤、核内レセプターリガンド、細胞増殖因子、サイト力イン、へキサメチレンビスァセ トアミド、ジメチルァセトアミド、ジブチル cAMP、ジメチォルスルホキシド、ョードデォ キシゥリジン、ヒドロキシル尿素、シトシンァラビノシド、マイトマイシン C、酪酸ナトリウ ム、ァフイディコリン、フルォロデオキシゥリジン、ポリプレンおよびセレンからなる群よ り選択される少なくとも 1つの分ィ匕因子を含む培養液において行われる。  [0059] In one embodiment, the differentiation is performed by using a DNA demethylating agent, a histone deacetylating agent, a nuclear receptor ligand, a cell growth factor, a cytokinin, hexamethylene bisacetamide. Dimethylacetamide, dibutyl cAMP, dimethylsulfoxide, eododexperidine, hydroxylurea, cytosine arabinoside, mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polyprene and selenium. It is performed in a culture solution containing at least one selected factor.
[0060] 他の局面において、本発明は、本発明の臓器、組織または細胞を再生するための 移植物を調製するための方法によって調製される、臓器、組織または細胞を提供す る。  [0060] In another aspect, the present invention provides an organ, tissue or cell prepared by the method for preparing a transplant for regenerating an organ, tissue or cell of the present invention.
[0061] 一つの局面において、本発明は、臓器、組織または細胞を再生するためのシステ ムであって: A)容器;および B)上記容器上に播種される、正常細胞と、細胞株とを含 むフィーダ一細胞、を備える、システムを提供する。  [0061] In one aspect, the present invention provides a system for regenerating an organ, tissue or cell, comprising: A) a container; and B) a normal cell and a cell line seeded on the container. And a feeder cell containing the same.
[0062] 一つの実施形態において、上記フィーダ一細胞調製物は、上記フィーダ一細胞調 製物記載の実施形態の任意の形態を採り得る。 [0062] In one embodiment, the feeder-cell preparation can take any form of the embodiment described in the feeder-cell preparation.
[0063] 一つの局面において、本発明は、正常細胞と細胞株との混合物を提供する。 [0063] In one aspect, the present invention provides a mixture of a normal cell and a cell line.
[0064] 一つの実施形態にお!ヽて、この混合物は、繊維芽細胞初代培養細胞と、繊維芽細 胞株とを含む。 [0064] In one embodiment, the mixture comprises primary fibroblast cells and a fibroblast cell line.
[0065] 一つの実施形態において、上記初代培養細胞は、マウス胎児初代培養繊維芽細 胞である。  [0065] In one embodiment, the primary cultured cells are mouse primary cultured fibroblasts.
[0066] 一つの実施形態において、上記繊維芽細胞株は、 STO株である。  [0066] In one embodiment, the fibroblast cell line is an STO line.
[0067] 一つの実施形態において、上記正常細胞は、マウス繊維芽細胞初代培養細胞で あり、上記細胞株は、 SL10株である。 [0068] 一つの局面において、本発明は、臓器、糸且織または細胞を再生するための方法で あって: A)所望の臓器、組織または細胞に分ィ匕し得る幹細胞を提供する工程; B) 上記幹細胞を、正常細胞と、細胞株とを含むフィーダ一細胞上で培養する工程;およ び C)上記培養された上記幹細胞を上記被検体の処置されるべき部位に移植するェ 程、を包含する、方法を提供する。 [0067] In one embodiment, the normal cells are mouse fibroblast primary culture cells, and the cell line is the SL10 strain. [0068] In one aspect, the present invention relates to a method for regenerating an organ, fibrous tissue or cell, comprising: A) providing a stem cell capable of dividing into a desired organ, tissue or cell; B) a step of culturing the stem cells on a feeder cell containing normal cells and a cell line; and C) a step of transplanting the cultured stem cells to a site of the subject to be treated. A method is provided.
[0069] 一つの実施形態において、上記フィーダ一細胞調製物は、上記フィーダ一細胞調 製物記載の実施形態の任意の形態を採り得る。  [0069] In one embodiment, the feeder-cell preparation can take any form of the embodiments described in the feeder-cell preparation.
[0070] 一つの実施形態において、本発明は、 D)上記幹細胞を分化させる工程をさらに包 含する。  [0070] In one embodiment, the present invention further includes D) a step of differentiating the stem cells.
[0071] 一つの局面において、本発明は、フィーダ一細胞としての、繊維芽細胞初代培養 細胞と、繊維芽細胞株とを含む細胞混合物の使用を提供する。  [0071] In one aspect, the present invention provides use of a cell mixture containing primary fibroblast cells and a fibroblast cell line as a feeder cell.
[0072] 一つの局面において、本発明は、フィーダ一細胞を含む医薬を製造するための、 繊維芽細胞初代培養細胞と、繊維芽細胞株とを含む細胞混合物の使用を提供する  [0072] In one aspect, the present invention provides use of a cell mixture containing a primary fibroblast cell line and a fibroblast cell line for producing a medicament containing a feeder cell.
[0073] 別の局面において、本発明は、フィーダ一細胞調製物としての、正常細胞と、細胞 株とを含む細胞混合物の使用を提供する。 [0073] In another aspect, the present invention provides the use of a cell mixture containing normal cells and a cell line as a feeder-cell preparation.
[0074] 一つの実施形態において、上記フィーダ一細胞調製物は、上記フィーダ一細胞調 製物記載の実施形態の任意の形態を採り得る。 [0074] In one embodiment, the feeder-cell preparation can take any form of the embodiment described in the feeder-cell preparation.
[0075] 一つの実施形態にお!、て、上記フィーダ一細胞調製物は、増殖しな!、ように処理さ れている。 [0075] In one embodiment, the feeder cell preparation is treated so as not to proliferate.
[0076] 一つの実施形態において、上記フィーダ一細胞調製物は、マイトマイシン Cにより 処理されている。  [0076] In one embodiment, the feeder cell preparation is treated with mitomycin C.
[0077] 別の局面において、本発明は、幹細胞の使用が適切である疾患、障害または状態 の処置または予防のための、幹細胞を含む医薬の製造における、正常細胞と、細胞 株とを含むフィーダ一細胞調製物の使用を提供する。  [0077] In another aspect, the present invention provides a feeder comprising a normal cell and a cell line in the manufacture of a medicament containing a stem cell for treating or preventing a disease, disorder or condition in which the use of the stem cell is appropriate. The use of a single cell preparation is provided.
[0078] 一つの実施形態において、上記フィーダ一細胞調製物は、上記フィーダ一細胞調 製物記載の実施形態の任意の形態を採り得る。  [0078] In one embodiment, the feeder-cell preparation can take any form of the embodiments described in the feeder-cell preparation.
[0079] 一つの実施形態にお!、て、上記フィーダ一細胞調製物は、増殖しな!、ように処理さ れている。 [0079] In one embodiment, the feeder-cell preparation is treated so as not to proliferate! It is.
[0080] 一つの実施形態において、上記フィーダ一細胞調製物は、マイトマイシン Cにより 処理されている。  [0080] In one embodiment, the feeder-cell preparation is treated with mitomycin C.
[0081] 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 説明を読みかつ理解すれば、当業者には明白〖こなることが理解される。  [0081] Accordingly, it is understood that these and other advantages of the present invention will be apparent to those of ordinary skill in the art upon reading and understanding the following detailed description, with reference to the accompanying drawings.
発明の効果  The invention's effect
[0082] 本発明によって、特に幹細胞を効率よく榭立する技術が提供される。特に、霊長類 細胞では、本発明 50%以上 (好ましくは 80%以上)という従来達成不可能であった 高い効率で細胞株の樹立が可能となった。特に、本発明のフィーダ一細胞を用いて 調製された幹細胞調製物に含まれる分化細胞が 50%未満であり、 10%未満、ある いは約 1%未満という低率も達成し得ることが明らかになった。従って、本発明は、従 来達成できな力つた高純度の多能性細胞の調製物が提供される。  [0082] The present invention provides a technique for efficiently establishing stem cells in particular. In particular, in primate cells, it has become possible to establish a cell line with a high efficiency of 50% or more (preferably 80% or more) of the present invention, which has not been achieved conventionally. In particular, it is clear that the stem cell preparation prepared using the feeder cell of the present invention contains less than 50% of differentiated cells, and can achieve a low rate of less than 10% or less than about 1%. Became. Thus, the present invention provides a powerful and highly pure preparation of pluripotent cells that could not be achieved conventionally.
図面の簡単な説明  Brief Description of Drawings
[0083] [図 1A]図 1Aは、本発明の方法により樹立したサル ES細胞株の解説写真である。ノ 一は 100 /z mを示す。  FIG. 1A is an explanatory photograph of a monkey ES cell line established by the method of the present invention. No. 1 indicates 100 / zm.
[図 1B]図 1Bは、この方法により樹立したサル ES細胞株の解説写真である。バーは 5 0 μ mを示す。  [FIG. 1B] FIG. 1B is an explanatory photograph of a monkey ES cell line established by this method. Bars indicate 50 μm.
[図 2A]図 2Aは、ヒト ES細胞の写真である。バーは、 275 μ mを示す。  FIG. 2A is a photograph of human ES cells. Bars indicate 275 μm.
[図 2B]図 2Bは、ヒト ES細胞の写真である。バーは、 70 mを示す。  FIG. 2B is a photograph of human ES cells. The bar indicates 70 m.
[図 3]図 3は、未分化状態特異的マーカー発現である。  FIG. 3 shows undifferentiated state-specific marker expression.
[図 4]図 4は、神経細胞への分ィ匕である。  [FIG. 4] FIG. 4 is a diagram illustrating the division of nerve cells.
[図 5]図 5は、テラトーマ形成による多分ィ匕能の証明である。  [FIG. 5] FIG. 5 is a proof of the supposedly terrifying ability by teratoma formation.
[図 6]図 6は、 ES細胞榭立スキーム例を示す。  FIG. 6 shows an example of an ES cell establishment scheme.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0084] 以下、本発明を説明する。本明細書の全体にわたり、単数形の表現は、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。従って、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。したがって、他に定義されない限り、本明細書中で使用さ れる全ての専門用語および科学技術用語は、本発明の属する分野の当業者によつ て一般的に理解されるのと同じ意味を有する。矛盾する場合、本明細書 (定義を含め て)が優先する。 Hereinafter, the present invention will be described. It should be understood that throughout this specification, the use of the singular includes the plural concept unless specifically stated otherwise. Therefore, it is to be understood that singular articles (eg, "a", "an", "the", etc. in English) also include the concept of the plural unless specifically stated otherwise. . Also used in this specification It should be understood that the terms used are used in the meaning commonly used in the art unless otherwise specified. Thus, unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. . In case of conflict, the present specification, including definitions, will control.
[0085] 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し、必要な技術の説 明を行い、本発明を説明する。  [0085] The definitions of terms used particularly in the present specification are listed below, necessary techniques are explained, and the present invention is described.
[0086] 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 意味と同様に定義され、多細胞生物の組織の構成単位であって、外界を隔離する膜 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、へマトキシリンーェォシン (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色素によって染め分け 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)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、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が 挙げられるがそれらに限定されない。  [0086] As used herein, "cell" is defined in the same broadest sense as used in the art, and is a constituent unit of a tissue of a multicellular organism, wrapped in a membrane structure that isolates the outside world. Rarely, it refers to an organism that has a self-renewal ability inside and has genetic information and its expression mechanism. In the present invention, any cell can be targeted. The number of "cells" used in the present invention can be counted through a light microscope. When counting through an optical microscope, count by counting the number of nuclei. The tissue is used as a slice of a tissue section, and extracellular matrix (for example, elastin or collagen) and nuclei derived from cells are stained with a dye by hematoxylin-eosin (HE) staining. This tissue section can be examined under an optical microscope, and the number of nuclei per specific area (for example, 200 m × 200 m) can be counted by estimating the number of cells. The cells used herein may be naturally occurring cells or artificially modified cells (eg, fusion cells, genetically modified cells). The source of cells can be, for example, a single cell culture, or a source of cells from a normally grown transgenic animal embryo, blood, or body tissue, or a normally grown cell line. Such cell mixtures include, but are not limited to.
[0087] 本発明において使用されるフィーダ一細胞として使用される細胞は、正常細胞と細 胞株とが存在し、好ましくは、繊維芽細胞またはその対応物がある限り、どの生物由 来の細胞 (例えば、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)由来の細胞)が用いられてもよい。 1つの実施形態では 、霊長類 (たとえば、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い。別の実施形態では、好ましくは、フィーダ一 細胞としては、げっ歯類細胞が用いられ、より好ましくは、マウス細胞がフィーダ一細 胞として用いられる。 [0087] The cells used as the feeder cells used in the present invention include normal cells and cell lines, and preferably, cells derived from any organism as long as fibroblasts or their counterparts are present. (E.g., mammals (e.g., monotremes, marsupials, oligodonts, skin wings, winged fins, carnivores, carnivores, long nose, odd ungulates, artiodactyla, tubulars, Cells derived from squamous species, marine species, cetaceans, whales, primates, rodents, apogonidae, etc.) may be used. In one embodiment, cells from primates (eg, chimpanzees, macaques, humans), particularly cells from humans, Cells are used, but are not limited thereto. In another embodiment, preferably, the feeder cell is a rodent cell, and more preferably, a mouse cell is used as the feeder cell.
[0088] 本発明で用いられる幹細胞として使用される細胞は、幹細胞またはその対応物が ある限り、どの生物由来の細胞 (例えば、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)由来の細胞)が用いられてもよい 。さらに好ましくは、幹細胞としては、霊長類 (たとえば、チンパンジー、二ホンザル、ヒ ト)由来の細胞が用いられる。最も好ましくは、幹細胞としてはヒト由来の細胞が用い られる。  [0088] The cells used as stem cells used in the present invention include cells derived from any organism (eg, mammals (eg, monopores, marsupials, oligodentates, Skin wings, winged hands, carnivores, carnivores, longnoses, hoofed horses, artiodactyla, tube teeth, scales, squids, cetaceans, primates, primates, rodents, etc. ) -Derived cells) may be used. More preferably, cells derived from primates (for example, chimpanzees, Japanese macaques, and humans) are used as stem cells. Most preferably, human-derived cells are used as the stem cells.
[0089] 本明細書において「対応物」とは、例えば、細胞について用いられる場合、ある種の 生物中の細胞に関し、同様の性質および Zまたは機能を有する別の種の細胞をいう 。そのような同様の性質は、分泌するサイト力イン、増殖因子、発現する細胞マーカ 一などによって同定することができる。  [0089] As used herein, the term "counterpart", for example, when used for a cell, refers to a cell in a certain organism, and refers to a cell of another species having similar properties and Z or function. Such similar properties can be identified by secreted cytokins, growth factors, expressed cellular markers, and the like.
[0090] 本明細書にぉ 、て「幹細胞」とは、自己複製能を有し、多分化能 (すなわち多能性)  [0090] As used herein, the term "stem cell" has a self-renewal ability and a pluripotency (that is,
(「pluripotency」)を有する細胞をいう。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。本明細書では幹細胞は、 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)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 (たと えば、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。 ("Pluripot e ncy"). Stem cells can usually regenerate tissue when it is damaged. As used herein, a stem cell can be, but is not limited to, an ES cell or a tissue stem cell (also a tissue stem cell, a tissue-specific stem cell, or a somatic stem cell). In addition, artificially created cells (eg, reprogrammed cells, etc.) can also be stem cells, as long as they have the above-mentioned ability.
[0091] 本明細書において「胚性幹細胞」および「ES細胞」は、交換可能に用いられ、初期 胚に由来する任意の多能性幹細胞をいう。通常 ES細胞は、全能性またはほぼ全能 性を有するとされる。この ES細胞を正常な宿主胚盤胞へ導入し仮親子宮へ戻すこと によってキメラ作製を行ったところ、高いキメラ形成能を持つ、生殖系列キメラ (ES細 胞由来の機能的生殖細胞を持つキメラマウス)が得られた (A. Bradley et al. : N ature, 309, 255, 1984)。 ES細胞株は、培養下で、種々の遺伝子導入法(例えば リン酸カルシウム法、レトロウイルスベクター法、リボゾーム法、エレクト口ポレーシヨン 法等)の適用が可能である。また、遺伝子が組込まれた細胞を選別する方法を工夫 し、相同遺伝子組換え(homologous recombination)を利用し、特定の遺伝子を 狙って改変(置換、欠失、挿入)させた細胞のクローンを得ることもできる。インビトロで このような処理をした ES細胞株は生殖系列への分ィ匕能を保持することから、ある特 定の遺伝子の機能を個体レベルで調べる研究が現在盛んに行われている(M. R. Capecchi: Science, 244, 1288, 1989)。 ES細胞を利用したトランスジエニックマ ウス作出法は、ある特定の遺伝子のみを任意に改変させた個体を得ることを可能に した点でマイクロインジェクション法によるトランスジエニック動物作出法にはない多く の利点が考えられる。特に、特定の遺伝子を不活ィ匕させたノックアウト動物を作出で きるようになり、遺伝子の機能を解明したり、外来性の遺伝子のみを発現させることが できる。従って、 ES細胞の榭立が容易になれば、その効果は図り知れない。 [0091] As used herein, "embryonic stem cells" and "ES cells" are used interchangeably and refer to any pluripotent stem cells derived from an early embryo. Generally, ES cells are considered to be totipotent or nearly totipotent. When the chimera was prepared by introducing the ES cells into a normal host blastocyst and returning to the foster parent uterus, a germline chimera (a chimeric mouse having functional germ cells derived from ES cells) having high chimera-forming ability was obtained. ) Was obtained (A. Bradley et al .: Nature, 309, 255, 1984). Various gene transfer methods (for example, a calcium phosphate method, a retrovirus vector method, a ribosome method, an electoral poration method, etc.) can be applied to the ES cell line in culture. We also devised a method to select cells into which genes have been incorporated. However, it is also possible to obtain a clone of a cell that has been modified (substitution, deletion, insertion) by targeting homologous gene recombination (homologous recombination). Since ES cell lines treated in this way retain their ability to germline in vitro, studies to examine the function of certain genes at the individual level are currently being actively conducted (MR Capecchi : Science, 244, 1288, 1989). The transgenic mouse production method using ES cells has many advantages over the transgenic animal production method using microinjection in that it makes it possible to obtain individuals in which only certain genes have been arbitrarily modified. Benefits are conceivable. In particular, a knockout animal in which a specific gene has been inactivated can be produced, and the function of the gene can be elucidated, or only the exogenous gene can be expressed. Therefore, if the establishment of ES cells is facilitated, the effect is immeasurable.
[0092] 本明細書において「組織幹細胞」とは、 ES細胞とは異なり、分化の方向が限定され ている細胞をいう。通常、組織幹細胞は、組織中の特定の位置に存在し、未分化な 細胞内構造をしている。従って、組織幹細胞は多能性のレベルが低い。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、多分ィ匕 能を有し、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、幹細胞としては、好ましくは ES細胞が使用され得るが組織幹 細胞もまた維持の対象であり得る。 [0092] As used herein, the term "tissue stem cell" refers to a cell that has a limited direction of differentiation, unlike ES cells. Normally, tissue stem cells are present at specific positions in a tissue and have an undifferentiated intracellular structure. Thus, tissue stem cells have a low level of pluripotency. Tissue stem cells are poor in organelles with a high nuclear Z cytoplasmic ratio. Tissue stem cells generally have a probable ability to maintain their proliferative capacity over the life of an individual whose cell cycle is slow. As used herein, ES cells are preferably used as stem cells, but tissue stem cells may also be the object of maintenance.
[0093] ES細胞株は、キメラマウス、ノックアウトマウスなどの作製において非常に重要であ り、これらの技術により遺伝子機能の解析が飛躍的に進歩した。 ES細胞から特定の 糸且織を分ィヒ誘導する系の開発が進んでおり、移植医療への応用の現実味を帯びて いる。また、ヒト ES細胞を利用した臨床応用を考えた場合、サルなどの適切なモデル 動物の ES細胞を利用して前臨床研究をすることは必須に近い程度に重要であると 考えられ、その榭立もまた重要である。  [0093] ES cell lines are extremely important in producing chimeric mice, knockout mice, and the like, and the analysis of gene function has been dramatically advanced by these techniques. The development of a system for inducing specific fibrous tissue from ES cells has been progressing, and the system has been applied to transplantation medicine. Considering clinical application using human ES cells, preclinical research using ES cells from appropriate model animals such as monkeys is considered to be almost as important as essential. Standing is also important.
[0094] ES細胞株の榭立は、フィーダ一細胞を用いて行われる。通常、 ES細胞は、胚盤胞 力 分離した内部細胞塊 (ICM)をフィーダ一細胞上で培養することによって榭立さ れる。ここで従来は、マウス胎仔繊維芽細胞あるいはそれ由来の細胞株 STOが単独 で用いられてきた。しかし、これらの細胞では、榭立効率が低ぐ特に霊長類幹細胞 では、 10%— 30%程度と顕著に低ぐ分化細胞が混入することが報告されている。ま た、この効率は、使用する胚盤胞のステージ、 ICMの分離法などによっても変動する 。従って、榭立効率が低い現在の系では、榭立に成功するまでには何回も試行錯誤 が必要であった。本発明を用いると、効率は 80%以上となり、分化細胞の混入も激 減し、そのような試行錯誤の必要はなくなる。 [0094] Establishment of an ES cell line is performed using a single feeder cell. Usually, ES cells are established by culturing a blastocyst-separated inner cell mass (ICM) on a single feeder cell. Heretofore, mouse fetal fibroblasts or cell line STO derived therefrom have conventionally been used alone. However, it has been reported that these cells have a low establishment efficiency, especially primate stem cells, and contaminated cells with a remarkably low level of about 10% to 30%. Ma In addition, this efficiency varies depending on the stage of the blastocyst used, the ICM separation method, and the like. Therefore, in the current system with low establishment efficiency, several trials and errors were necessary to succeed in establishment. Using the present invention, the efficiency is over 80%, the contamination of differentiated cells is drastically reduced, and the need for such trial and error is eliminated.
[0095] 次に、本発明において用いることが可能な ES細胞の一般的な榭立法を解説する。  Next, a general method for establishing ES cells that can be used in the present invention will be described.
マウスで説明すると、通常、受精後 3. 5日で胚は胚盤胞に発生する。胚盤胞から胚 本体を形成する未分化幹細胞である内部細胞塊を免疫手術により分離する。この IC Mは、胎盤などが作る細胞である栄養外胚葉への分化能を有する。このため、 3. 5 日胚から直接 ICMを分離すると ICMが栄養外胚葉へと分化することから未分化細胞 力 S喪失することがよくあるとされる。 3. 5日胚をさら〖こ 1日培養し、 4. 0-4. 5日胚とす ると、胚盤胞は透明帯を脱出する。このステージでは ICMはもはや栄養外胚葉には 分化しない。このような状態から、 ICMを分離し、培養を開始すると、未分化細胞の 割合を高めることができる。従って、本発明における幹細胞の榭立の際も、このような 日数の未分ィ匕細胞を用いることができる。また、ヒト ES細胞の場合は、この日数を参 考にして、適宜変更して用いることができる。ヒトの場合は、受精後 5— 8日のもの(凍 結期間を除く)を使用することが多いが、それに限定されない。  In terms of mice, embryos usually develop into blastocysts 3.5 days after fertilization. The inner cell mass, which is an undifferentiated stem cell forming the embryo body, is separated from the blastocyst by immunosurgery. This ICM has the ability to differentiate into trophectoderm, a cell made by the placenta and the like. Therefore, if ICM is directly isolated from 3.5-day embryos, undifferentiated cell strength S is often lost due to the differentiation of ICM into trophectoderm. 3. After culturing the 5-day embryo for 1 day, 4.0-4.5 days, the blastocyst escapes the zona pellucida. At this stage, the ICM no longer differentiates into trophectoderm. From such a state, if the ICM is separated and culture is started, the ratio of undifferentiated cells can be increased. Therefore, even when stem cells are established in the present invention, undivided cells having such a number of days can be used. In addition, in the case of human ES cells, the number of days can be used as a reference and used as appropriate. In the case of humans, those that are 5-8 days after fertilization (excluding the freezing period) are often used, but are not limited thereto.
[0096] ES細胞を榭立する際には、胚盤胞は、フィーダ一細胞上に直接培養することもで きるが、直接の場合は、胚盤胞が接着し栄養外胚葉が伸展して ICMが露出すること 力もあまり好ましくない。これを分離し培養したものを用いてもよいが、好ましくは免疫 手術が用いられる。模式的な ES細胞株榭立スキームを図 6に示す。簡単に言うと、 免疫手術で分離した ICMをフィーダ一細胞上で培養し、継代を続けると分ィ匕した細 胞に混じって、未分化幹細胞のコロニーが現れる。これを分離し、継代を行うことで、 安定して維持することができる ES細胞株を榭立することができる。本発明を用いると 、榭立率が 80%を超えることから、安定して維持することができる ES細胞を得る効率 も格段に上がる。  [0096] When establishing ES cells, blastocysts can be cultured directly on feeder cells, but in those cases, blastocysts adhere and nutritive ectoderm extends. Exposure of the ICM is also less favorable. A culture obtained by separating and culturing this may be used, but immunosurgery is preferably used. Fig. 6 shows a schematic ES cell line establishment scheme. Briefly, ICM isolated by immunosurgery is cultured on a single feeder cell, and as the passage is continued, undifferentiated stem cell colonies appear in the mixed cells. By separating and subculturing this, an ES cell line that can be stably maintained can be established. According to the present invention, since the establishment rate exceeds 80%, the efficiency of obtaining ES cells that can be stably maintained is significantly improved.
[0097] ES細胞を榭立するためには、本発明のフィーダ一細胞のほ力、 PBS (Ca、 Mg不 含)、 0. 25%トリプシン ' ImM EDTA in PBS、 ES細胞用培地(DMEM (ノヽィグ ルコース) 400ml、ゥシ胎仔血清 100ml、非必須アミノ酸溶液(Gibco) 4ml、ヌクレ オシド溶液(ヌクレオシド、グアノシン、シチジン、ゥリジン各 3mM、チミジン ImM水 溶液、 40°Cに加熱して溶解し、濾過滅菌し、 20°Cで保存する) 4ml、 2 -メルカプト エタノール 4 1(0. ImM)、 LIF 100 ^ l (2000U/ml) ) , Μ2培地(Sigmaなどか ら入手可能)、 M16培地(Sigmaなど力も入手可能)、酸性タイロード液(Sigmaなど から入手可能)、抗マウス血清 (マウス脾臓細胞またはリンパ球 5 X 108細胞をゥサギ に 2週間おきに 3回静脈免疫し、最終免疫から 2週間後に採血する。これを抗血清と して非働化し 80°Cで保存する)、モルモット補体、流動パラフィン (軽質;ナカラィテ スクなど力 入手可能)を用意することが必要である。実験器具としては、 COインキ [0097] In order to establish the ES cells, the feeder cell of the present invention was used to prepare the cells of the feeder, PBS (containing no Ca and Mg), 0.25% trypsin 'ImM EDTA in PBS, a medium for ES cells (DMEM ( Noigram course) 400ml, fetal calf serum 100ml, non-essential amino acid solution (Gibco) 4ml, Nucle Oside solution (nucleoside, guanosine, cytidine, peridine 3 mM each, thymidine ImM aqueous solution, dissolve by heating at 40 ° C, filter sterilize, and store at 20 ° C) 4 ml, 2-mercaptoethanol 4 1 (0. ImM), LIF 100 ^ l (2000 U / ml)), Μ2 medium (available from Sigma etc.), M16 medium (available from Sigma etc.), acidic Tyrode's solution (available from Sigma etc.), anti-mouse serum (5 x 10 8 mouse spleen cells or lymphocytes are immunized intravenously three times every 2 weeks into a egret, and blood is collected 2 weeks after the final immunization. This is inactivated as an antiserum and stored at 80 ° C ), Guinea pig complement, and liquid paraffin (light; available as nacalitesk) are necessary. As an experimental device, CO ink
2 ュベータ、実体顕微鏡、キヤビラリ一、マルチウエルプレート(例えば、 4ゥエル、 12ゥ エル、 NUNCなど力 入手可能)を用意するとよい。  It is advisable to prepare a 2 incubator, a stereo microscope, a capillary, and a multi-well plate (for example, 4-well, 12-well, NUNC, etc. are available).
[0098] 以下に ES細胞の榭立の例示的なプロトコール記載する。このプロトコールは、京都 大学再生医科学研究所においてまとめられたヒト ES細胞株榭立計画書に準じて記 載される力 本明細書では、この特定のプロトコールに限定されず、いずれのプロトコ ールであっても用いることができる。  [0098] An exemplary protocol for establishing ES cells is described below. This protocol is based on the protocol described in the Human ES Cell Line Establishment Program compiled at the Institute of Regenerative Medicine at Kyoto University. In this specification, the protocol is not limited to this particular protocol, but may be any protocol. Can also be used.
[0099] (1)凍結胚の解凍と胚盤胞期までの培養  [0099] (1) Thawing of frozen embryos and culture up to the blastocyst stage
凍結保存されたヒト受精卵または胚盤胞期までの初期胚を順次解凍して培養を行 う。個々の凍結胚容器からは提供者を同定できるものは除去されているため、各回の 解凍 ·培養実験に使用されたヒト受精胚の出自は同定され得ない。し力 ながら、各 々のヒト受精胚の取扱がおろそかにならないように、凍結胚として受け入れた時点か らひとつの凍結容器内に納められたヒト受精胚を榭立研究の過程で個々の存在とし て尊重し、どのような経過をたどつたかを記録する。  Cryopreserved human fertilized eggs or early embryos up to the blastocyst stage are sequentially thawed and cultured. Since the substance that can identify the donor is removed from each frozen embryo container, the origin of the human fertilized embryo used in each thawing and culture experiment cannot be identified. However, in order to prevent the handling of each human fertilized embryo from being neglected, the human fertilized embryos stored in one cryocontainer from the time of their acceptance as frozen embryos were identified as individual entities during the establishment research. And record what progress has been made.
[0100] (2)内部細胞塊の分離と細胞株の榭立  (2) Separation of inner cell mass and establishment of cell line
胚盤胞期まで到達した胚 (受精後の発生期間が約 14日以内のもの)について、抗 ヒト血清による免疫手術などの方法によって内部細胞塊を分離したのち、フィーダ一 細胞層の上で培養する。フィーダ一細胞としては、本発明のフィーダ一細胞を用いる 。内部細胞塊を採取した後の残部についても、礼節をもって取扱う。フィーダ一細胞 の上で増殖した細胞を適時に解離して分割継代し、幹細胞と思われる細胞コロニー の選別培養などを行ないながら、 ES細胞と思われる細胞株を榭立する。この間に、 培養維持方法や細胞凍結保存方法などの改良を目指した研究を行う。 Embryos that have reached the blastocyst stage (the development period after fertilization is about 14 days or less) are separated from the inner cell mass by immunosurgery with anti-human serum, and then cultured on the feeder cell layer I do. The feeder cell of the present invention is used as the feeder cell. The remainder after collecting the inner cell mass is also handled with courtesy. The cells grown on a single feeder cell are dissociated in a timely manner, divided and subcultured, and cell cultures that are likely to be ES cells are established while performing selective culture of cell colonies that are thought to be stem cells. During this time, We will conduct research aimed at improving the culture maintenance method and cell cryopreservation method.
[0101] (3)幹細胞マーカー発現の有無および染色体検定  [0101] (3) Stem cell marker expression and chromosome assay
ES細胞であることを確認するために、幹細胞マーカー(アルカリ性フォスファターゼ 活性や特異的抗原)の検出を行なう。また核型解析を行なって染色体数や形態が正 常かどうかを検定する。  Detection of stem cell markers (alkaline phosphatase activity and specific antigens) is performed to confirm that the cells are ES cells. In addition, karyotype analysis is performed to test whether the number and morphology of chromosomes are normal.
[0102] (4)分化能の検定  [0102] (4) Differentiation ability test
培養下での分ィ匕能を検定するために、培養条件の変更や細胞凝集塊作成による細 胞分化の誘導と各種機能細胞への分化能の解析を行なう。また免疫不全マウスなど への移植を行なってテラトーマ形成による組織分ィ匕能の解析を行なう。  In order to test the ability to divide cells in culture, induction of cell differentiation by changing culture conditions and creation of cell aggregates and analysis of the ability to differentiate into various functional cells are performed. In addition, transplantation into an immunodeficient mouse or the like is performed to analyze the tissue division ability by teratoma formation.
[0103] (5)安全性確保と事故防止  [0103] (5) Ensuring safety and preventing accidents
ヒト凍結胚の一時的保存は専用の液体窒素タンクを用いることによって、榭立計画に 用いる以外の細胞や動物胚などに由来するウィルスと微生物による汚染を防ぐ。また 細胞培養実験には専用の炭酸ガスインキュベーターを用いることによって、他種類の 細胞との混合を防ぎ、ウィルスや微生物による汚染の可能性を小さくする。細胞培養 に用いた培養液や培養器具は、実験室内で加圧高温滅菌処理を行なったのちに廃 棄する。ヒト受精胚の保存および細胞株榭立を行なう実験室への入室者の管理を厳 重に行う。  Temporary storage of human frozen embryos will use a dedicated liquid nitrogen tank to prevent contamination by viruses and microorganisms derived from cells and animal embryos other than those used for the establishment plan. In addition, by using a dedicated carbon dioxide incubator for cell culture experiments, mixing with other types of cells is prevented, and the possibility of contamination by viruses and microorganisms is reduced. The culture medium and culture equipment used for cell culture are subjected to autoclaving in a laboratory and then discarded. Strictly control the entry to the laboratory where human fertilized embryos are stored and cell lines are established.
[0104] 本明細書において「多能性」または「多分ィ匕能」とは、互換可能に用いられ、細胞の 性質をいい、 1以上、好ましくは 2以上の種々の組織または臓器に分ィ匕し得る能力を いう。従って、「多能性」および「多分化能」は、本明細書においては特に言及しない 限り「未分化」と互換可能に用いられる。通常、細胞の多能性は発生が進むにつれて 制限され、成体では一つの組織または器官の構成細胞が別のものの細胞に変化す ることは少ない。従って多能性は通常失われている。とくに上皮性の細胞は他の上皮 性細胞に変化しにくい。これが起きる場合通常病的な状態であり、化生 (metaplasia )と呼ばれる。し力し間葉系細胞では比較的単純な刺激で他の間葉性細胞にかわり 化生を起こしやすいので多能性の程度は高い。 ES細胞は、多能性を有する。組織 幹細胞は、多能性を有する。本明細書において、多能性のうち、受精卵のように生体 を構成する全ての種類の細胞に分ィ匕する能力は全能性といい、多能性は全能性の 概念を包含し得る。ある細胞が多能性を有するかどうかは、たとえば、体外培養系に おける、胚様体 (Embryoid Body)の形成、分ィ匕誘導条件下での培養等が挙げら れるがそれらに限定されない。また、生体を用いた多能性の有無についてのアツセィ 法としては、免疫不全マウスへの移植による奇形種 (テラトーマ)の形成、胚盤胞への 注入によるキメラ胚の形成、生体組織への移植、腹水への注入による増殖等が挙げ られるがそれらに限定されない。 [0104] In the present specification, "pluripotency" or "probably dandelion" is used interchangeably and refers to a property of a cell, and is divided into one or more, preferably two or more various tissues or organs. The ability to dagger. Therefore, “pluripotency” and “pluripotency” are used interchangeably with “undifferentiated” in this specification unless otherwise specified. Normally, cell pluripotency is limited as development progresses, and in adults, the constituent cells of one tissue or organ rarely change into cells of another. Thus pluripotency is usually lost. In particular, epithelial cells are unlikely to change into other epithelial cells. When this happens, it is usually a pathological condition and is called metaplasia. Forced mesenchymal cells have a high degree of pluripotency because metastasis is likely to occur in other mesenchymal cells with relatively simple stimulation. ES cells have pluripotency. Tissue Stem cells are pluripotent. In the present specification, among pluripotency, the ability to divide into all kinds of cells constituting a living body such as a fertilized egg is called totipotency, and pluripotency is totipotency. It can encompass concepts. Whether a certain cell has pluripotency includes, but is not limited to, for example, formation of an embryoid body (Embryoid Body) in an in vitro culture system, culture under conditions for inducing induction, and the like. Assays for the presence or absence of pluripotency using living organisms include the formation of teratomas by transplantation into immunodeficient mice, the formation of chimeric embryos by injection into blastocysts, and the transfer into living tissues. And proliferation by infusion into ascites, but are not limited thereto.
[0105] 本明細書において、多能性のうち、受精卵のように生体を構成する全ての種類の 細胞に分ィ匕する能力は「全能性」といい、多能性は全能性の概念を包含し得る。ただ し、明確に区別する場合は、全能性と多能性とは区別され得、前者はどのような細胞 へも分ィ匕し得る能力をいい、後者は、複数の方向を有するが、生物が可能なすべて の方向には分ィ匕できない能力を有することをいう。また、 1つの方向にのみ分ィ匕する 能力は、単能性ともいう。  [0105] In the present specification, among the pluripotency, the ability to divide into all kinds of cells constituting a living body such as a fertilized egg is called "totipotency", and pluripotency is a concept of totipotency. May be included. However, when the distinction is made clearly, totipotency and pluripotency can be distinguished, the former refers to the ability to divide into any cell, and the latter has multiple directions, Has the ability to divide in all possible directions. The ability to divide in only one direction is also referred to as singularity.
[0106] 本明細書において全能性と多能性とは、例えば、受精後の日数により判定すること ができ、例えば、マウスであれば、受精後約 8日を基準として区別され得る。理論に束 縛されないが、マウスでは、受精後、以下のような経過をたどることが通常である。受 精後 6. 5日(E6. 5とも表記する)では、原始線条 (原条ともいう)がェピブラストの片 側に出現し、胚の将来の前後軸が明らかになる。原条は、胚の将来の後方端を示し 、外胚葉を横切って円筒の遠位端まで達する。原条は、細胞運動が行われる領域で あり、その結果、将来の内胚葉と中胚葉とが形成されることになる。 E7. 5までに結節 の前方に頭部突起が出現し、この部分には脊索と、それを取り囲んで下層には将来 の内胚葉、上層には神経板が形成されることになる。結節は、 E6. 5日ごろカゝら現れ 、後方へと移動し、軸構造が前から後ろへと形成される。 E8. 5日までに胚は幾分丈 が長くなり、その前端には大部分前方神経板力もなる大きな頭部ヒダが形成される。 体節は E8日力 1. 5時間に 1個の割合で前方力も後方へと形成され始める。この時 期を越えた細胞は、仮に胎盤に戻したとしても、脱分ィ匕をしない限りもはや全能性を 示さず、個体を形成しない。これより前では特別の処理をしなくても全能性を示し得る ことから、この点が全能性の分岐点であるといえる。このことは、 ES細胞がこれ以降の 胚力 樹立することが困難であり、これ以降の胚力 は通常 EG (生殖細胞由来)細胞 と呼ばれる細胞が榭立されることから、そのような意味でも分岐点であるといえる。 [0106] In the present specification, totipotency and pluripotency can be determined, for example, by the number of days after fertilization. For example, in the case of mice, they can be distinguished on the basis of about 8 days after fertilization. Without being bound by theory, mice typically follow the following course after fertilization. 6.5 days after fertilization (also referred to as E6.5), the primitive streak (also referred to as the original streak) appears on one side of the epiblast, revealing the future anterior-posterior axis of the embryo. The original streak indicates the future posterior end of the embryo, extending across the ectoderm to the distal end of the cylinder. The original streak is the area where cell motility takes place, resulting in the formation of future endoderm and mesoderm. By E7.5, a head process appears in front of the nodule, which forms the notochord, surrounding it, the future endoderm in the lower layer and the neural plate in the upper layer. The nodules appear around E6.5 and move backwards, forming an axial structure from front to back. E8.5 By day 5, the embryos have grown somewhat longer, and large anterior folds are formed at their anterior ends, most of which also have anterior neural plate strength. The somites begin to evolve backwards at a rate of one every 1.5 hours at E8. Cells beyond this time will no longer exhibit totipotency and will not form an individual, even if they are returned to the placenta, unless dissociated. Before this point, it is possible to show totipotency without any special treatment, so this point can be said to be a branch point of totipotency. This means that it is difficult for ES cells to establish subsequent embryonic power, and subsequent embryonic power is usually EG (germ cell-derived) cells In this sense, it is a divergence point because a cell called と is established.
[0107] 本明細書において「胎児」または「胎仔」とは、交換可能に用いられ、哺乳動物の子 が各器官原基の分化を完了し,成長期に入ったとき力 出産までの期間にある生物 体をいう。  [0107] As used herein, the term "fetus" or "fetus" is used interchangeably and refers to a period in which the offspring of a mammal complete differentiation of each organ primordium and enter the anagen phase until power delivery. A certain organism.
[0108] 本明細書において、「榭立された」または「確立された」細胞とは、特定の性質 (例え ば、多分化能)を維持し、かつ、細胞が培養条件下で安定に増殖し続けるようになつ た状態をいう。したがって、榭立された幹細胞は、多分化能を維持する。榭立された 分化細胞は、特定の確定した機能を有する。榭立された分化細胞は癌化しているこ とが多いが、それに限定されない。  [0108] As used herein, an "established" or "established" cell refers to a cell that maintains a specific property (eg, pluripotency) and proliferates stably under culture conditions. It is the state that has started to continue. Therefore, established stem cells maintain pluripotency. Established differentiated cells have certain defined functions. Established differentiated cells are often cancerous, but are not limited thereto.
[0109] 本明細書において本明細書において「分化」とは、一般的には、 1つの系が 2っ以 上の質的に異なる系に分離することをいい、細胞、糸且織または臓器について用いら れるとき、機能および Zまたは形態が特殊ィ匕することをいう。分化に伴い、通常、多能 性は減少または消失する。  [0109] As used herein, the term "differentiation" as used herein generally refers to the separation of one system into two or more qualitatively different systems. When used for, it means that the function and Z or form are special. With differentiation, pluripotency usually decreases or disappears.
[0110] 本明細書にぉ 、て「分ィ匕 (した)細胞」とは、機能および形態が特殊化した細胞 (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、脾管細胞、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 もよい。分ィ匕細胞の幹細胞調製物への混入は、幹細胞の利用において有害であるこ とが多ぐそのような分ィ匕細胞の混入を減少させることは従来技術では困難であった  [0110] As used herein, the term "divided cells" refers to cells whose functions and morphology are specialized (for example, muscle cells, nerve cells, and the like). No or little potential. The differentiated cells include, for example, epidermal cells, spleen parenchymal cells, splenic duct cells, hepatocytes, blood cells, cardiomyocytes, skeletal muscle cells, osteoblasts, skeletal myoblasts, nerve cells, vascular endothelial cells, and pigments Cells, smooth muscle cells, fat cells, bone cells, chondrocytes and the like. When used in the present invention, the sorting cells may be in the form of a population or a tissue. Incorporation of shunt cells into a stem cell preparation is often detrimental in the use of stem cells. It has been difficult in the prior art to reduce such shunt cells.
[0111] 本明細書において「体細胞」とは、卵子、精子などの生殖細胞以外の細胞であり、 その DNAを次世代に直接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、 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よい。 [0111] In the present specification, "somatic cells" are cells other than germ cells such as eggs and sperm, and refer to all cells that do not directly transfer their DNA to the next generation. Somatic cells usually have a limited or pluripotent force or disappear. As used herein, somatic cells may be naturally occurring or genetically modified.
[0112] 本明細書において「正常細胞」とは、病理状態にない任意の細胞をいう。従って、 そのような正常細胞は、分裂回数が有限であることが多い。そのような有限の分裂回 数は、通常 50— 80回であることが多い。 [0112] As used herein, "normal cells" refers to any cells that are not in a pathological state. Therefore, such normal cells often have a finite number of divisions. Such finite split times The number is usually 50-80 times.
[0113] 本明細書において「不死化」とは、有限の分裂回数 (例えば、 50— 80回)を超えて も細胞が死滅しな 、ことを 、う。  [0113] As used herein, "immortalization" refers to the fact that cells do not die even after a finite number of divisions (for example, 50 to 80 times).
[0114] 本明細書にぉ 、て「細胞株」または「株化細胞」とは、上記有限の分裂回数を超え ても生存する細胞をいう。そのような細胞株は、初代培養細胞など力 得られた継代 細胞を順次継代し、正常細胞が通常死滅する継代数を超えて細胞を継代した後も 生存する細胞を選択することによって榭立することができる。そのような榭立方法は、 当該分野において公知である。  [0114] As used herein, the term "cell line" or "cell line" refers to a cell that survives the above-mentioned finite number of divisions. Such cell lines are obtained by sequentially subculturing passaged cells, such as primary cultured cells, and selecting cells that survive after passage beyond the number of passages at which normal cells usually die. Can be established. Such establishment methods are known in the art.
[0115] 本明細書にぉ ヽて「繊維芽細胞」とは、支持組織の繊維成分を供給し,繊維性結 合組織の重要な成分をなす細胞をいう。組織切片図では、扁平で長目の外形をもち 、不規則な突起を示すことが多い。細胞質は、ミトコンドリア、ゴルジ体、中心体、小脂 肪球などを含むが、そのほかに特殊な分ィ匕は示さない。核は楕円形をしており、しば しば膠原繊維に密接して存在する。  [0115] As used herein, the term "fibroblast" refers to a cell that supplies a fiber component of a supporting tissue and forms an important component of a fibrous connective tissue. Tissue section diagrams often have a flat, long outline and irregular projections. The cytoplasm contains mitochondria, Golgi apparatus, centrosomes, small fat globules, etc., but does not show any special restriction. The nucleus is elliptical and often lies close to collagen fibers.
[0116] 本明細書において「フィーダ一層」または「フィーダ一細胞」 (feeder layerまたは f eeder cell)とは、互換可能に用いられ、培養基質に設けられる、単独では培養維 持することのできない細胞種の増殖および Zまたは分ィ匕形質発現を可能にするよう な、他の細胞種による支持細胞層をいう。組織細胞には、通常の細胞培養条件下で は,分ィ匕形質発現はもとより増殖すらできない細胞種も多いといわれており、そのよう な細胞としては、例えば、幹細胞 (特に、 ES細胞、神経幹細胞、間葉系幹細胞、造 血幹細胞など)、胚性生殖細胞 (EG細胞)、生殖幹細胞 (GS細胞)などが挙げられる がそれらに限定されない。これらの細胞種は一般に、栄養要求性が高く特異的な増 殖因子、分化誘導因子を必要とする。このような細胞種でも生体内でのその細胞種 の支持細胞あるいはそれと類似の細胞が形成する特定の細胞の層を培養基質に活 用することで、培養される細胞種が要求する因子および Zまたは栄養源が供給され ることによって、増殖および分ィ匕をするようになる。フィーダ一細胞として用いる細胞 種は、対象となる細胞種によって選択されるが、 UV照射などの方法で細胞増殖を抑 制して用いることが多い。生殖細胞、 ES細胞、造血幹細胞などの培養が可能になつ たのは,フィーダ一層の活用に負うところが大きい。従来は、フィーダ一細胞として、 マウス繊維芽細胞 (初代培養細胞または細胞株)が使用されており、幹細胞榭立効 率は 10%程度であるとされている。本明細書において、このようなフィーダ一細胞を 含む調製物は、「フィーダ一細胞調製物」という。また、本発明において、二種以上の 細胞を混合した細胞であって、フィーダ一細胞として使用される場合は、「フィーダ一 細胞混合物」または「細胞混合物」 t 、うことがある。 [0116] In the present specification, "feeder layer" or "feeder cell" (feeder layer or feeder cell) is used interchangeably and is a cell provided on a culture substrate and which cannot be maintained alone by culture. Refers to a feeder layer with other cell types that allows the growth of the species and the expression of the Z or sliver trait. It is said that there are many types of tissue cells that cannot express the shunting phenotype or even proliferate under normal cell culture conditions. Such cells include, for example, stem cells (especially, ES cells, neural cells). Stem cells, mesenchymal stem cells, hematopoietic stem cells, etc.), embryonic germ cells (EG cells), germ stem cells (GS cells), and the like, but are not limited thereto. These cell types generally require specific auxotrophic growth and differentiation inducing factors. By utilizing a specific cell layer formed by supporting cells of the cell type or cells similar to the cell type in the living body as a culture substrate, the factors required by the cell type to be cultured and Z Or, by being supplied with a nutrient source, they grow and divide. The cell type to be used as a feeder cell is selected depending on the target cell type, but it is often used by suppressing cell proliferation by a method such as UV irradiation. The ability to culture germ cells, ES cells, hematopoietic stem cells, etc., is largely dependent on the further utilization of feeders. Conventionally, as a feeder cell, Mouse fibroblasts (primary cultured cells or cell lines) are used, and the efficiency of stem cell establishment is said to be about 10%. In the present specification, a preparation containing such a feeder-cell is referred to as a “feeder-cell preparation”. Further, in the present invention, when a cell is a mixture of two or more cells and is used as a single feeder cell, it may be referred to as “a single feeder cell mixture” or “a single cell mixture”.
[0117] 本明細書において「STO細胞」とは、榭立されたマウス繊維芽細胞株であって、 A TCCにおいて CRL— 1503として寄託されている細胞を含む。 STO細胞は、 SIM繊 維芽細胞株に由来し、 6-チォグァニンおよびゥァバインに対して抵抗性を示し、 HG PRT— (HPRT—)であり、 HATに感受性を有するとされる。従来より、 STO細胞は、 フィーダ一細胞として用いられており、その対象には、 ES細胞のほ力、テラトカルシノ 一マ、ハイプリドーマなどが含まれている。 STO細胞の維持には、適切である限り任 意の培養条件を用いることができる力 例えば、 DMEMに 4mM L—グルタミンを加 え、 1. 5gZL重炭酸ナトリウムおよび 4. 5gZLグルコース、およびゥシ胎仔血清 (F CS) 10%をカ卩えたものを用いて培養することができる。 STO細胞の継代には、培地 を取り除き、 0. 25%トリプシン、 0. 03% EDTA溶液を用いてリンスし、その溶液を 除いて、さらに 1— 2mlのトリプシン- EDTA溶液をカ卩える。その後、容器を室温 (また は 37°C)中に移動させて細胞を剥離させる。その後新鮮な培地を入れて、吸引し、そ の後新たな培養容器へと分配して継代を完成する。培養容器は、ゼラチンコーティン グされていてもよい。継代は、 1 : 3— 1 : 10で行うことが推奨されるがそれに限定され ない。 STO細胞としては、上記 ATCCに寄託されたものを用いてもよいし、あるいは 、照射された STO細胞(例えば、 ATCCでは、 56— Xとして寄託されている)を用いて ちょい。  [0117] As used herein, the term "STO cell" refers to an established mouse fibroblast cell line, and includes a cell deposited as CRL-1503 in the ATCC. STO cells are derived from the SIM fibroblast cell line, are resistant to 6-thioguanine and ゥ abain, are HG PRT— (HPRT—), and are said to be sensitive to HAT. Conventionally, STO cells have been used as feeder cells, and their targets include the strength of ES cells, teratocarcinoma, and hybridomas. The ability to use any culture conditions as appropriate to maintain STO cells.For example, add 4 mM L-glutamine to DMEM, add 1.5 gZL sodium bicarbonate and 4.5 gZL glucose, and It can be cultured using 10% serum (FCS). For passage of STO cells, remove the medium, rinse with 0.25% trypsin, 0.03% EDTA solution, remove the solution, and add 1-2 ml of trypsin-EDTA solution. Then, move the container to room temperature (or 37 ° C) to detach the cells. Then fill with fresh medium, aspirate, and then dispense into new culture vessels to complete the passage. The culture vessel may be coated with gelatin. It is recommended, but not limited to, passage at 1: 3—1: 10. As the STO cells, those deposited with the ATCC may be used, or irradiated STO cells (eg, deposited with ATCC as 56-X) may be used.
[0118] 本明細書において「SL10細胞」とは、 STO細胞株力 サブ樹立した細胞株であり 、その榭立方法は、 Kawase E. , et al. , Int. J. Dev. Biol. 38 : 385—390 (19 94)、フィーダ一細胞の調製などについては、川瀬らの方法 [川瀬ら、実験医学, Vol . 10. No. 13 (増刊), 1575-1580, 1992]に準じて実施できる。 Robertson E . J. (ed) Teratocarsmomas and Emryonic Stem Cells, IRL Press, Ne w York (1987)に記載されており、本明細書においてその記載を参考として援用 する。手短に述べると、 STO細胞を neo1遺伝子を含むベクターでトランスフエタトする 。その後、ネオマイシン抵抗性になったコロニーを増幅し、そしてフィーダ一細胞とし ての適切性を確認する。この場合 ES細胞株 CCEを試験用に用いることができるがそ れに限定されない。 SL10細胞は、 CCEの未分化幹細胞コロニーをよく維持するもの として選択される。 SL10細胞は、ゼラチンへの接着性の良さからフィーダ一として適 しているとされる。 [0118] In the present specification, "SL10 cell" is a sub-established cell line of STO cell line, and its establishment method is described in Kawase E., et al., Int. J. Dev. Biol. 38: 385-390 (1994), preparation of feeder cells can be performed according to the method of Kawase et al. [Kawase et al., Experimental Medicine, Vol. 10. No. 13 (extra), 1575-1580, 1992]. . Robertson E. J. (ed) Teratocarsmomas and Emryonic Stem Cells, IRL Press, New York (1987), which is incorporated herein by reference. To do. Briefly, transfected Hue Tato the STO cells with the vector containing the neo 1 gene. Thereafter, the colonies that have become neomycin resistant are amplified, and their suitability as feeder cells is confirmed. In this case, the ES cell line CCE can be used for the test, but is not limited thereto. SL10 cells are selected for good maintenance of undifferentiated stem cell colonies of CCE. SL10 cells are said to be suitable as a feeder because of their good adhesion to gelatin.
[0119] 本明細書において「初代培養細胞」とは、体から分離した細胞、組織、器官などを 植え込み,第 1回目の継代を行うまでの培養の状態にある細胞をいうが、本明細書で は、継代を 5回程度まで行うものは本明細書において初代培養細胞の定義内に入る ことが理解される。初代培養細胞は、通常何も改変を施されていない細胞を用いるこ とが好ま 、が、マイトマイシン Cまたは X線で不活ィ匕したものを用いることもできる。  [0119] As used herein, the term "primary cultured cell" refers to a cell that has been cultured until the first passage, in which cells, tissues, organs, and the like separated from the body are implanted. It is understood in this document that passages performed up to about 5 times fall within the definition of primary cultured cells herein. As the primary culture cells, it is usually preferable to use cells without any modification, but it is also possible to use cells inactivated with mitomycin C or X-rays.
[0120] 本明細書にぉ ヽて継代された後の細胞は、「継代細胞」とも 、う。 [0120] The cells that have been passaged herein are also referred to as "passaged cells".
[0121] 本発明の細胞は、細胞の維持または所望の分化細胞へ分化する限り、任意の培養 液を用いることができる。そのような培養液としては、例えば、 DMEM、 M2、 M16、 P199、 MEM、 HBSS、 Ham' s F12、 BME、 RPMI1640、 MCDB104、 MCDB 153 (KGM)、 M199、 MEM a、 DMEM/F12= 1: 1、 L— 15などが挙げられるが それらに限定されない。このような培養液には、デキサメタゾンなどの副腎皮質ステロ イド、インスリン、グルコース、インドメタシン、イソブチルーメチルキサンチン(IBMX)、 ァスコルべ一トー 2—ホスフェート、ァスコルビン酸およびその誘導体、グリセ口ホスフエ ート、エストロゲンおよびその誘導体、プロゲステロンおよびその誘導体、アンドロゲン およびその誘導体、 aFGF、 bFGF、 EGF、 IGF、 TGF jS、 ECGF、 BMP, PDGFな どの増殖因子、下垂体エキス、松果体エキス、レチノイン酸、ビタミン D、甲状腺ホル モン、子牛血清、馬血清、ヒト血清、へパリン、炭酸水素ナトリウム、 HEPES、アルブ ミン、トランスフェリン、セレン酸(亜セレン酸ナトリウムなど)、リノレン酸、 3—イソブチル —1ーメチルキサンチン、 5—ァザンシチジンなどの脱メチル化剤、トリコスタチンなどの ヒストン脱ァセチル化剤、ァクチビン、 '1しー2 '1し—6などのサィトカィン、へキサメ チレンビスァセトアミド(HMBA)、ジメチルァセトアミド(DMA)、ジブチル cAMP (db cAMP)、ジメチルスルホキシド(DMSO)、ョードデォキシゥリジン(IdU)、ヒドロキシ ゥレア(HU)、シトシンァラビノシド (AraC)、マイトマイシン C (MMC)、酪酸ナトリウム (NaBu)、ポリブレン、セレニウムなどを 1つまたはその組み合わせとして含ませてお いてもよい。 [0121] As the cells of the present invention, any culture solution can be used as long as the cells are maintained or differentiated into desired differentiated cells. As such a culture solution, for example, DMEM, M2, M16, P199, MEM, HBSS, Ham's F12, BME, RPMI1640, MCDB104, MCDB153 (KGM), M199, MEMa, DMEM / F12 = 1: 1, L-15 and the like, but are not limited thereto. Such cultures include adrenocortical steroids such as dexamethasone, insulin, glucose, indomethacin, isobutyl-methylxanthine (IBMX), ascorbate 2-phosphate, ascorbic acid and its derivatives, glycemic phosphate, Estrogen and its derivatives, progesterone and its derivatives, androgen and its derivatives, growth factors such as aFGF, bFGF, EGF, IGF, TGF jS, ECGF, BMP, PDGF, pituitary extract, pineal extract, retinoic acid, vitamin D , Thyroid hormone, calf serum, calf serum, horse serum, human serum, heparin, sodium bicarbonate, HEPES, albumin, transferrin, selenate (such as sodium selenite), linolenic acid, 3-isobutyl-1-methylxanthine Demethylating agents such as 5-azancytidine; Histone deacetylating agents such as costatin, activin, sitin such as'1-2'-1'-6, hexamethylene bisacetamide (HMBA), dimethylacetamide (DMA), dibutyl cAMP (db cAMP) , Dimethylsulfoxide (DMSO), eododexperidine (IdU), hydroxy ゥ Rare (HU), cytosine arabinoside (AraC), mitomycin C (MMC), sodium butyrate (NaBu), polybrene, selenium, etc. may be contained as one or a combination thereof.
[0122] 本明細書にぉ 、て「分ィ匕因子」とは、「分化促進因子」とも 、、分化細胞への分ィ匕 を促進することが知られている因子 (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、 DNA脱メチル化剤(5—ァザシチジンなど)、ヒストン脱ァセ チル化剤(トリコスタチンなど)、核内レセプターリガンド (例えば、レチノイン酸 (ATR A)、ビタミン D、 T3など)、細胞増殖因子(ァクチビン、 IGF— 1、 FGF, PDGF、 TG  [0122] As used herein, the term "diversion factor" refers to both a "differentiation promoting factor" and a factor known to promote diversion to differentiated cells (for example, a chemical substance, Temperature, etc.). Such factors include, for example, various environmental factors, such as temperature, humidity, pH, salt concentration, nutrition, metals, gases, organic solvents, pressure, and chemicals. (Eg, steroids, antibiotics, etc.) and any combination thereof, but are not limited thereto. Representative of such factors are DNA demethylating agents (such as 5-azacytidine), histone deacetylating agents (such as trichostatin), nuclear receptor ligands (such as retinoic acid (ATR A), vitamin D, T3, etc.), cell growth factors (activin, IGF-1, FGF, PDGF, TG
3  Three
F- β、 BMP2/4など)、サイト力イン(LIFゝ IL 2、 IL— 6など)、へキサメチレンビス ァセトアミド、ジメチルァセトアミド、ジブチル cAMP、ジメチォルスルホキシド、ョード デォキシゥリジン、ヒドロキシル尿素、シトシンァラビノシド、マイトマイシン C、酪酸ナト リウム、ァフイディコリン、フルォロデオキシゥリジン、ポリプレン、セレンなどが挙げられ るがそれらに限定されない。  F-β, BMP2 / 4, etc.), cytotoxicity (LIF ゝ IL2, IL-6, etc.), hexamethylenebisacetamide, dimethylacetamide, dibutyl cAMP, dimethylthiol sulfoxide, iododexiridine, hydroxyl urea, Examples include, but are not limited to, cytosine arabinoside, mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polypropylene, selenium, and the like.
[0123] 具体的な分ィ匕因子としては、以下が挙げられる。  [0123] Specific examples of the diversion factor include the following.
[0124] A)神経: NGF、 CNTF, EGF、 FGF;  [0124] A) Nerve: NGF, CNTF, EGF, FGF;
B)血液細胞: VEGF、 EPO、インターロイキン類およびインターフェロン類(例えば 、インターロイキン 2など);  B) Blood cells: VEGF, EPO, interleukins and interferons (eg, interleukin 2, etc.);
C)肝臓: HGF  C) Liver: HGF
D)骨,軟骨: BMP。  D) Bone, cartilage: BMP.
[0125] 本明細書において「生体内」または「インビボ」(in vivo)とは、生体の内部をいう。  [0125] As used herein, "in vivo" or "in vivo" (in vivo) refers to the inside of a living body.
特定の文脈において、「生体内」は、目的とする組織または器官が配置されるべき位 置をいう。  In a particular context, "in vivo" refers to the location where the tissue or organ of interest is to be located.
[0126] 本明細書にぉ 、て「インビトロ」 (in vitro)とは、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いる状態をいう。 インビボと対照をなす用語である。 [0126] As used herein, the term "in vitro" refers to a state in which a part of a living body is extirpated or released (for example, in a test tube) for various research purposes. Say. A term that contrasts with in vivo.
[0127] 本明細書にぉ 、て「ェキソビボ」(ex vivo)とは、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。 [0127] As used herein, "ex vivo" (ex vivo) refers to the extraction of a target cell for gene transfer from a subject, the introduction of a therapeutic gene in vitro, and then the same subject again. When returning, a series of actions is called ex vivo.
[0128] 本明細書において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、組織、臓器など)をいう。本明細書にお いて自己というときは、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。 [0128] In this specification, the term "self" or "self" refers to an individual or a part thereof (eg, a cell, a tissue, an organ, etc.) derived from the individual. As used herein, the term self may broadly include a transplant from another genetically identical individual (eg, an identical twin).
[0129] 本明細書において同種(同種異系)とは、同種であっても遺伝的には異なる他個体 カゝら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。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。  [0129] In the present specification, the allogeneic (allogeneic) refers to an individual who is the same species but is genetically different from another individual to be transplanted or a part thereof (eg, cells, tissues, organs, etc.). Say. Due to their genetic differences, allogenes can elicit an immune response in the transplanted individual (recipient). Examples of such cells include, but are not limited to, cells derived from the parent.
[0130] 本明細書において異種とは、異種個体力 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いう。  [0130] In the present specification, the term "heterologous" refers to a substance which is transplanted into a heterologous individual. Thus, for example, where a human is the recipient, a transplant from a pig is referred to as a xenograft.
[0131] 本明細書において「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っても異なっていてもよい  [0131] As used herein, the term "recipient" (recipient) refers to an individual that receives transplanted cells and the like, and is also referred to as a "host". On the other hand, an individual that provides cells or the like to be transplanted is called a “donor” (donor). Recipient and donor may be the same or different
[0132] 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 U、が、拒絶反応が問題でな 、場合同種異系由来であってもよ 、。 [0132] The cells used in the present invention may be derived from syngeneic (autologous (autologous)), allogeneic (from another individual (heterologous)), or heterologous. Autologous cells are preferred because rejection is likely, but if rejection is not an issue, it may be allogeneic.
[0133] 本明細書において「移植」とは、本発明の細胞、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意味する。本発明は、以下のよ うな治療部位 (例えば、骨などなど)への導入方法,導入形態および導入量が使用さ れ得る:本発明の医薬などの障害部位への直接注入し、貼付後に縫合し、挿入する 等の方法があげられる。本発明の脂肪幹細胞と、分ィ匕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力 治療混合物としてとも に投与される提示を含み、そして組み合わせた薬剤が、別々であるが同時に(例え ば、分化促進因子)投与される手順もまた含む。「組み合わせ」投与または移植は、 第 1に与えられ、続いて第 2に与えられる細胞、医薬、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。 [0133] In the present specification, "transplantation" means that the cells, compositions, medicaments, and the like of the present invention are transferred into the body alone or in combination with other therapeutic agents. The present invention can use the following methods, forms and amounts of introduction into a treatment site (eg, bone, etc.): And then insert it. Combinations of the adipose stem cells of the present invention and the sorting cells may be, for example, simultaneously as a mixture, separately but simultaneously or in parallel; or sequentially. May be administered. This includes presentation in which the combined drug power is administered together as a therapeutic mixture, and also includes procedures in which the combined drugs are administered separately but simultaneously (eg, a differentiation promoting factor). "Combination" administration or transplantation further includes separately administering one of the cells, drugs, compounds or agents given first, followed by the second.
[0134] 本明細書において「維持」とは、細胞、組織または臓器について用いられるとき、そ の機能および Zまたは形態を実質的に保持させることをいう。例えば、角膜の維持と は、被検体が通常有するべき角膜の機能および Zまたは形態を実質的に損傷しな いで有することをいい、機能としては、視力の維持、形態としては、外観の保持が挙 げられるがそれに限定されない。  [0134] As used herein, "maintenance", when used for a cell, tissue or organ, refers to substantially retaining its function and Z or morphology. For example, maintaining the cornea means having the function and Z or morphology of the cornea that the subject should normally have without substantially damaging the subject. The function includes maintaining the visual acuity and the form includes maintaining the appearance. Listed, but not limited to.
[0135] 本明細書において使用される「再生」(regeneration)とは,個体の組織または臓 器の一部が失われた際に、欠如した組織が補填されて復元される現象をいう。動物 種間または同一個体における組織種に応じて、再生のその程度および様式は変動 する。ヒト組織の多くはその再生能が限られており、大きく失われると完全再生は望め ない。大きな傷害では、失われた組織とは異なる増殖力の強い組織が増殖し,不完 全に組織が再生され機能が回復できない状態で終わる不完全再生が起こり得る。こ の場合には,生体内吸収性材料力もなる構造物を用いて、組織欠損部への増殖力 の強 、組織の侵入を阻止することで本来の組織が増殖できる空間を確保し,さらに 細胞増殖因子を補充することで本来の組織の再生能力を高める再生医療が行われ ている。この例として、軟骨、骨および末梢神経の再生医療がある。あるいは、本発 明の方法で榭立された幹細胞を用いれば、どのような組織の再生も原理的には行う ことができ、そのようにして調製された本発明の臓器、組織および細胞が再生のため の移植物として提供される。細胞は、由来により、外胚葉、中胚葉および内胚葉に由 来する幹細胞に分類され得る。外胚葉由来の細胞は、主に脳に存在し、神経幹細胞 およびその分ィ匕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、膝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、細胞はどのような胚葉由来でもよい。 [0135] As used herein, "regeneration" refers to a phenomenon in which, when a part of an individual's tissue or organ is lost, the missing tissue is supplemented and restored. Depending on the species between animals or tissue in the same individual, the extent and mode of regeneration will vary. Many human tissues have a limited ability to regenerate, and complete loss cannot be expected if they are severely lost. In a major injury, incomplete regeneration can occur in which a highly proliferative tissue that is different from the lost tissue proliferates, incompletely regenerating the tissue and ending in a state in which function cannot be restored. In this case, by using a structure that also has a bioabsorbable material strength, the proliferation power to the tissue defect is prevented, and the invasion of the tissue is prevented, thereby securing a space in which the original tissue can proliferate. Regenerative medicine is being used to increase the ability of native tissues to regenerate by supplementing growth factors. An example of this is regenerative medicine for cartilage, bone and peripheral nerves. Alternatively, if the stem cells established by the method of the present invention are used, any tissue can be regenerated in principle, and the organs, tissues and cells of the present invention thus prepared can be regenerated. Provided as an implant for Cells can be classified according to their origin into ectoderm, mesoderm and endoderm-derived stem cells. The cells derived from the ectoderm mainly exist in the brain, and include neural stem cells and their cells. Mesodermal-derived cells are mainly present in bone marrow, and include vascular stem cells and their differentiated cells, hematopoietic stem cells and their differentiated cells, mesenchymal stem cells and their cells, and the like. Endoderm-derived cells are mainly present in organs, and include hepatic stem cells and their cells, knee stem cells and their cells. Honmei In the text, the cells can be from any germ layer.
[0136] 本発明にお 、て、幹細胞は遺伝子改変することができる。幹細胞は培養細胞であ るので、他の培養細胞と全く同様に種々の遺伝子導入法 (例えばリン酸カルシウム法 、リボゾーム法、マイクロインジェクション法、エレクト口ポレーシヨン法等)が利用できる 。また、導入できる遺伝子も何ら制限されるものではなぐ細菌、動物またはヒトの染 色体に由来する遺伝子などを挙げることができるがそれに限定されない。同様に、 E S細胞株を利用するジーンターゲッティングの方法として使用されるターゲッティング ベクターを用いた内在遺伝子の相同遺伝子組換えによる改変も可能である。  In the present invention, stem cells can be genetically modified. Since stem cells are cultured cells, various gene transfer methods (eg, calcium phosphate method, ribosome method, microinjection method, electoral poration method, etc.) can be used just like other cultured cells. The gene that can be introduced is not limited, and examples thereof include genes derived from bacterial, animal or human chromosomes, but are not limited thereto. Similarly, modification by homologous gene recombination of an endogenous gene using a targeting vector used as a method of gene targeting using an ES cell line is also possible.
[0137] 本発明の方法が対象とし得る疾患、障害、状態は、臓器または組織の再生が所望 される任意の疾患、障害、状態を含む。本発明は特に、フィーダ一細胞がなければ 再生し得ない幹細胞などを必要とする臓器、糸且織または細胞に関連する疾患、障害 、状態が対象として特に有利である。  [0137] The disease, disorder, or condition that can be targeted by the method of the present invention includes any disease, disorder, or condition in which regeneration of an organ or tissue is desired. The present invention is particularly advantageous for diseases, disorders and conditions related to organs, fibrils or cells requiring stem cells which cannot be regenerated without feeder cells.
[0138] そのような本発明が対象とし得る疾患および障害としては、循環器系(血液細胞な ど)の疾患または障害;神経系の疾患または障害;免疫系の疾患または障害;運動器 •骨格系の疾患または障害;皮膚系の疾患または障害;内分泌系の疾患または障害; 呼吸器系の疾患または障害;消化器系の疾患または障害;、泌尿器系の疾患または障 害;生殖器系の疾患または障害などが挙げられるがそれらに限定されない。  [0138] Such diseases and disorders that can be targeted by the present invention include: diseases or disorders of the circulatory system (such as blood cells); diseases or disorders of the nervous system; diseases or disorders of the immune system; Diseases or disorders of the skin; diseases or disorders of the skin; diseases or disorders of the endocrine system; diseases or disorders of the respiratory system; diseases or disorders of the digestive system; diseases or disorders of the urinary system; Obstacles, but are not limited to them.
[0139] 本発明が対象とする動物は、幹細胞を有する動物であれば、どのような動物 (たと 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 (たとえば、チン パンジー、力-クイサル、二ホンザル、ヒト)が用いられる。最も好ましくはヒトが対象で める。  [0139] The animal targeted by the present invention is any animal that has stem cells (for example, metal eel, scallop, cartilaginous fish, teleost, amphibian, reptile, bird, mammal, etc.). It may be. Preferably, such an animal is a mammal (e.g., monotremes, marsupials, oligodentates, dermis, winged fins, carnivores, carnivores, longnoses, snouts, even Hoofs, tube rodents, squamata, sponges, cetaceans, primates, rodents, egrets). Exemplary subjects include, but are not limited to, animals such as, for example, horses, pigs, horses, chickens, cats, dogs, and the like. More preferably, primates (eg, chimpanzees, power-quisars, diplomatic monkeys, humans) are used. Most preferably, it is intended for humans.
[0140] 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。本発明の医薬に含まれる薬学的に受容可能なキャリアと しては、当該分野において公知の任意の物質が挙げられる。 [0140] When the present invention is used as a medicament, such a medicament may further include a pharmaceutically acceptable carrier and the like. A pharmaceutically acceptable carrier contained in the medicament of the present invention; For example, any substance known in the art can be used.
[0141] そのような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、希釈剤、賦形剤および/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、本発明の細胞を、 1っ以 上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、注射用水、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な他の物質を補充 することが可能である。  [0141] Such suitable formulation materials or pharmaceutically acceptable carriers include antioxidants, preservatives, colorants, flavors, diluents, emulsifiers, suspending agents, solvents, fillers, bulking agents , Buffering agents, delivery vehicles, diluents, excipients and / or pharmaceutical adjuvants. Typically, the medicament of the invention will be administered in the form of a composition comprising the cells of the invention together with one or more physiologically acceptable carriers, excipients or diluents. For example, suitable vehicles may be water for injection, physiological solutions, or artificial cerebrospinal fluid, which may be supplemented with other materials common in compositions for parenteral delivery. is there.
[0142] 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、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、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、 a— 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、ポリビュルピロリドン);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を含む); 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 ) )などが挙げられるがそれらに限定されな 、。  [0142] As used herein, an acceptable carrier, excipient, or stabilizer is non-toxic to the recipient, and is preferably inert at the dosages and concentrations employed. Eg, phosphate, citrate or other organic acids; ascorbic acid, a-tocopherol; low molecular weight polypeptides; proteins (eg, serum albumin, gelatin or immunoglobulin); hydrophilic polymers (eg, polybutyl) Amino acids (eg, glycine, glutamine, asparagine, arginine or lysine); monosaccharides, disaccharides and other carbohydrates (including glucose, mannose, or dextrin); chelating agents (eg, EDTA); sugar alcohols (eg, EDTA); For example, mannitol or sorbitol); salt-forming counterion (Eg, sodium); and Z or non-ionic surfactants (eg, Tween, pluronic or polyethylene glycol (PEG)), and the like.
[0143] 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。  [0143] Exemplary suitable carriers include neutral buffered saline or saline mixed with serum albumin. Preferably, the product is formulated as a lyophilizate using suitable excipients (eg, sucrose). Other standard carriers, diluents and excipients may be included as desired.
[0144] 必要に応じて本発明の医薬は、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 、。  [0144] If necessary, the medicament of the present invention may contain a coloring agent, a preservative, a fragrance, a flavoring agent, a sweetener, and the like, and other agents.
[0145] 本発明の処置方法において使用される医薬の量は、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、毎日一数ケ月に 1回(例えば、 1週間に 1回一 1ヶ月に 1回)の投与が挙げられる。 1週間一 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。 [0145] The amount of the drug used in the treatment method of the present invention is determined in consideration of the purpose of use, the target disease (type, severity, etc.), the age, weight, sex, medical history, cell morphology or type of the patient, etc. Then, those skilled in the art can easily determine. The frequency of applying the treatment method of the present invention to a subject (or patient) also depends on the purpose of use, the target disease (type, severity, etc.), the age, weight, sex, medical history, and course of treatment of the patient. A person skilled in the art can easily determine this in consideration of such factors. The frequency includes, for example, administration once every few months (for example, once a week, once a month). It is preferable to administer once a week once a month while observing the progress. The amount to be administered can be determined by estimating the amount required by the site to be treated.
[0146] 本明細書において「指示書」は、本発明の医薬などを投与する方法または診断する 方法などを医師、患者など投与を行う人、診断する人 (患者本人であり得る)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手順を指 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)のような形態でも提供され得る。  [0146] In the present specification, the "instruction" refers to a method of administering or diagnosing the medicament or the like of the present invention for a physician, a patient or the like who administers or diagnoses (possibly a patient). It is described. This instruction describes a word indicating a procedure for administering the diagnostic agent, the medicine and the like of the present invention. This instruction is prepared in accordance with the format prescribed by the competent authority of the country where the present invention is implemented (for example, the Ministry of Health, Labor and Welfare in Japan and the Food and Drug Administration (FDA) in the United States) and is issued by the competent authority. The approval is clearly stated. Instructions are so-called package inserts, which are usually provided on paper media, but are not limited to, for example, electronic media (eg, homepages (websites) provided on the Internet, emails, etc.). ) Can also be provided.
[0147] 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上記疾患 (例えば、神経疾患、心臓疾 患など)に特徴的な臨床症状の消滅によって支持され得る。治療は、上記疾患 (例え ば、神経疾患、心臓疾患など)の再発により再開することができる。  [0147] Judgment of the termination of treatment by the method of the present invention may be based on the results of standard clinical tests using commercially available atseys or devices or the above-mentioned diseases (eg, neurological diseases, cardiac diseases, etc.). Can be supported by the disappearance of various clinical symptoms. Treatment can be resumed upon recurrence of the disease (eg, neurological disease, heart disease, etc.).
[0148] 本発明はまた、本発明の医薬の 1つ以上の成分 (例えば、 ES細胞のような幹細胞) 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 知力 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。  [0148] The present invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more components (eg, stem cells such as ES cells) of the medicament of the present invention. A notice in the form of a government agency regulating the manufacture, use or sale of pharmaceuticals or biological products. Or a government approval for sale.
[0149] 本発明が研究用試薬として使用される場合、本発明の幹細胞は、当局の定める倫 理規定に従って分配されることが望ましい。そのような規定としては、例えば、日本政 府が定めるものが挙げられるがそれに限定されない。以下にその規定の例を記載す る。 [0149] When the present invention is used as a research reagent, the stem cells of the present invention are desirably distributed according to ethical rules set by the authorities. Such rules include, but are not limited to, those stipulated by the Japanese government. The following is an example of the rules. The
[0150] 榭立機関の業務および義務として政府指針などに定められているもの従い、ヒト ES 細胞の榭立および使用に関する指針に従って承認された研究計画をもつ使用機関 からの要請を受けて、榭立後特性解析を行った細胞株を分配する。分配の開始の時 期としては、多分ィ匕能と正常核型の検定と確認を行った細胞株を増殖させて、細胞 株保存のために必要な数の凍結細胞サンプルが得られた時点から分配を始めること が企図される。  [0150] In accordance with the tasks and duties of the institution, as stipulated in government guidelines, etc., and in response to a request from the institution having a research plan approved in accordance with the guidelines on the establishment and use of human ES cells, Distribute cell lines that have undergone post-mortem characterization. The timing of the start of the distribution is probably from the time when the cell lines that have been tested and confirmed for normal karyotype and the number of frozen cell samples necessary for cell line preservation are obtained. It is contemplated to begin distribution.
[0151] 榭立と分配を開始する時点では、分配機関が分配に責任を持って担当する。分配 機関には細胞培養に必要なクリーンルームと機器および細胞凍結保存用液体窒素 タンクが整備されるべきである。ヒト ES細胞株の榭立チームが使用し、その人員構成 は、例えば、研究者 5名(内 2名は医師免許保持者が好ましい)力もなる。細胞の分配 を開始する時点では、さらに実験補佐員 2名程度と事務補佐員 1名程度が実務を分 担してもよい。細胞分配および研修の要請の増加に応じて、あらたに人員および施 設を増加させることが好まし 、。  [0151] At the start of establishment and distribution, the distributor is responsible for the distribution. Distributors should have clean rooms and equipment necessary for cell culture and a liquid nitrogen tank for cell cryopreservation. It is used by the human ES cell line establishment team, and its staffing can be, for example, five researchers (two of whom are preferably doctor's license holders). At the start of cell distribution, about two additional assistants for experimentation and one assistant clerk for office work may be assigned to work. It is preferable to increase the number of personnel and facilities in response to the increasing demand for cell distribution and training.
[0152] 分配の方法および条件は例えば以下のようなものを挙げることができる: ES細胞株 の維持および増殖の継続に必要な榭立機関における細胞ストックの確保が優先され るべさである。  [0152] Examples of the distribution method and conditions include the following: The priority should be given to securing a cell stock at an institution required for maintaining and growing the ES cell line.
[0153] 幹細胞の提供者がヒトである場合、通常インフォームド 'コンセントを得る必要がある  [0153] If the donor of stem cells is a human, it is usually necessary to obtain informed consent
[0154] ヒト受精胚の提供候補者については、 ES細胞研究に凍結胚を提供依頼にあたつ てのインフォームド 'コンセントによる同意が与えられるかどうかを、次のような手^!に 従って確認する。不妊治療の開始から凍結胚の廃棄決定に至る手続き、および関連 文書、その後に始まる ES細胞研究への提供に関するプロセスの例示的概要を示す [0154] For human fertilized embryo donor candidates, it is determined whether or not informed consent is given when requesting a frozen embryo for ES cell research according to the following procedure ^! Confirm. Provides an exemplary overview of the process from initiation of fertility treatment to the decision to dispose of frozen embryos, as well as related documentation, followed by a process for submission to ES cell research
[0155] (1) 不妊治療の結果作られて凍結保存されているヒト受精胚が、子宮へ移植さ れることなく廃棄させることが決定するまでは、 ES細胞研究とは全く無関係な不妊治 療プロセスとして患者と不妊治療担当医師による臨床的問題である。 [0155] (1) Until it is decided that human fertilized embryos created and cryopreserved as a result of fertility treatment should be discarded without being transferred to the uterus, fertility treatment completely unrelated to ES cell research The process is a clinical problem with the patient and the fertility physician.
[0156] (2) 廃棄させることが決定した凍結胚について、不妊治療担当医師が中立的立 場を保ちながら、 ES細胞の研究について説明を受けるかどうか提供候補者に質問 する。 [0156] (2) The fertility treatment physician is responsible for neutralizing frozen embryos that have been decided to be discarded. Ask the candidate to receive an explanation of ES cell research while keeping the field.
[0157] (3) 説明を受けたいとの意思を示した提供候補者に対して、榭立機関の説明者( 榭立責任者以外)が ES細胞とはなにか、将来の医療への応用の可能性、研究内容 の概要、 ES細胞株榭立によって提供者は利益も不利益も受けないこと、提供者のプ ライパシーは保護されること、などについて十分に説明する。これらの説明は、全く中 立の立場で行われる。  [3] (3) For the donors who indicated their willingness to receive explanations, the explainer of the institution (other than the chief of establishment) asked the ES cell what is ES cell and how to apply it to future medical treatment. A thorough explanation of the possibilities, an overview of the research, that the establishment of the ES cell line does not benefit or detriment the donor, and that the privacy of the donor will be protected, will be fully explained. These explanations are given in a completely neutral way.
[0158] (4) 提供候補者は説明を受けた後に、提供医療機関の長に対して提供に同意す るかどうかを回答する。同意はインフォームド 'コンセントの書類への署名を必要とし、 その書類は提供医療機関が厳重に保管する。特定の提供候補者による同意あるい は不同意に関する結果は榭立関係者には伝えない。  [0158] (4) After receiving the explanation, the providing candidate responds to the head of the providing medical institution whether or not he / she agrees with the providing. Consent requires the signing of informed 'consent' documents, which will be kept securely by the provider. The results of consent or disagreement by a particular candidate will not be communicated to the relevant parties.
[0159] (5) 提供候補者による同意の署名力も一ヶ月間以上の猶予期間をおいて、提供 候補者の意思に変更がない場合は、凍結胚を榭立機関に移送する。その際、複数 の提供者力もの凍結胚を同時に引渡すとともに、凍結胚容器力 は提供者を同定で きるラベルなどを完全に除去しておく。従って、榭立機関の説明担当者が複数の提 供候補者と面会はするが、その氏名などの個人情報は知ることがなぐまたそれらの 候補のうち誰が同意して提供者となった力も知ることがないので、提供者の匿名性は 保証され得る。さらに、複数の提供者からの凍結胚を使ってその一部のみ力 細胞 株が樹立されるので、どの提供者の胚から実際に ES細胞株が樹立されたかは榭立 機関と提供医療機関の両者ともに知ることができな 、ようにする。  [5] (5) The signing ability of the consent of the donor is also given a grace period of at least one month, and if there is no change in the intention of the donor, the frozen embryo is transferred to the institution. At that time, multiple frozen embryos from the donor are delivered at the same time, and the label for identifying the donor is completely removed from the frozen embryo container. Therefore, the instructor of the institution will meet with multiple donors, but will not know the personal information such as their names, nor will they know who has agreed to become a donor. Since there is no such thing, the anonymity of the provider can be guaranteed. In addition, since only a part of the embryonic cell lines are established using frozen embryos from multiple donors, it is important to determine which donor's embryos actually established the ES cell lines between Do not know both.
[0160] 本明細書において「キット」とは、通常 2つ以上の区画に分けて、提供されるべき部 分 (例えば、試薬、粒子など)が提供されるユニットをいう。混合されて提供されるべき でなぐ使用直前に混合して使用することが好ましいような組成物の提供を目的とす るときに、このキットの形態は好ましい。そのようなキットは、好ましくは、提供される部 分 (例えば、試薬、粒子など)をどのように処理すべき力を記載する説明書を備えて いることが有利である。このような説明書は、どのような媒体であってもよぐ例えば、 そのような媒体としては、紙媒体、伝送媒体、記録媒体などが挙げられるがそれらに 限定されない。伝送媒体としては、例えば、インターネット、イントラネット、エタストラネ ット、 LANなどが挙げられるがそれらに限定されない。記録媒体としては、 CD— RO M、 CD-R,フレキシブルディスク、 DVD— ROM、 MD、ミニディスク、 MO、メモリー スティックなどが挙げられるがそれらに限定されない。 [0160] In the present specification, the "kit" refers to a unit to which a part to be provided (eg, a reagent, a particle, or the like) is provided, usually divided into two or more sections. This kit form is preferred when it is intended to provide a composition such that it should preferably be mixed and used shortly before use, which should be provided as a mixture. Advantageously, such kits will preferably include instructions describing how to process the provided components (eg, reagents, particles, etc.). Such instructions may be in any medium. Examples of such medium include, but are not limited to, a paper medium, a transmission medium, and a recording medium. Transmission media include, for example, the Internet, intranets, , LAN, etc., but are not limited to them. Examples of the recording medium include, but are not limited to, CD-ROM, CD-R, flexible disk, DVD-ROM, MD, mini disk, MO, and memory stick.
[0161] (トランスジエニック生物)  [0161] (Transgenic organisms)
本発明の幹細胞は、トランスジエニックマウスを作製するのに使用することができる。 トランスジェニックマウスを作製するための一般的な技術は、国際公開 WO01Z1315 0 (Ludwig Inst. Cancer Res. )に記載されている。米国特許第 4, 873, 191号 ( Wagner et al . )は、哺乳動物接合体への DNAのマイクロインジェクションによつ て得られた、外因性 DNAを有する哺乳動物を教示しており、これらの説明内容を本 明細書において援用する。。  The stem cells of the present invention can be used to generate transgenic mice. General techniques for producing transgenic mice are described in International Publication WO 01Z13150 (Ludwig Inst. Cancer Res.). U.S. Pat.No. 4,873,191 (Wagner et al.) Teaches mammals having exogenous DNA obtained by microinjection of DNA into mammalian zygotes; The description is incorporated herein. .
[0162] このほかにもまた、トランスジエニック生物を作り出すための様々な方法は、例えば、 M. Markkulaら、 Rev. Reprod. , 1, 97—106 (1996); R. T. Wallら J. Dairy S ci. , 80, 2213-2224 (1997); J. C. Dalton、ら、 Adv. Exp. Med. Biol. , 411 , 419-428 (1997);ぉよび H. Lubonら、 Transfus. Med. Rev. , 10, 131—143 (1996)などが挙げられるがそれらに限定されない。これらの文献の各々は、本明細 書において参考として援用される。  [0162] In addition, various methods for producing transgenic organisms are described, for example, in M. Markkula et al., Rev. Reprod., 1, 97-106 (1996); RT Wall et al., J. Dairy S. Ci., 80, 2213-2224 (1997); JC Dalton, et al., Adv. Exp. Med. Biol., 411, 419-428 (1997); Pu and H. Lubon et al., Transfus. Med. Rev., 10 , 131-143 (1996), but are not limited thereto. Each of these documents is incorporated herein by reference.
[0163] そのような中、最近 10年間ほどで、遺伝子機能の解析を目的として、胚性幹 (ES) 細胞の相同組換えを介したトランスジエニック(ノックアウト、ノックインを含む)動物の 解析が重要な手段となってきて 、る。  [0163] Under such circumstances, in the last decade or so, analysis of transgenic (including knockout and knockin) animals through homologous recombination of embryonic stem (ES) cells has been conducted for the purpose of analyzing gene function. It has become an important tool.
[0164] 高等生物では、例えば、ネオマイシン耐性遺伝子を用いる陽性選択および HSVの チミジンキナーゼ遺伝子またはジフテリア毒素遺伝子を用いる陰性選択により組換え 体の効率的な選別が行われて 、る。 PCRまたはサザンプロット法により相同組換え 体の選択が行われる。すなわち、標的遺伝子の一部を陽性選択用のネオマイシン耐 性遺伝子等で置換し、その末端に陰性選択用の HSVTK遺伝子等を連結したター ゲティングベクターを作成し、エレクト口ポレーシヨンにより ES細胞に導入し、 G418お よびガンシクロビルの存在下で選択して、生じたコロニーを単離し、さらに PCRまたは サザンブロットにより相同組換え体を選択する。  [0164] In higher organisms, efficient selection of recombinants is performed by, for example, positive selection using a neomycin resistance gene and negative selection using the thymidine kinase gene or diphtheria toxin gene of HSV. Homologous recombinants are selected by PCR or Southern blotting. In other words, a part of the target gene is replaced with a neomycin resistance gene for positive selection, etc., and a targeting vector having the HSVTK gene, etc. for negative selection ligated to its end, and introduced into ES cells by electoporation. , G418 and ganciclovir, and the resulting colonies are isolated and homologous recombinants are selected by PCR or Southern blot.
[0165] このように、内在する標的遺伝子を置換または破壊して、機能が喪失した力または 変更された変異を有するトランスジエニック (標的遺伝子組換え)マウスを作製する方 法は、標的とした遺伝子だけに変異が導入されるので、その遺伝子機能の解析に有 用である。 [0165] As described above, the force or the loss of function by replacing or destroying the endogenous target gene The method of producing a transgenic (target transgenic) mouse having an altered mutation is useful for analyzing the function of the gene because the mutation is introduced only into the targeted gene.
[0166] 所望の相同組換え体を選択した後、得られた組換え ES細胞を胚盤注入法または 集合キメラ法により正常な胚と混合して ES細胞と宿主胚とのキメラマウスを作製する。 胚盤注入法では、 ES細胞を胚盤胞にガラスピペットで注入する。集合キメラ法では、 ES細胞の塊と透明帯を除去した 8細胞期の胚とを接着させる。 ES細胞を導入した胚 盤胞を偽妊娠させた代理母の子宮に移植してキメラマウスを得る。 ES細胞は、全能 性を有するので、生体内では、生殖細胞を含め、あらゆる種類の細胞に分ィ匕すること ができる。 ES細胞由来の生殖細胞を有するキメラマウスと正常マウスを交配させると ES細胞の染色体をへテロに有するマウスが得られ、このマウス同士を交配すると ES 細胞の改変染色体をホモに有するトランスジヱニックマウスが得られる。得られたキメ ラマウスから改変染色体をホモに有するトランスジエニックマウスを得るには、雄性キメ ラマウスと雌性野生型マウスとを交配して、 F1世代のへテロ接合体マウスを産出させ 、生まれた雄性および雌性のへテロ接合体マウスを交配して、 F2世代のホモ接合体 マウスを選択する。 F1および F2の各世代において所望の遺伝子変異が導入されて いるか否かは、組換え ES細胞のアツセィと同様に、サザンブロッテイング、 PCR、塩 基配列の解読など当該分野において慣用される方法を用いて分析され得る。  [0166] After selecting a desired homologous recombinant, the obtained recombinant ES cells are mixed with normal embryos by a scutellum injection method or an assembly chimera method to produce a chimeric mouse of ES cells and a host embryo. . In blastocyst injection, ES cells are injected into blastocysts with a glass pipette. In the assembly chimera method, a mass of ES cells is adhered to an 8-cell stage embryo from which the zona pellucida has been removed. The blastocyst into which the ES cells have been introduced is transplanted into the uterus of a surrogate mother who has been pseudopregnant to obtain a chimeric mouse. Since ES cells have totipotency, they can be divided into all kinds of cells including germ cells in vivo. When a chimeric mouse having germ cells derived from ES cells is crossed with a normal mouse, a mouse having a heterologous ES cell chromosome is obtained. A mouse is obtained. To obtain transgenic mice homozygous for the modified chromosome from the resulting chimeric mice, male chimeric mice and female wild-type mice are bred to produce F1 generation heterozygous mice. And breeding female heterozygous mice to select F2 generation homozygous mice. Whether or not the desired gene mutation has been introduced into each generation of F1 and F2 can be determined by methods commonly used in the art, such as Southern blotting, PCR, and decoding of the base sequence, as in the case of recombinant ES cells. Can be analyzed using
[0167] 多様な遺伝子機能を選択的に解析することができないという問題を克服する次世 代技術として、 Creレコンビナーゼの細胞種特異的発現と Cre— ΙοχΡの部位特異的 組み換えを併用する技術が注目されて 、る。 Cre— ΙοχΡを用いるトランスジエニックマ ウスは、標的遺伝子の発現を阻害しな ヽ位置にネオマイシン耐性遺伝子を導入し、 後に削除するェキソンをはさむようにして ΙοχΡ配列を挿入したターゲテイングべクタ 一を ES細胞に導入し、その後相同組換え体を単離する。この単離したクローンから キメラマウスを得、遺伝子改変マウスが作製される。次に、大腸菌の P1ファージ由来 の部位特異的組換え酵素 Creを組織特異的に発現するトランスジエニックマウスとこ のマウスを交配させると、 Creを発現する組織中でのみ遺伝子が破壊される(ここでは 、 Creは、 ΙοχΡ配列(34bp)を特異的に認識して、 2つの ΙοχΡ配列にはさまれた配列 で組換えを起こさせ、これが破壊される)。臓器特異的なプロモータに連結した Cre遺 伝子を有するトランスジヱニックマウスと交配させる力、または Cre遺伝子を有するウイ ルスベクターを使用して、成体で Creを発現させることができる。 [0167] As a next-generation technology that overcomes the inability to selectively analyze various gene functions, a technology that combines cell type-specific expression of Cre recombinase and site-specific recombination of Cre- CreοχΡ has attracted attention. Being done. A transgenic mouse using Cre-ΙοχΡ inserts a neomycin resistance gene at a position that does not inhibit the expression of the target gene, and inserts a targeting vector that inserts the す る οχΡ sequence with an exon to be deleted later. The cells are introduced, and then the homologous recombinant is isolated. A chimeric mouse is obtained from the isolated clone, and a genetically modified mouse is produced. Next, when this mouse is crossed with a transgenic mouse that expresses Cre, a site-specific recombination enzyme derived from the P1 phage of Escherichia coli, the gene is disrupted only in the tissue that expresses Cre. In the Cre, the ΙοχΡ sequence (34bp) is specifically recognized, and the sequence sandwiched between two ΙοχΡ sequences To cause recombination, which is destroyed). Cre can be expressed in adults using the ability to breed with a transgenic mouse having a Cre gene linked to an organ-specific promoter, or a virus vector having a Cre gene.
[0168] 特定の遺伝子を解析する方法としてジーントラップ (遺伝子トラップ)法が注目され ている。ジーントラップ法では、プロモータを有しないレポーター遺伝子が細胞に導 入され、その遺伝子が偶発的にゲノム上に挿入されると、レポーター遺伝子が発現 することを利用して、新規な遺伝子を単離 (トラップ)される。ジーントラップ法は、マウ ス初期胚操作法, ES細胞培養法,相同組換えによる遺伝子ターゲテイング法に基づ ぐ効率的な挿入変異と未知遺伝子同定のための方法である(Stanford WL. , et al. , Nature Genetics 2 : 756— 768 (2001) )。ジーントラップ法では、遺伝子 の導入ならびに挿入変異体の選択およびその表現型解析が比較的に容易である。  [0168] As a method for analyzing a specific gene, a gene trap (gene trap) method has attracted attention. In the gene trap method, a reporter gene without a promoter is introduced into a cell, and when that gene is accidentally inserted into the genome, the novel gene is isolated by utilizing the expression of the reporter gene ( Trap). The gene trap method is a method for efficient insertion mutation and identification of unknown genes based on mouse early embryo manipulation, ES cell culture, and gene targeting by homologous recombination (Stanford WL., Et al.). al., Nature Genetics 2: 756—768 (2001)). In the gene trap method, introduction of a gene, selection of an inserted mutant, and phenotyping thereof are relatively easy.
[0169] ジーントラップ法では、例えば、スプライシング Zァクセプター配列とポリ A付加シグ ナルとの間に lacZと neoとの融合遺伝子である β geoを連結したジーントラップべク ターを ES細胞に導入し、 G418で選択すると、 ES細胞で発現している遺伝子を偶然 にトラップしたクローンだけが選択される。  [0169] In the gene trap method, for example, a gene trap vector in which β geo, a fusion gene of lacZ and neo, is linked between a splicing Z receptor sequence and a poly-A-added signal is introduced into ES cells. When selecting with G418, only clones that accidentally trap the gene expressed in ES cells are selected.
[0170] このようにして得られたクローン力 キメラ胚を作製すると、トラップした遺伝子の発 現パターンにより、さまざまな X— galの染色パターンを示す。このようにして、ジーント ラップ法では、未知の遺伝子が単離され、その遺伝子発現パターンが解析され、また その遺伝子が破壊される。  When the clonal chimeric embryo thus obtained is produced, various X-gal staining patterns are exhibited depending on the expression pattern of the trapped gene. Thus, in the gene trap method, an unknown gene is isolated, its gene expression pattern is analyzed, and the gene is destroyed.
[0171] 本発明は、このように、種々のトランスジエニック生物の作製に利用することができる  [0171] As described above, the present invention can be used for production of various transgenic organisms.
[0172] (一般技術) [0172] (General technology)
本明細書において用いられる分子生物学的手法、生化学的手法、微生物学的手 法は、当該分野において周知であり慣用されるものであり、例えば、 Sambrook J. et al. (1989). Molecularし loning: A Laboratory Manual, Cold pring Harborおよびその 3rd Ed. (2001); Ausubel, F. M. (1987). Current Protocolsin Molecular Biology, Greene Pub. AssociatES and Wiley- Interscience; Ausubel, F.M. (1989). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates and Wiley— Interscience;Innis, M. A. (1990). PCR Protocols: A Guide to Methods and Applications , Academic Press; Ausubel, F. M. (1992). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates; Ausubel, F. M. (1995). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates; Innis, M. A. et al. (1995). PCR Strategies, Academic Press; Ausubel, F. M. (1999). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Wiley, and annual updates;Sninsky, J. J. et al. The molecular biological, biochemical, and microbiological techniques used herein are well known and commonly used in the art, and are described, for example, in Sambrook J. et al. (1989). Loning: A Laboratory Manual, Cold pring Harbor and its 3rd Ed. (2001); Ausubel, FM (1987) .Current Protocols in Molecular Biology, Greene Pub. AssociatES and Wiley- Interscience; Ausubel, FM (1989). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates and Wiley—Interscience; Innis, MA (1990) .PCR Protocols: A Guide to Methods and Applications, Academic Press; Ausubel, FM (1992) .Short Protocols in Molecular Biology: A Compendium of Methods. from Current Protocols in Molecular Biology, Greene Pub. Associates; Ausubel, FM (1995) .Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates; Innis, MA et al. (1995) PCR Strategies, Academic Press; Ausubel, FM (1999) .Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Wiley, and annual updates; Sninsky, JJ et al.
(1999). PCR Applications: Protocols for FunctionalGenomics , Academic Press ¾'J冊 実験医学「遺伝子導入 &発現解析実験法」羊土社、 1997などに記載されており、こ れらは本明細書において関連する部分 (全部であり得る)が参考として援用される。  (1999) .PCR Applications: Protocols for Functional Genomics, Academic Press ¾'J, Experimental Medicine “Experimental Methods for Gene Transfer and Expression Analysis,” Yodosha, 1997, and the like. Portions (which can be all) are incorporated by reference.
[0173] 人工的に合成した遺伝子を作製するための DNA合成技術および核酸化学につい ては、 f列えば、 Gait, M. J. (1985). Oligonucleotide Synthesis: A Practical  [0173] For DNA synthesis technology and nucleic acid chemistry for producing artificially synthesized genes, see Gait, M. J. (1985). Oligonucleotide Synthesis: A Practical.
Approach, IRL Press; Gait, M. J. (1990). Oligonucleotide Synthesis: A Practical Approach, IRL Press; Eckstein, F. (1991). Oligonucleotides and Analogues: A PracticalApproach, IRL Press; Adams, R. L. et al. (1992). The Biochemistry of the Nucleic Acids, Chapman & Hall; Shabarova, Z. et al. (1994). AdvancedOrganic Chemistry of Nucleic Acids, Weinheim; Blackburn, G. M. et al. (1996). Nucleic Acids in Chemistry and Biology, Oxford University Press; Hermanson, G.T. (1996).  Oligonucleotide Synthesis: A Practical Approach, IRL Press; Eckstein, F. (1991) .Oliigonucleotides and Analogues: A PracticalApproach, IRL Press; Adams, RL et al. (1992). The Biochemistry of the Nucleic Acids, Chapman &Hall; Shabarova, Z. et al. (1994) .AdvancedOrganic Chemistry of Nucleic Acids, Weinheim; Blackburn, GM et al. (1996) .Nucleic Acids in Chemistry and Biology, Oxford University Press. ; Hermanson, GT (1996).
Bioconjugate Techniques, Academic Pressなどに記載されており、これらは本明細書 において関連する部分が参考として援用される。  Bioconjugate Techniques, Academic Press, etc., and the relevant portions are incorporated herein by reference.
[0174] (好ましい実施形態の説明)  (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ことが理解される。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 it is understood that the scope of the present invention should not be limited to the following description. Therefore, it is apparent that those skilled in the art can appropriately make modifications within the scope of the present invention in view of the description in the present specification.
[0175] (フィーダ一細胞調製物) 1つの局面において、本発明は、正常細胞と、細胞株とを含む、幹細胞を調製する ためのフィーダ一細胞調製物を提供する。このような複数の性質を有するフィーダ一 細胞の混合物をフィーダ一細胞調製物として利用した例はこれまでなぐまた、従来 は、フィーダ一細胞として使用可能な細胞種はいくつか知られていた力 それらの間 に本質的な差違の存在は想定されておらず、従って細胞を組み合わせることによつ てフィーダ一細胞機能が著しく向上することは予想されて 、な力つた。本発明にある ような細胞の組み合わせにより幹細胞株の榭立維持の効率が非常に向上する機作 につ!/、てはフィーダ一細胞の機能が分子レベルで解明されて!、な!/、ため明らかでな いが、上に挙げたような因子の供給が単独の細胞の使用では完全に行われない為 では無 、かと思われる。よってフィーダ一細胞を複数種組み合わせることにより予想 外の効果が得られたと考えられる。従って、ある局面では、本発明は、任意の複数の フィーダ一細胞の混合物を用いることができることが理解される。本明細書において 使用されるフィーダ一細胞は、従来フィーダ一細胞として使用される細胞であれば、 どのような細胞であっても使用され得ることが理解される。 [0175] (Feeder-one cell preparation) In one aspect, the present invention provides a feeder-one cell preparation for preparing a stem cell, including a normal cell and a cell line. There have been no examples in which a mixture of feeder cells having such a plurality of properties has been used as a feeder cell preparation.Furthermore, conventionally, several cell types that can be used as feeder cells have been known. No substantial difference was expected between the two, and it was anticipated that combining cells would significantly improve feeder-cell function. The mechanism by which the combination of cells as in the present invention greatly improves the efficiency of stem cell line establishment and maintenance! /, And the functions of feeder cells are elucidated at the molecular level! Therefore, it is not clear that the above-mentioned factors are not completely supplied by using a single cell. Therefore, it is considered that an unexpected effect was obtained by combining a plurality of feeder cells. Thus, it is understood that, in one aspect, the invention can employ a mixture of any plurality of feeder-cells. It is understood that the feeder cell used in the present specification can be any cell that is conventionally used as a feeder cell.
[0176] 本明細書において使用される正常細胞は、フィーダ一細胞として適切に使用され ている細胞であれば、使用され得ることが理解される。そのような正常細胞としては、 例えば、マウス初代培養繊維芽細胞、ラット初代培養繊維芽細胞、サル初代培養繊 維芽細胞、ヒト初代培養繊維芽細胞などを挙げることができるがそれらに限定されな い。  [0176] It is understood that the normal cell used in the present specification can be used as long as the cell is appropriately used as a feeder cell. Examples of such normal cells include, but are not limited to, mouse primary cultured fibroblasts, rat primary cultured fibroblasts, monkey primary cultured fibroblasts, and human primary cultured fibroblasts. No.
[0177] そのような初代培養繊維芽細胞の調製方法としては、例えば、 11 16日令胎児か ら、頭部、内臓等を取り除いたものを細断、酵素処理で主として繊維芽細胞を取り出 すと 、う方法を用いて行うことができる。  [0177] As a method for preparing such primary cultured fibroblasts, for example, a fetus from which a head, internal organs, and the like are removed from an 11-16-day-old fetus is shredded, and the fibroblast is mainly extracted by enzymatic treatment. Then, it can be performed by using a method.
[0178] 具体的には、  [0178] Specifically,
1.受精後 12日目(11一 16日目が好ましい。)のマウスを準備する。  1. Prepare a mouse on day 12 (preferably day 11 to 16) after fertilization.
2.胎児を摘出し胎仔を傷つけないように羊膜'胎盤を取り除き、新しいペトリディッシ ュへ移す o  2. Remove the fetus, remove the amniotic membrane's placenta without damaging the fetus, and transfer to a new Petri dish o
3.実体顕微鏡下で胎仔の頭部および腹部の肝臓、腸などの内臓をできるだけピン セットで取り除く。 PBS (—)で胎仔についた余分な臓器を洗い流し、新しいペトリディ 、移す。 3. Remove the internal organs such as the liver and intestine of the fetal head and abdomen with tweezers as much as possible under a stereoscopic microscope. Rinse excess organs from the fetus with PBS (-), ,Move.
4.クリーンベンチ内で 100 mm細胞培養ディッシュに培地を 10 mlずつ分注する 。 21G針とシリンジを準備する。シリンジの中に胎仔を 1つ入れ、ディッシュ中の培地 をゆっくりと吸い上げ、そのまま同じディッシュ中へゆっくりと培地をもどす。この操作 を 2— 3回繰り返す。  4. Dispense 10 ml of medium into 100 mm cell culture dishes in a clean bench. Prepare a 21G needle and syringe. Put one fetus in a syringe, slowly suck up the medium in the dish, and slowly return the medium to the same dish. Repeat this operation 2-3 times.
5.胎仔が細片になっていることを確認する。  5. Make sure that the fetus is in pieces.
6. 37°C、 COインキュベータでー晚培養する。  6. Incubate at 37 ° C in a CO incubator.
2  2
t 、う方法によって、繊維芽細胞を調製することができる。  t, Fibroblasts can be prepared according to the method.
[0179] 好ましくは、上記正常細胞は不死化していないことが有利である。理論に束縛され ることは望まないが、不死化していないことによって、分化させずに幹細胞を維持する ために必要な因子の一部が供給されると考えられるからである。  [0179] Preferably, the normal cells are not immortalized. While not wishing to be bound by theory, it is believed that non-immortalization provides some of the factors necessary to maintain stem cells without differentiation.
[0180] 本発明において使用される正常細胞は、繊維芽細胞であってもよいが、そうでなく てもよい。好ましくは、繊維芽細胞が使用される。理論に束縛されることを望まないが 、繊維芽細胞は、フィーダ一細胞としての適性を付与することが多ぐ分化させずに 幹細胞を維持するために必要な因子の一部が供給されると考えられる力 である。  [0180] The normal cells used in the present invention may be fibroblasts, but need not be. Preferably, fibroblasts are used. While not wishing to be bound by theory, it is important to note that fibroblasts, when given some of the factors necessary to maintain stem cells without differentiation, often do not confer suitability as feeder cells It is a conceivable force.
[0181] より好ましい実施形態では、本発明において用いられる正常細胞は、繊維芽細胞 初代培養細胞である。より好ましくは、本発明において用いられる正常細胞は、マウ ス由来である。理論に束縛されることを望まないが、マウスであることが好ましい理由 は、調製の容易さにある。従って、任意の哺乳動物が使用され、簡易な調製法が別 の存在する場合は、他の哺乳動物の細胞を用いることによって好ましく使用され得る [0181] In a more preferred embodiment, the normal cells used in the present invention are fibroblast primary culture cells. More preferably, the normal cells used in the present invention are mouse-derived. While not wishing to be bound by theory, a mouse is preferred because of its ease of preparation. Therefore, any mammal can be used, and if another simple preparation method exists, it can be preferably used by using cells of another mammal.
。理論に束縛されることは望まないが、経験的には、ゥシおよびゥサギの ES榭立研究 で同種の繊維芽細胞を使用しても、マウスより良い効果は見られないようであり、一般 的にはマウスの細胞が良 、と考えられて 、る。 . While not wishing to be bound by theory, empirical evidence suggests that the use of allogeneic fibroblasts in ES-establishment studies of Escherichia coli and Egret does not seem to be as effective as mice. Mouse cells are generally considered to be good.
[0182] 好ましい実施形態では、本発明において用いられる正常細胞は、胎児 (胎仔)由来 であり得る。理論に束縛されることを望まないが、胎児由来の細胞は、分化させずに 幹細胞を維持するために必要な因子の一部を提供し、フィーダ一細胞としてより適切 であると考えられるからである。また、本発明において使用される細胞株とは、細胞学 的にみて、最も異なる性質を有すると考えられ、従って、細胞株が提供できない幹細 胞の維持に必要な因子を供給する能力を有すると考えられるからである。 [0182] In a preferred embodiment, the normal cells used in the present invention may be derived from a fetus (fetus). While not wishing to be bound by theory, it is believed that cells from the fetus provide some of the factors necessary to maintain stem cells without differentiation and are more suitable as feeder cells. is there. In addition, the cell line used in the present invention is considered to have the most different properties from the viewpoint of cytology, and therefore, cannot be provided by a cell line. This is because it is considered to have the ability to supply factors necessary for the maintenance of cells.
[0183] 従って、本発明では、フィーダ一細胞としての能力を提供することができる限り、ど のような継代数の細胞でも用いることができるが、好ましくは、初代培養細胞について は継代数の少ないもの (4, 5回程度)が推奨され、継代数が 5回以下、 4回以下、 3回 以下、 2回以下、 1回または継代なしの細胞が用いられる。  [0183] Therefore, in the present invention, any passage number of cells can be used as long as the ability as a feeder cell can be provided, but it is preferable that primary culture cells have a low passage number. Cells (about 4 or 5 times) are recommended, and cells with 5 or less passages, 4 or less, 3 or less, 2 or less, 1 passage, or no passage are used.
[0184] ある好ましい実施形態では、本発明において使用される正常細胞は、マウス 11一 1 6日齢胎児から酵素処理により得られた繊維芽細胞である。  [0184] In a preferred embodiment, the normal cells used in the present invention are fibroblasts obtained by enzymatic treatment from a mouse 11-16 day old fetus.
[0185] 本明細書において使用される細胞株は、フィーダ一細胞としての能力を有する限り 、どのような細胞であっても使用することができることが理解される。そのような細胞株 は、代表的には、不死化している。理論に束縛されることを望まないが、不死化した 細胞を用いることによって、初代細胞株などの正常細胞では供給されないが、分ィ匕さ せずに幹細胞を維持するために必要な因子の他の一部が供給されると考えられるか らである。  [0185] It is understood that any cell line can be used as long as it has the ability as a feeder cell as used herein. Such cell lines are typically immortalized. Although not wishing to be bound by theory, the use of immortalized cells does not provide for normal cells such as primary cell lines, but does not provide other factors necessary to maintain stem cells without fragmentation. It is considered that some of this will be supplied.
[0186] 好ましくは、本発明において使用される細胞株は、繊維芽細胞株である。  [0186] Preferably, the cell line used in the present invention is a fibroblast cell line.
[0187] 本発明において使用される細胞株は、継代期間は、どのような期間であっても良い 力 好ましくは、継代された期間が 2ヶ月以下であり、より好ましくは、継代された期間 力 ヶ月以下である細胞を用いることが有利であり得る。理論に束縛されることを望ま ないが、そのような継代期間が短い細胞株を用いることによって、初期の目的の因子 を分泌する細胞を安定して供給することができ、分化させずに幹細胞を維持するた めに必要な因子の他の一部が十分に供給されると考えられるからである力 必ずしも 継代期間が短くある必要はない。 [0187] The cell line used in the present invention may have any passage period. Preferably, the passage period is 2 months or less, and more preferably, the passage period is not more than 2 months. It may be advantageous to use cells that have been for a period of less than one month. Without wishing to be bound by theory, it is possible to stably supply cells secreting the initial factor of interest by using such a cell line with a short passage time, and to obtain stem cells without differentiation. It is not necessary that the passage period be short, because some of the other factors necessary to maintain the force will be supplied adequately.
[0188] 本発明において使用される細胞株は、マウス由来であり得るが、それに限定されな い。理論に束縛されることを望まないが、マウスであることが好ましい理由は、調製の 容易さにある。従って、任意の哺乳動物が使用され、簡易な調製法が別の存在する 場合は、他の哺乳動物の細胞を用いることによって好ましく使用され得る。理論に束 縛されることは望まないが、経験的には、ゥシおよびゥサギの ES榭立研究で同種の 繊維芽細胞株を使用しても、マウスより良い効果は見られないようであり、一般的には マウスの細胞が良 、と考えられて 、る。 [0189] 好ま 、実施形態では、本発明にお 、て用いられる細胞株は、胎児 (胎仔)由来で あり得る。理論に束縛されることを望まないが、胎児由来の細胞は、分化させずに幹 細胞を維持するために必要な因子の一部を提供し、フィーダ一細胞としてより適切で あると考えられるからである。また、本発明において使用される正常細胞とは、細胞学 的にみて、最も異なる性質を有すると考えられ、従って、正常細胞が提供できない幹 細胞の維持に必要な因子を供給する能力を有すると考えられるからである。 [0188] The cell line used in the present invention may be derived from a mouse, but is not limited thereto. While not wishing to be bound by theory, a mouse is preferred because of its ease of preparation. Therefore, any mammal can be used, and if another simple preparation method exists, it can be preferably used by using cells of another mammal. While not wishing to be bound by theory, it has been empirically shown that the use of allogeneic fibroblast cell lines in ES-establishment studies in Escherichia coli and egrets does not appear to be as effective as in mice. Generally, mouse cells are considered to be good. [0189] Preferably, in embodiments, the cell line used in the present invention may be derived from a fetus (fetus). While not wishing to be bound by theory, it is believed that fetal-derived cells provide some of the factors needed to maintain stem cells without differentiation and are more suitable as feeder cells It is. The normal cells used in the present invention are considered to have the most different properties from a cytological point of view, and therefore have the ability to supply factors necessary for maintaining stem cells that normal cells cannot provide. Because it is possible.
[0190] 好ま ヽ実施形態では、使用される細胞株は、 STO株である。この細胞株はネオマ イシン耐性 (neo抵抗性)の性質を持って ヽることが好まし ヽ。最も好まし!/ヽ実施形態 では、本発明において使用される細胞株は、 SL10株である。  [0190] In a preferred embodiment, the cell line used is an STO line. Preferably, this cell line has the properties of neomycin resistance (neo resistance). Most preferred! In the ヽ embodiment, the cell line used in the present invention is the SL10 strain.
[0191] 本発明のフィーダ一細胞調製物において含まれる正常細胞 (たとえば、繊維芽細 胞初代培養細胞)と細胞株 (たとえば、繊維芽細胞株)とは、代表的に、約 1 : 10— 10 : 1で存在し得る。理論に束縛されることを望まないが、 1 : 10— 10 : 1の範囲外では、 フィーダ一細胞を複数種使用する効果がほとんど見られないが、 1: 10— 10 : 1の範 囲内では、幹細胞の維持において効果が有意に見られているからである。  [0191] Normal cells (for example, primary fibroblast cells) and cell lines (for example, fibroblast cell lines) contained in the feeder-cell preparation of the present invention are typically about 1: 10- Can exist at 10: 1. Without wishing to be bound by theory, outside the range of 1:10 to 10: 1, there is little effect of using multiple feeder cells, but within the range of 1:10 to 10: 1. This is because the effect is significantly observed in the maintenance of stem cells.
[0192] 好ま 、実施形態では、正常細胞 (たとえば、繊維芽細胞初代培養細胞)と細胞株  [0192] Preferably, in embodiments, normal cells (eg, primary fibroblast cells) and cell lines
(たとえば、繊維芽細胞株)との比率は、約 1 :4一 4 : 1であり得る。理論に束縛される ことを望まないが、複数種の細胞を含むフィーダ一細胞調製物を利用することによつ て、幹細胞の維持および多分ィ匕能の維持の効果が顕著に改善されるからである。よ り好ましい細胞比率は、正常細胞 (たとえば、繊維芽細胞初代培養細胞)と細胞株( たとえば、繊維芽細胞株)との比率が約 1: 3—約 3: 1であり得る。  (Eg, a fibroblast cell line) can be about 1: 4 to 4: 1. While not wishing to be bound by theory, the use of a feeder-one cell preparation containing multiple types of cells significantly improves the effects of maintaining stem cells and possibly maintaining dangling ability. It is. A more preferred cell ratio may be a ratio of about 1: 3 to about 3: 1 normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts).
[0193] 最も好まし!/、実施形態では、正常細胞 (たとえば、繊維芽細胞初代培養細胞)と細 胞株 (たとえば、繊維芽細胞株)との比率は、ほぼ等量である。理論に束縛されること を望まないが、ほぼ等量の複数種の細胞を用いることによって、相互に補完すべき幹 細胞を維持するための成分が互いに十分含まれることになるからである。従って、当 業者は、本明細書においてほぼ等量というとき、等量に近いがフィーダ一細胞として 最適な任意の比率を意味することを理解する。  [0193] Most preferred! In embodiments, the ratio of normal cells (eg, primary fibroblasts) to cell lines (eg, fibroblasts) is approximately equal. Without wishing to be bound by theory, it is because using approximately equal amounts of multiple types of cells results in sufficient components of each other to maintain stem cells to complement each other. Therefore, those skilled in the art understand that, when referring to substantially equivalent amounts in this specification, means any ratio that is close to equivalent amounts but is optimal as a feeder cell.
[0194] 1つの実施形態では、本発明のフィーダ一細胞調製物が目的とする細胞は、胚性 幹細胞であり、より好ましくは、ヒト胚性幹細胞である。ヒト胚性幹細胞のような細胞が 多能性を維持しながら増殖させることができ、しかも 50%未満の分化細胞混入率 (好 ましくは、 30%未満、より好ましくは 10%未満、最も好ましくは 1%未満)は、これまで の技術では達成できな力つた格別の効果であるといえる。 [0194] In one embodiment, the cell of interest in the feeder-one-cell preparation of the present invention is an embryonic stem cell, more preferably a human embryonic stem cell. Cells like human embryonic stem cells It can be grown while maintaining pluripotency and has less than 50% differentiated cell contamination (preferably less than 30%, more preferably less than 10%, and most preferably less than 1%). It can be said that this is a powerful effect that cannot be achieved with the technology described above.
[0195] (幹細胞調製法)  [0195] (Stem cell preparation method)
1つの局面において、本発明は、正常細胞 (例えば、繊維芽細胞初代培養細胞)と 、細胞株 (例えば、繊維芽細胞株)とを含むフィーダ一細胞調製物の上で、幹細胞を 培養する工程を包含する、幹細胞を調製するための方法を提供する。正常細胞 (例 えば、繊維芽細胞初代培養細胞)および細胞株 (例えば、繊維芽細胞株)は、おの おの正常細胞 (例えば、初代培養細胞)であり、株化したものである限り、任意のもの を使用することができる。フィーダ一細胞としては、これらの 2種の細胞を含んでいる 限りどのような含まれ方であっても使用することができるが、好ましくは、均一に混合し ていることが有利である。そのような混合比率としては、好ましくは、約 10:1— 1:10、 約 9:1一 1:9、約 8:1— 1:8、約 7:1— 1:7、約 6:1— 1:6、約 5:1— 1:5、約 4:1一 1 :4、約 3: 1— 1: 3、約 2: 1— 1: 2、約 1: 1 (ほぼ等量を意味する)などが挙げられる。 好ましくは、ほぼ等量混合されることが有利である。理論に束縛されないが、正常細 胞 (例えば、繊維芽細胞初代培養細胞)と、細胞株 (例えば、繊維芽細胞株)とは、相 互に相乗的な効果を有する因子 (例えば、幹細胞維持に必要なもの)を相補的に分 泌することによって、本発明の予想外の効果を達成しているものと考えられる。  In one aspect, the present invention provides a process for culturing a stem cell on a feeder-one cell preparation containing a normal cell (eg, a primary fibroblast cell) and a cell line (eg, a fibroblast cell line). There is provided a method for preparing a stem cell, comprising: Normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts) are each normal cells (eg, primary cells) and, as long as they are established, Anything can be used. As the feeder cell, any cell may be used as long as it contains these two types of cells, but it is advantageous that the cells are uniformly mixed. Such mixing ratios are preferably about 10: 1 to 1:10, about 9: 1 to 1: 9, about 8: 1 to 1: 8, about 7: 1 to 1: 7, about 6: 1: 1: 6, approx. 5: 1—1: 5, approx. 4: 1: 1-1: 4, approx. 3: 1-1—1: 3, approx. 2: 1-1—1: 2, approx. 1: 1 (almost equivalent And the like). Preferably, approximately equal amounts are mixed. Without being bound by theory, normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts) are factors that have mutually synergistic effects (eg, stem cell maintenance). It is considered that the unexpected effect of the present invention has been achieved by complementarily secreting (required).
[0196] 1つの実施形態において、本発明のフィーダ一細胞において用いられる正常細胞( 例えば、繊維芽細胞初代培養細胞)は、マウス由来であることが有利であるがそれに 限定されない。従って、本発明では、任意の動物を用いることができる。マウスは、技 術的に充分に確立した実験動物であり、初代培養細胞などの正常細胞の入手方法 もまた、当業者によく知られるところとなっているからである。  [0196] In one embodiment, the normal cells (eg, primary fibroblasts) used in the feeder cell of the present invention are advantageously derived from a mouse, but are not limited thereto. Therefore, any animal can be used in the present invention. Mice are technically well established experimental animals, and methods for obtaining normal cells such as primary cultured cells are also well known to those skilled in the art.
[0197] 好ま 、実施形態にぉ 、て、本発明にお 、て使用される正常細胞 (例えば、繊維 芽細胞初代培養細胞)は、胎児由来であることが有利であるがそれに限定されない。 従来、例えば、繊維芽細胞初代培養細胞は、フィーダ一細胞として用いられているが 、胎児由来のものが主に用いられている。胎児由来の繊維芽細胞初代培養細胞を 単独で用いた場合でも、幹細胞の榭立効率は、それほど高いものではなぐせいせ い 10— 30%程度であった。翻って、本発明のフィーダ一細胞調製物および細胞混合 物では、その榭立効率は、 50%をはるかに超え、 80%程度の場合もある。従って、こ のような高率の榭立高率は、従来技術からは想像を超えるものであったといえる。ここ で、正常細胞は、厳密な意味での初代培養細胞であることが好ましいが、継代培養 した細胞であっても、継代数が少ないものであれば、初代培養細胞とほぼ類似する 性質を有し得ることから、本発明において有利に使用することができる。そのような継 代数としては、例えば、約 5継代以下、 4継代以下、 3継代以下、 2継代以下、 1継代 などが挙げられるがそれらに限定されない。 [0197] Preferably, in the embodiments, the normal cells (eg, primary cultured fibroblast cells) used in the present invention are advantageously derived from a fetus, but are not limited thereto. Conventionally, for example, primary fibroblast cells have been used as feeder cells, but those derived from the fetus are mainly used. Even when primary cultures of fetal fibroblasts are used alone, the efficiency of stem cell establishment is not so high. About 10-30%. In turn, with the feeder-cell preparations and cell mixtures of the present invention, the establishment efficiency is well over 50%, and in some cases as high as 80%. Therefore, it can be said that such a high rate of establishment was beyond imagination from the prior art. Here, a normal cell is preferably a primary cultured cell in a strict sense, but even if it is a subcultured cell, if it has a small number of passages, it has properties similar to those of the primary cultured cell. Since it can have, it can be used advantageously in the present invention. Examples of such a passage number include, but are not limited to, about 5 passages or less, 4 passages or less, 3 passages or less, 2 passages or less, 1 passage, and the like.
[0198] 1つの好ま 、実施形態にぉ 、て、本発明のフィーダ一細胞に含まれる細胞株 (例 えば、繊維芽細胞株)は、 STO株であることが有利である。従来 STO株は、幹細胞 のフィーダ一細胞として用いられているが、幹細胞の榭立効率は、やはり 30%以下と 低率にとどまっており、本発明の細胞混合物および細胞調製物をフィーダ一細胞調 製物として使用した場合では、幹細胞の榭立効率は、 50%をはるかに超え、 80%程 度の場合もある。従って、このような高率の榭立高率は、従来技術からは想像を超え るものであったといえる。  [0198] In one preferred embodiment, the cell line (eg, fibroblast cell line) contained in the feeder cell of the present invention is advantageously an STO line. Conventionally, the STO strain has been used as a feeder cell for stem cells, but the establishment efficiency of stem cells is still as low as 30% or less, and the cell mixture and the cell preparation of the present invention can be prepared using a feeder cell. When used as a product, the efficiency of stem cell establishment is well over 50% and can be as high as 80%. Therefore, it can be said that such a high rate of establishment was beyond imagination from the prior art.
[0199] より好ましい実施形態では、 STO細胞からの ES細胞未分ィ匕状態支持能の高い細 胞の分離法を用いてより好ましい細胞株を榭立することができる。そのような方法を簡 単に説明すると、以下のようになる。  [0199] In a more preferred embodiment, a more preferable cell line can be established by using a method for separating cells having a high ability to support an ES cell in an undivided state from STO cells. The following is a brief description of such a method.
[0200] 1.定法に従い STO細胞を細胞一つ一つになるまで解離する。  [0200] 1. Dissociate STO cells into individual cells according to a standard method.
[0201] 2.培養皿に 100— 500個程度の解離した STO細胞を播種する。  [0201] 2. Seed 100 to 500 dissociated STO cells in a culture dish.
[0202] 3.数日間培養すると播種した細胞一つ一つの細胞それぞれからコロニーが形成さ れるので十分な大きさのコロニーが形成されるまで培養を続ける。  [0202] 3. After culturing for several days, a colony is formed from each of the seeded cells. Therefore, culture is continued until a colony of a sufficient size is formed.
[0203] 4.それぞれのコロニーから細胞を回収し、別々に培養して十分な量にまで増殖さ せ 100クローン程度のサブラインを確立する。  [0203] 4. Collect cells from each colony, culture them separately, grow to a sufficient amount, and establish a subline of about 100 clones.
[0204] 5.それぞれのサブクローンの細胞を用いてフィーダ一細胞とし、それを用いて ES 細胞の培養を行 、、未分ィ匕状態の支持能の高 、サブラインを選別する。  [0204] 5. One feeder cell is used using the cells of each subclone, and ES cells are cultured using the cells to select a subline having a high ability to support an undivided state.
[0205] ひとつの好ましい実施形態では、本発明において使用される STO株は、 neo抵抗 性であることが有利である。理論に束縛されないが、そのような neo抵抗性にして細 胞株を選択すると、フィーダ一効果の高いものが選択されやすくなる力もである。 neo 抵抗性にするには、 neo1遺伝子を当該分野において公知の任意の遺伝子導入手法 (例えば、形質転換、トランスフエクシヨンなど)を用いて導入することによって達成す ることがでさる。 [0205] In one preferred embodiment, the STO strain used in the present invention is advantageously neo-resistant. Without being bound by theory, such neo-resistant Choosing a cell strain also has the power to make it easier to select one with the highest feeder effect. To neo resistance, neo 1 gene any known gene transfer techniques in the art (e.g., transformation, full Ekushi Yung, etc.) you to achieved by introducing with the leave in.
[0206] 1つの好ましい実施形態において、本発明において使用される細胞株は、 SL10株 であることが有利である。 SL10株は、上記のような方法に基づいて分離された細胞 株である。 SL10株は、明治乳業力 入手可能であり、京都大学再生医科学研究所 において分配されるものであり、第三者は容易に入手することができるものであること が理解される。  [0206] In one preferred embodiment, the cell line used in the present invention is advantageously the SL10 strain. The SL10 strain is a cell strain isolated based on the above method. It is understood that the SL10 strain is available in Meiji Dairy Power, distributed at Kyoto University's Institute of Regenerative Medicine, and readily available to third parties.
[0207] 本発明が支持する幹細胞は、任意の幹細胞が含まれ、例えば、 ES細胞、組織幹 細胞などが挙げられるがそれに限定されない。好ましい実施形態において、本発明 が支持する幹細胞は、 ES細胞である。 ES細胞は、榭立効率の高いフィーダ一細胞 がなぐ本発明は、 ES細胞に関し従来達成可能であった上限である 30%をはるかに 超える 80%もの榭立効率を達成した点で注目されるべきである。  [0207] The stem cells supported by the present invention include any stem cells, and include, but are not limited to, ES cells, tissue stem cells, and the like. In a preferred embodiment, the stem cells supported by the present invention are ES cells. ES cells are a feeder cell with high establishment efficiency The present invention is noted for achieving an establishment efficiency of 80%, far exceeding the previously achievable upper limit of 30% for ES cells Should.
[0208] 別の実施形態では、本発明が支持する幹細胞は、組織幹細胞である。そのような 組織幹細胞としては、例えば、造血幹細胞、神経幹細胞、間葉系幹細胞などが挙げ られるがそれらに限定されない。組織幹細胞もまた、フィーダ一細胞を必要とするも のが多ぐ従来技術では、それほど満足力 ^、くほどに榭立効率を高く達成した技術は 存在していない。従って、本発明は、組織幹細胞にも応用可能であることが理解され る。  [0208] In another embodiment, the stem cells supported by the present invention are tissue stem cells. Examples of such tissue stem cells include, but are not limited to, hematopoietic stem cells, neural stem cells, mesenchymal stem cells, and the like. Conventional techniques, which also require a single feeder cell for tissue stem cells, do not have a technology that achieves so much satisfaction and a particularly high establishment efficiency. Therefore, it is understood that the present invention is also applicable to tissue stem cells.
[0209] より好ましい実施形態では、本発明が支持する幹細胞は、霊長類の細胞であり、よ り好ましくはヒト幹細胞である。ヒトを含む霊長類の細胞は、ごく最近になって ES細胞 が榭立したば力りであるという事情もあり、そのフィーダ一細胞に関する研究はまだま だ発展途上にある。特に、満足のいく榭立効率をもたらすフィーダ一細胞は、霊長類 幹細胞、より好ましくはヒト幹細胞に関してはまったく存在しないといっても過言では ないということにかんがみると、本発明における榭立効率の高さは、特に霊長類幹細 胞、より好ましくはヒト幹細胞にぉ 、て意義は高 、。  [0209] In a more preferred embodiment, the stem cells supported by the present invention are primate cells, more preferably human stem cells. Primate cells, including humans, have only recently been developed as ES cells, and research on feeder cells is still under development. In particular, it is not an exaggeration to say that feeder cells that provide satisfactory establishment efficiency do not exist at all with respect to primate stem cells, more preferably human stem cells. The height is particularly high for primate stem cells, more preferably human stem cells, and the significance is high.
[0210] 本発明のフィーダ一細胞が配置される培養ディッシュはゼラチンコーティングされて いることが好ましいが、それに限定されず、培養効率を高めるものであれば、どのよう なコーティングがされていてもよい。そのようなコーティングとしては、例えば、任意の 細胞外マトリクス (例えば、フイブロネクチン、コラーゲン、ビトロネクチンなど)が挙げら れるがそれらに限定されない。培養ディッシュは、どのような材質のものであってもよく 、例えば、容器の材質は、生体適合性のものが使用されることが望ましいが、生体に 毒性を与えるものが使用されていない限り、どのようなものであっても使用することが できることが理解される。そのような容器の材質としては、例えば、ガラス、ポリエチレ ン、エチレン、ポリプロピレン、ポリイソブチレン、ポリエチレンテレフタレート、不飽和 ポリエステル、含フッ素榭脂、ポリ塩化ビニル、ポリ塩ィ匕ビユリデン、ポリ酢酸ビュル、 ポリビュルアルコール、ポリビュルァセタール、アクリル榭脂、ポリアクリロニトリル、ポリ スチレン、ァセタール榭脂、ポリカーボネート、ポリアミド、フエノール榭脂、ユリア榭脂 、エポキシ榭脂、メラミン榭脂、スチレン'アクリロニトリル共重合体、アクリロニトリルブ タジエンスチレン共重合体、シリコーン榭脂、ポリフエ-レンオキサイド、ポリスルホン 等の有機材料、あるいは、シラン、ポリ Lリジンコートなどでコーティングされたものが 使用され得る。 [0210] The culture dish in which the feeder cells of the present invention are arranged is coated with gelatin. However, the coating is not limited thereto, and any coating may be applied as long as it enhances the culture efficiency. Such coatings include, but are not limited to, for example, any extracellular matrix (eg, fibronectin, collagen, vitronectin, etc.). The culture dish may be made of any material.For example, it is desirable that the material of the container is biocompatible, but unless a material that gives toxicity to the living body is used. It is understood that anything can be used. Examples of the material of such a container include glass, polyethylene, ethylene, polypropylene, polyisobutylene, polyethylene terephthalate, unsaturated polyester, fluorine-containing resin, polyvinyl chloride, polyvinyl chloride, polyvinyl acetate, and polyvinyl acetate. Bull alcohol, polybutyl acetal, acrylic resin, polyacrylonitrile, polystyrene, acetal resin, polycarbonate, polyamide, phenol resin, urea resin, epoxy resin, melamine resin, styrene 'acrylonitrile copolymer, acrylonitrile Organic materials such as butadiene styrene copolymer, silicone resin, polyphenylene oxide, and polysulfone, or those coated with silane, poly-L-lysine, and the like can be used.
[0211] 1つの好ましい実施形態において、本発明のフィーダ一細胞は、約 1 X 104細胞 Z cm2—約 1 X 105細胞/ cm2で播種され、好ましくは、 2— 5 X 105細胞/ cm2で播種 される。理論に束縛されないが、このような適度な細胞密度によって、分泌される細胞 生理活性物質が最適な状態になるからであると考えられる。 [0211] In one preferred embodiment, the feeder single cell of the present invention, about 1 X 10 4 cells Z cm 2 - seeded at approximately 1 X 10 5 cells / cm 2, preferably, 2-5 X 10 5 It is seeded in a cell / cm 2. Without being bound by theory, it is considered that such an appropriate cell density results in an optimal state of the secreted cell bioactive substance.
[0212] 本発明の方法において、本発明のフィーダ一細胞は、通常、混合して播種された 後 5日以内に使用され、好ましくは、 3日以内、より好ましくは 2日以内、さらに好ましく は 1日以内に使用される。ただし、混合後一定時間を置くことが好ましい。理論に束 縛されないが、混合した後、一定時間(例えば、 1時間)を置くことによって、幹細胞の 維持に必要な成分が出てくるに十分な条件が成立する力 である。  [0212] In the method of the present invention, the feeder-cell of the present invention is usually used within 5 days after being mixed and inoculated, preferably within 3 days, more preferably within 2 days, and still more preferably. Used within one day. However, it is preferable to set a fixed time after mixing. Without being bound by theory, after mixing, a certain period of time (eg, 1 hour) is a force that establishes sufficient conditions for the components necessary for stem cell maintenance to emerge.
[0213] 1つの実施形態において、本発明において使用される幹細胞は、ノックアウト血清 代替添加物(KSR)を含む培地中で培養されることが有利である。 KSRは、 Invitrog enなど力も入手可能である。 KSRを用いた場合、全般に細胞の増殖が遅くなる傾向 が見られる力 本発明の目的にかなう限り特に限定して用いることができる。あるいは 、本発明において使用される幹細胞は、ゥシ胎仔血清を含む液体培地中で榭立する ことが好ましい。ゥシ胎仔血清に関しては、 ES細胞を長期間安定に維持できるもの であれば、通常問題なく榭立にも使用することができることが理解される。 [0213] In one embodiment, the stem cells used in the present invention are advantageously cultured in a medium containing knockout serum replacement supplement (KSR). KSR is also available in power, such as Invitrogen. When KSR is used, the ability to generally show a tendency for cell growth to be slow can be used, as long as the object of the present invention is satisfied. Or Preferably, the stem cells used in the present invention are established in a liquid medium containing fetal bovine serum. It is understood that fetal serum can be used without any problem as long as ES cells can be stably maintained for a long period of time.
[0214] 本発明の方法では、継代する工程がさらに包含され、この継代工程において、コラ ゲナーゼが使用される。コラゲナーゼの使用により、細胞回収がスムーズに行われ、 榭立効率を改善するのに寄与すると考えられる。  [0214] The method of the present invention further includes a sub-passing step, in which collagenase is used. It is thought that the use of collagenase facilitates cell recovery and contributes to improving the establishment efficiency.
[0215] (幹細胞)  [0215] (Stem cells)
別の局面において、本発明は、本発明の幹細胞調製法によって調製された幹細胞 調製物を提供する。このような幹細胞は、従来の方法で得られる幹細胞よりも、より未 分ィ匕な状態を保っているとか考えられるという点で従来方法によって調製される幹細 胞とは異なる有利な効果を有すると考えられる。この細胞は、好ましくは、 ES細胞で あり、より好ましくは、霊長類 ES細胞であり、なおさらに好ましくは、ヒト ES細胞である 。このよう ES細胞 (特に、ヒトのような霊長類)は、より発生段階の初期にある未分ィ匕な 状態を保っているという点で従来方法によって調製される幹細胞とは異なる有利な効 果を有すると考えられる。従って、本発明の幹細胞調製物は、 1)従来の方法で得ら れる幹細胞よりも,より未分ィ匕な状態を保って 、る; 2)より正常に近 、組織を支持する 幹細胞を榭立することができる; 3)より正常な組織に近 、性質を有して 、るなどの有 利な点を持っていることがわかる。従来の方法 (例えば、非特許文献 5、 7など)よりも 、本発明の方法ではより簡便に安定して未分ィ匕状態を維持したまま培養できており、 他の方法では常に分ィ匕細胞が混入するのに比較し、本発明の方法では分ィ匕細胞は ほとんど現れないことが示されている。従来の報告では、具体的な数値での報告は 少ないが Peraのグループ (NBt;非特許文献 7)では 50%ほどが部分的に分ィ匕した細 胞集団であるとしているのに対して、本発明では、実施例においても例証されるよう に、 50%未満どころ力 1%程度以下という数値も達成できることが示されている。こ のような幹細胞調製物は、従来は調製できな力つたことから、調製物自体が新規であ ることが理解される。  In another aspect, the present invention provides a stem cell preparation prepared by the method for preparing a stem cell of the present invention. Such stem cells have an advantageous effect different from stem cells prepared by the conventional method in that they are considered to remain in a more undivided state than stem cells obtained by the conventional method. It is thought that. The cells are preferably ES cells, more preferably primate ES cells, and even more preferably human ES cells. Such ES cells (particularly primates such as humans) have an advantageous effect that differs from stem cells prepared by conventional methods in that they remain in an undivided state at an earlier stage of development. It is considered to have Therefore, the stem cell preparation of the present invention can: 1) maintain a more undivided state than stem cells obtained by conventional methods; 2) reduce stem cells that are more normal and support tissue. 3) It is found that it has advantages such as being closer to a normal tissue, having properties, and being more normal. Compared with the conventional method (for example, Non-Patent Documents 5 and 7), the method of the present invention allows the culture to be performed more easily and stably while maintaining the undivided state. It has been shown that compared to the contamination with cells, the method of the present invention shows almost no shunt cells. In conventional reports, there are few reports with specific numerical values, but in the group of Pera (NBt; Non-Patent Document 7), about 50% are cell populations that are partially divided, In the present invention, as exemplified in the examples, it is shown that a numerical value of less than 50% and a force of about 1% or less can be achieved. It is understood that such a stem cell preparation is novel because the preparation itself is novel because it has been difficult to prepare conventionally.
[0216] (再生移植物の調製) [0216] (Preparation of regenerated transplant)
別の局面において、本発明は、臓器、組織または細胞を再生するための移植物を 調製するための方法を提供する。この方法は、 A)所望の臓器、組織または細胞に分 化し得る幹細胞を提供する工程; B)該幹細胞を、正常細胞 (例えば、繊維芽細胞初 代培養細胞)と、細胞株 (例えば、繊維芽細胞株)とを含むフィーダ一細胞調製物の 上で培養する工程;および C)該幹細胞を所望の臓器、組織または細胞を再生する ための移植物へと分ィ匕させる工程、を包含する。ここで、フィーダ一細胞としては、上 記「幹細胞調製法」および「フィーダ一細胞調製物」節で詳細に説明した任意の形態 を用いることができる。幹細胞は、所望の臓器、組織または細胞に分ィ匕し得るもので ある限り、任意のものを使用することができる。所望の臓器、組織または細胞に分化し 得るかどうかは、当該分野において公知の技術を用いて確認することができる。その ような手法としては、分ィ匕条件にその幹細胞を晒し、その後一定期間培養した後、所 望の分ィ匕をしているかどうかを、例えば、肉眼でみたり、顕微鏡により観察したり、ある いは、分子生物学的に観察したり、細胞表面マーカーなどを同定することによるもの などが挙げられるがそれらに限定されない。 In another aspect, the present invention provides a transplant for regenerating an organ, tissue or cell. A method for preparing is provided. The method comprises the steps of: A) providing a stem cell capable of differentiating into a desired organ, tissue or cell; B) converting the stem cell into a normal cell (eg, a primary fibroblast cell) and a cell line (eg, a fiber Culturing on a feeder-cell preparation containing the blast cell line); and C) separating the stem cells into a transplant for regenerating a desired organ, tissue or cell. . Here, as the feeder-cell, any of the forms described in detail in the “Stem cell preparation method” and “Feeder-cell preparation” section can be used. Any stem cells can be used as long as they can be separated into desired organs, tissues or cells. Whether it can be differentiated into a desired organ, tissue or cell can be confirmed using a technique known in the art. As such a technique, after exposing the stem cells to shading conditions and then culturing for a certain period of time, it is checked whether or not the shading is performed as desired, for example, with the naked eye or by observing with a microscope. Alternatively, examples include, but are not limited to, observations by molecular biology and identification of cell surface markers.
[0217] 本発明の再生移植物の調製において、分ィ匕工程は、当該分野において公知の任 意の技術を用いることができる。一つの好ましい実施形態において、本発明の方法 における培養のために、分ィ匕因子をカ卩えることができる。そのような分化因子としては 、例えば、 DNA脱メチル化剤(5—ァザシチジンなど)、ヒストン脱ァセチル化剤(トリコ スタチンなど)、核内レセプターリガンド(例えば、レチノイン酸 (ATRA)、ビタミン D [0217] In the preparation of the regenerative transplant of the present invention, any technique known in the art can be used for the sorting step. In one preferred embodiment, the dangling factor can be removed for culturing in the method of the present invention. Such differentiation factors include, for example, DNA demethylating agents (eg, 5-azacytidine), histone deacetylating agents (eg, trichostatin), nuclear receptor ligands (eg, retinoic acid (ATRA), vitamin D
3、 3,
T3など)、細胞増殖因子(ァクチビン、 IGF— 1、 FGF, PDGF、 TGF— j8、 BMP2/ 4など)、サイト力イン (LIF、 IL-2、 IL— 6など)、へキサメチレンビスァセトアミド、ジメ チルァセトアミド、ジブチル cAMP、ジメチォルスルホキシド、ョードデォキシゥリジン 、ヒドロキシル尿素、シトシンァラビノシド、マイトマイシン C、酪酸ナトリウム、ァフィディ コリン、フルォロデオキシゥリジン、ポリプレン、セレンなどが挙げられるがそれらに限 定されない。神経への分ィ匕には、 NGF、 FGFという分ィ匕因子をカ卩えることができる。 血液細胞への分ィ匕には、インターロイキン、 EPOという分ィ匕因子をカ卩えることができる T3), cell growth factors (activin, IGF-1, FGF, PDGF, TGF-j8, BMP2 / 4, etc.), cytodynamics (LIF, IL-2, IL-6, etc.), hexamethylene bisacetate Amide, dimethyl acetoamide, dibutyl cAMP, dimethyl sulfoxide, ododeoxyperidine, hydroxyl urea, cytosine arabinoside, mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polyprene, selenium And the like, but are not limited thereto. In order to divide the nerve, NGF and FGF can be added. In order to divide blood cells, interleukin and EPO can be added.
[0218] 本発明の再生移植物の調製が目的とする所望の臓器、組織または細胞としては、 神経、血球、骨、軟骨、心臓、心膜、血管、筋肉、眼、肝臓、脾臓、腸、胃、肺、気管、 毛、皮膚などが挙げられるがそれらに限定されない。 [0218] Desired organs, tissues or cells for the purpose of preparing the regenerative transplant of the present invention include nerves, blood cells, bones, cartilage, heart, pericardium, blood vessels, muscles, eyes, liver, spleen, intestine, Stomach, lungs, trachea, Hair, skin, and the like, but are not limited thereto.
[0219] 1つの実施形態にお!、て、上述の所望の臓器、糸且織または細胞と前記フィーダ一 細胞とは、同じ種である。同じ種であることによって、免疫反応を最低限に抑えること も可能であり、適合性という点で好ましい。  [0219] In one embodiment, the desired organ, fibrous tissue or cell and the feeder cell are of the same species. By being the same species, it is also possible to minimize the immune response, which is preferable in terms of compatibility.
[0220] 別の実施形態において、上述の所望の臓器、組織または細胞は霊長類のものであ り、フィーダ一細胞はマウス由来であることが好ましい。この組み合わせで幹細胞の 榭立効率が 80%以上であるという高率を達成することができることがわ力つた力もで ある。しかし、この組み合わせ以外の種の組み合わせもまた、同様に本発明において 好ましくあり得ることが理解されるべきである。従って、フィーダ一細胞としてヒトなどの 霊長類由来のものを使用してもよいことが理解される。  [0220] In another embodiment, the above-mentioned desired organ, tissue or cell is a primate, and the feeder cell is preferably derived from a mouse. This combination is a powerful factor in achieving a high rate of stem cell establishment efficiency of 80% or more. However, it should be understood that combinations of species other than this combination may also be preferred in the present invention. Therefore, it is understood that feeder cells derived from primates such as humans may be used.
[0221] 別の実施形態において、本発明の培養は、ェキソビボ (すなわち、 自己へもどすこ とを目的とする)で行われる。  [0221] In another embodiment, the culture of the present invention is performed ex vivo (ie, for the purpose of returning to self).
[0222] 1つの実施形態において、本発明の幹細胞は、被検体から摘出されてすぐのもの であるか、または凍結保存されたものであり得る。摘出されてすぐ使用することが好ま しい。榭立効率が高いからである。  [0222] In one embodiment, the stem cells of the present invention may be those that have just been removed from a subject or those that have been cryopreserved. It is preferable to use it as soon as it is removed. This is because the establishment efficiency is high.
[0223] 別の実施形態において、本発明の方法における培養は、 37°C、飽和湿度中で 5% CO下であることが好ましぐ分ィ匕のために加えられる分ィ匕因子としては、 DNA脱メ [0223] In another embodiment, the cultivation in the method of the present invention may be performed at 37 ° C under a saturated humidity of 5% CO under a 5% CO concentration. , DNA removal
2 2
チル化剤(5—ァザシチジンなど)、ヒストン脱ァセチル化剤(トリコスタチンなど)、核内 レセプターリガンド (例えば、レチノイン酸 (ATRA)、ビタミン D  Tilating agents (eg, 5-azacytidine), histone deacetylating agents (eg, trichostatin), nuclear receptor ligands (eg, retinoic acid (ATRA), vitamin D)
3、 T3など)、細胞増殖 因子(ァクチビン、 IGF— 1、 FGF, PDGF、 TGF— j8、 BMP2Z4など)、サイト力イン (LIF、 IL 2、 IL— 6など)、へキサメチレンビスァセトアミド、ジメチルァセトアミド、ジブ チル cAMP、ジメチォルスルホキシド、ョードデォキシゥリジン、ヒドロキシル尿素、シ トシンァラビノシド、マイトマイシン C、酪酸ナトリウム、ァフイディコリン、フルォロデオキ シゥリジン、ポリプレン、セレンなどを 1または複数を含んでいることが好ましい。  3, T3, etc.), cell growth factors (activin, IGF-1, FGF, PDGF, TGF-j8, BMP2Z4, etc.), cytodynamics (LIF, IL 2, IL-6, etc.), hexamethylene bisacetamide , Dimethylacetamide, dibutyl cAMP, dimethyl sulfoxide, eododexperidine, hydroxyl urea, cytosine arabinoside, mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polyprene, selenium, etc. Preferably, one or more are included.
[0224] (臓器など) [0224] (Organs, etc.)
別の局面において、本発明は、本発明の再生移植物の調製法によって調製される 、臓器、組織または細胞を提供する。このような臓器、組織または細胞は、より正常な 組織に近 、性質を有して 、ると 、う点で従来方法によって調製される再生臓器、組 織または細胞とは異なる有利な効果を有すると考えられる。 In another aspect, the present invention provides an organ, tissue or cell prepared by the method for preparing a regenerative transplant of the present invention. Such an organ, tissue or cell is similar to a normal tissue and has properties in that it can be a regenerated organ or tissue prepared by a conventional method. It is thought to have a different beneficial effect than tissue or cells.
[0225] (臓器等再生システム)  [0225] (Organ regeneration system)
別の局面において、本発明は、臓器、組織または細胞を再生するためのシステムを 提供する。このシステムは、 A)容器;および B)該容器上に播種される、正常細胞 (例 えば、繊維芽細胞初代培養細胞)と、細胞株 (例えば、繊維芽細胞株)とを含むフィ ーダー細胞調製物、を備える。ここで、このフィーダ一細胞調製物は、上記「フィーダ 一細胞調製物」および「幹細胞調製法」において説明される任意の形態をとることが できることが理解される。容器もまた、任意の物を利用することができ、例えば、「再生 移植物の調製」節において説明される任意の形態をとることができることが理解される  In another aspect, the present invention provides a system for regenerating an organ, tissue or cell. The system comprises: A) a container; and B) feeder cells seeded on the container, comprising normal cells (eg, primary fibroblasts) and a cell line (eg, a fibroblast cell line). Preparation. Here, it is understood that the feeder-one-cell preparation can take any form described in the above “feeder-one-cell preparation” and “stem cell preparation method”. It is understood that the container can also utilize any material, for example, take any of the forms described in the section "Preparing a Regenerated Implant".
[0226] (細胞混合物) [0226] (Cell mixture)
本発明は、正常細胞 (例えば、繊維芽細胞初代培養細胞)と、細胞株 (例えば、繊 維芽細胞株)とを含む混合物を提供する。この混合物は、本発明において実証され るように、幹細胞のフィーダ一細胞として使用することができる。正常細胞 (例えば、 繊維芽細胞初代培養細胞)と、細胞株 (例えば、繊維芽細胞株)とは、「幹細胞調製 法」および「フィーダ一細胞調製物」において説明されるような任意の形態をとること ができることが理解される。従って、実質的には、この細胞混合物は、フィーダ一細胞 調製物と同一のものであり得ることが理解され得る。  The present invention provides a mixture comprising normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts). This mixture can be used as a stem cell feeder cell, as demonstrated in the present invention. Normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts) can be any form as described in “Stem cell preparation method” and “Feeder-one cell preparation”. It is understood that it can be taken. Thus, it can be appreciated that substantially the cell mixture may be the same as the feeder-cell preparation.
[0227] 好ましい実施形態において、本発明の混合物中の初代培養細胞は、マウス胎児初 代培養繊維芽細胞であることが有利である。この形態が有利なのは、 STO細胞など 、他の細胞力 得られない因子などが供給され、従来にない効率で幹細胞を榭立す ることができること、より正常に近い組織を支持する幹細胞を榭立することができること など 、う効果が考えられるからである。  [0227] In a preferred embodiment, the primary cultured cells in the mixture of the present invention are advantageously mouse fetal primary cultured fibroblasts. This form is advantageous because stem cells can be established with unprecedented efficiency by supplying factors such as STO cells and other factors that cannot be obtained, and stem cells supporting more normal tissues can be established. This is because it is possible to do so.
[0228] 別の好ましい実施形態において、本発明の混合物中の繊維芽細胞株は、 STO株 であることが有利である。この形態が有利なのは、マウス胎仔繊維芽細胞から供給さ れる因子が得られることにより、従来にない効率で幹細胞を榭立することができること 、より正常に近い組織を支持する幹細胞を榭立することができることなどという効果が 考えられる力 である。 [0229] (臓器等再生方法) [0228] In another preferred embodiment, the fibroblast cell line in the mixture of the present invention is advantageously an STO line. The advantage of this form is that the factor supplied from mouse fetal fibroblasts can be obtained, so that stem cells can be established with unprecedented efficiency, and that stem cells that support more normal tissues can be established. It is a force that can be considered as an effect that can be done. [Methods for Regenerating Organs, etc.]
別の局面において、本発明は、臓器、組織または細胞を再生するための方法を提 供する。この方法は、 A)所望の臓器、組織または細胞に分ィ匕し得る幹細胞を提供す る工程; B)該幹細胞を、正常細胞 (例えば、繊維芽細胞初代培養細胞)と、細胞株( 例えば、繊維芽細胞株)とを含むフィーダ一細胞調製物上で培養する工程;および C )該培養された該幹細胞を被検体の処置されるべき部位に移植する工程、を包含す る。ここで使用されるフィーダ一細胞は、上記「フィーダ一細胞調製物」および「幹細 胞調製法」において説明される任意の形態をとることができることが理解される。幹細 胞もまた、上記「フィーダ一細胞調製物」および「幹細胞調製法」において説明される 任意の形態をとることができることが理解される。移植技術もまた、当該分野において 公知の任意の技術を用いて投与することができる。そのような技術としては、例えば、 カテーテルを用いて心筋層への幹細胞移植などが挙げられるがそれらに限定されな い。  In another aspect, the present invention provides a method for regenerating an organ, tissue or cell. This method comprises the steps of: A) providing a stem cell capable of dividing into a desired organ, tissue or cell; B) converting the stem cell into a normal cell (eg, a primary fibroblast cell) and a cell line (eg, , A fibroblast cell line) and C) transplanting the cultured stem cells to a site to be treated in a subject. It is understood that the feeder cell used herein can take any of the forms described in the above "feeder cell preparation" and "stem cell preparation method". It is understood that the stem cells can also take any of the forms described in the “Feeder Cell Preparations” and “Stem Cell Preparation Methods” above. Transplantation techniques can also be administered using any technique known in the art. Such techniques include, but are not limited to, stem cell transplantation into the myocardium using a catheter, for example.
[0230] 好ましい実施形態において、本発明の方法は、 D)前記幹細胞を分化させる工程を さらに包含する。このような分ィ匕工程は、当該分野において公知の方法を適用するこ とができる。このような分ィ匕条件は、分ィ匕の目的となる臓器、組織または細胞によって 変動し得ることが理解される。そのような変動を考慮して、当業者は、適宜、適切な分 化条件を決定し、選択することができることが理解される。  [0230] In a preferred embodiment, the method of the present invention further comprises D) a step of differentiating the stem cell. For such a dividing process, a method known in the art can be applied. It is understood that such conditions can vary depending on the target organ, tissue or cell. It is understood that those skilled in the art can, in consideration of such variations, appropriately determine and select appropriate fragmentation conditions.
[0231] (混合物のフィーダ一細胞としての使用)  [0231] (Use of mixture as feeder-cell)
別の局面において、本発明は、フィーダ一細胞調製物としての、正常細胞 (例えば 、繊維芽細胞初代培養細胞)と、細胞株 (例えば、繊維芽細胞株)とを含む細胞混合 物の使用を提供する。あるいは、フィーダ一細胞としての、繊維芽細胞初代培養細胞 と、繊維芽細胞株とを含む細胞混合物の使用が提供される。あるいは、幹細胞の使 用が適切である疾患、障害または状態の処置または予防のための、幹細胞を含む医 薬の製造における、繊維芽細胞初代培養細胞と、繊維芽細胞株とを含むフィーダ一 細胞調製物の使用が提供される。  In another aspect, the invention provides the use of a cell mixture comprising normal cells (eg, primary fibroblasts) and cell lines (eg, fibroblasts) as a feeder-cell preparation. provide. Alternatively, there is provided the use of a cell mixture comprising primary fibroblast cells and a fibroblast cell line as feeder cells. Alternatively, a feeder cell containing a primary cultured fibroblast cell and a fibroblast cell line in the manufacture of a medicament containing a stem cell for the treatment or prevention of a disease, disorder or condition for which the use of a stem cell is appropriate. Use of the preparation is provided.
[0232] ここで使用されるフィーダ一細胞は、上記「幹細胞調製法」および「フィーダ一細胞 調製物」において説明される任意の形態をとることができることが理解される。フィー ダー細胞は、任意の細胞を支持することができる力 好ましくは幹細胞を支持するこ とを目的とする。ここで、幹細胞もまた、上記「フィーダ一細胞調製物」および「幹細胞 調製法」において説明される任意の形態をとることができることが理解される。 [0232] It is understood that the feeder cell used herein can take any of the forms described in the above "stem cell preparation method" and "feeder cell preparation". Fee Dar cells are intended to support any cells, preferably stem cells. Here, it is understood that the stem cells can also take any of the forms described in the above “feeder-one cell preparation” and “stem cell preparation method”.
[0233] 他の局面において、本発明は、フィーダ一細胞を含む医薬を製造するための、正 常細胞 (例えば、繊維芽細胞初代培養細胞)と、細胞株 (例えば、繊維芽細胞株)と を含む細胞混合物の使用を提供する。ここで使用されるフィーダ一細胞は、上記「幹 細胞調製法」および「フィーダ一細胞調製物」において説明される任意の形態をとる ことができることが理解される。フィーダ一細胞は、任意の細胞を支持することができ るが、好ましくは幹細胞を支持することを目的とする。ここで、幹細胞もまた、上記「幹 細胞調製法」および「フィーダ一細胞調製物」において説明される任意の形態をとる ことができることが理解される。医薬を製造する技術は、当該分野において公知の技 術を用いることができる。医薬には、適宜薬学的に受容可能なキャリアを加えることが できる。  [0233] In another aspect, the present invention relates to a normal cell (for example, a primary fibroblast cell) and a cell line (for example, a fibroblast cell line) for producing a medicament containing a feeder cell. The use of a cell mixture comprising: It is understood that the feeder cells used herein can take any of the forms described in the “Stem cell preparation method” and “Feeder cell preparation” above. The feeder cell can support any cells, but preferably aims to support stem cells. Here, it is understood that the stem cells can also take any of the forms described in the above “Stem cell preparation method” and “Feeder-one cell preparation”. As a technique for producing a medicament, a technique known in the art can be used. A pharmaceutically acceptable carrier can be appropriately added to the medicament.
[0234] 好ましくは、上記フィーダ一細胞調製物は、増殖しないように処理されている(例え ば、マイトマイシン Cによる)。このことにより、フィーダ一細胞調製物は、パッケージに 入れて販売することが可能になる。  [0234] Preferably, the feeder-cell preparation is treated so as not to proliferate (for example, with mitomycin C). This allows the feeder-cell preparation to be sold in a package.
[0235] 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、上記発明の詳細な説明にも下記実施例 にも限定されるものではなぐ請求の範囲によってのみ限定される。  Hereinafter, the present invention will be described based on examples, but the following examples are provided for illustrative purposes only. Accordingly, the scope of the present invention is not limited to the above detailed description of the invention nor to the following Examples, but only by the appended claims.
実施例  Example
[0236] 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、 [0236] The reagents used in the examples shown below were Wako Pure Chemical unless otherwise specified.
Sigma力ら得た。動物の飼育 ίま、 National Society for Medical Researches 作成した「Principles of Laboratory Animal CareJおよひnstitute of Lab oratory Animal Resource力 S作成、 National Institute of Health力 S公表し た「Guide for the Care and Use of Laboratory Animals」 (NIH Publi cation, No. 86— 23, 1985,改訂)に遵つて、京都大学および日本政府が規 定する基準に遵ぃ、動物愛護精神に則って行った。ヒトを対象とする場合は、厚生労 働省または等価の政府当局の基準に従い、事前に同意を得た上で実験を行う。その 同意方法は、本明細書において記載されるとおりである。 Sigma obtained. Animal breeding Puma, created by the National Society for Medical Researches, `` Principles of Laboratory Animal Care J and the Institute of Laboratory Animal Resource S '', published by the National Institute of Health S, `` Guide for the Care and Use of Laboratory Animals ”(NIH Publication, No. 86-23, 1985, revised), in compliance with the standards stipulated by Kyoto University and the Japanese government, and in accordance with the spirit of animal protection. In the case of human subjects, conduct experiments with the prior consent of the Ministry of Health, Labor and Welfare or equivalent governmental authorities. That The consent method is as described herein.
[0237] (実施例 1:マウス胎仔繊維芽細胞の初代培養細胞 (MEF)の調製) (Example 1: Preparation of primary cultured cells (MEF) of mouse fetal fibroblasts)
マウス胎仔力も繊維芽細胞の初代培養細胞 (MEFともいう)を調製した。具体的に は、以下のとおりである。  Primary embryonic fibroblast cells (also referred to as MEFs) were prepared for mouse fetal power. Specifically, it is as follows.
[0238] 妊娠したマウス (ICR、 日本クレア)から、胎仔の部分を取り出し、細胞を分離して、 DMEM+ 10%ゥシ胎仔血清 (FBS)という培地中に播種した。取り出した細胞は、 D MEM + 10%FBS中で維持した。 [0238] A part of the fetus was removed from a pregnant mouse (ICR, CLEA Japan), the cells were separated, and seeded in a medium called DMEM + 10% fetal serum (FBS). The removed cells were maintained in DMEM + 10% FBS.
[0239] (実施例 2 : MEF細胞の前処理) (Example 2: Pretreatment of MEF cells)
次に、フィーダ一細胞としての効率を上げるために、実施例 1で調製した MEF細胞 を前処理した。前処理には、以下のものを使用した。  Next, in order to increase the efficiency as a single feeder cell, the MEF cells prepared in Example 1 were pretreated. The following were used for the pretreatment.
[0240] 培養液: DMEM + 10%FCS [0240] Culture solution: DMEM + 10% FCS
PBS (Ca, Mg不含)  PBS (without Ca, Mg)
0. 05%トリプシン + lmM EDTA  0.05% Trypsin + lmM EDTA
2mgZmlマイトマイシン C水溶液  2mgZml mitomycin C aqueous solution
0. 1%ゼラチン水溶液。  0.1% gelatin aqueous solution.
[0241] 前処理は以下のとおり行った。 [0241] Pretreatment was performed as follows.
[0242] (1)コンフルェントの細胞にマイトマイシン Cを 10 μ g/mlになるよう培地に加え、 1.5 一 2時間培養した。  (1) Mitomycin C was added to the confluent cells at a concentration of 10 μg / ml in the medium, and the cells were cultured for 1.5 to 12 hours.
[0243] (2)マイトマイシン C処理した培養皿から培地を除き PBSで 3回洗った。培地を加え [0243] (2) The medium was removed from the culture dish treated with mitomycin C and washed three times with PBS. Add medium
、数時間から一晩培養した。 And cultured for several hours to overnight.
[0244] (3)培地を除き PBSで 3回洗い、トリプシン処理により細胞を解離し、培地を加え細 胞をよく懸濁した後、遠心して上清を除いた。 [0244] (3) The medium was removed, washed three times with PBS, the cells were dissociated by trypsin treatment, the medium was added, the cells were well suspended, and the mixture was centrifuged to remove the supernatant.
[0245] (4)細胞を lxl05/mlの濃度に懸濁させた。 [0245] (4) was suspended in a concentration of cells lxl0 5 / ml.
[0246] このようにして調製した MEF細胞を、単独でまたは他の細胞と混合してフィーダ一 細胞として以下に実験に用いた。  [0246] The MEF cells thus prepared were used alone or in a mixture with other cells as feeder cells in the following experiments.
[0247] (実施例 3 : STO細胞の前処理) (Example 3: Pretreatment of STO cells)
STO細胞として、 STO細胞のサブクローン SL10細胞を使用した。 SL10細胞は、 明治乳業 (東京、 日本)から入手した。 SL10の場合、 2週間がフィーダ一細胞として の利用限度であり、 7— 10日で継代を行った。培地としては DMEM+ 10%FCSを 使用した。 As STO cells, subclone SL10 cells of STO cells were used. SL10 cells were obtained from Meiji Dairies (Tokyo, Japan). In the case of SL10, 2 weeks is one feeder cell It is the limit of use, and it was subcultured in 7-10 days. DMEM + 10% FCS was used as the medium.
[0248] この STO細胞を、フィーダ一細胞として使用するために前処理した。その手順は以 下のとおりである。前処理には、以下のものを使用した。  [0248] The STO cells were pretreated for use as feeder cells. The procedure is as follows. The following were used for the pretreatment.
[0249] 培養液: DMEM + 10%FCS [0249] Culture solution: DMEM + 10% FCS
PBS (Ca, Mg 不含)  PBS (Ca, Mg free)
0. 05%トリプシン + lmM EDTA  0.05% Trypsin + lmM EDTA
2mgZmlマイトマイシン C水溶液  2mgZml mitomycin C aqueous solution
0. 1%ゼラチン水溶液。  0.1% gelatin aqueous solution.
[0250] 前処理は以下のとおり行った。 [0250] Pretreatment was performed as follows.
[0251] (1)コンフルェントの細胞にマイトマイシン Cを 10 gZmlになるよう培地に加え、 1 . 5— 2時間培養した。  [0251] (1) Mitomycin C was added to the confluent cells to a medium at 10 gZml, and the cells were cultured for 1.5 to 2 hours.
[0252] (2)マイトマイシン C処理した培養皿から培地を除き PBSで 3回洗った。培地を加え (2) The culture medium was removed from the culture dish treated with mitomycin C and washed three times with PBS. Add medium
、数時間から一晩培養した。 And cultured for several hours to overnight.
[0253] (3)培地を除き PBSで 3回洗い、トリプシン処理により細胞を解離し、培地を加え細 胞をよく懸濁した後、遠心して上清を除いた。 (3) The medium was removed, washed three times with PBS, the cells were dissociated by trypsinization, the medium was added, the cells were well suspended, and the mixture was centrifuged to remove the supernatant.
[0254] (4)細胞を 1. 5 X 105/mlの濃度に懸濁した。 (4) The cells were suspended at a concentration of 1.5 × 10 5 / ml.
[0255] (実施例 4 :別の STO細胞の調製) (Example 4: Preparation of another STO cell)
別の実施例として、 STO細胞力も好適なクローンをサブクローユングする。その手 順は以下のとおりである。  As another example, STO cell strength also subclones suitable clones. The procedure is as follows.
[0256] マウスにおいて ES細胞株の榭立.継代維持に用いられる STO細胞は株細胞であ り均一な集団と考えられていた力 その中に特に ES細胞株の榭立 ·維持に優れた性 質を持つ細胞集団を含むことが示されて!/ヽる。 [0256] Establishment of ES cell line in mice. STO cells used for maintaining the passage were cell lines and were considered to be a uniform population. Among them, it was particularly excellent in establishing and maintaining ES cell lines. It has been shown to contain cell populations of the nature!
[0257] 同様の性質を持つ STO細胞のサブラインは以下のように単離できる。 [0257] A subline of STO cells with similar properties can be isolated as follows.
[0258] (1)定法に従い STO細胞を細胞一つ一つになるまで解離する。 [0258] (1) Dissociate STO cells into individual cells according to a standard method.
[0259] (2)培養皿に 100— 500個程度の解離した STO細胞を播種する。 (2) About 100 to 500 dissociated STO cells are seeded on a culture dish.
[0260] (3)数日間培養すると播種した細胞一つ一つの細胞それぞれ力 コロニーが形成 されるので十分な大きさのコロニーが形成されるまで培養を続ける。 [0261] (4)それぞれのコロニー力も細胞を回収し、別々に培養して十分な量にまで増殖さ せ 100クローン程度のサブラインを確立する。 [0260] (3) After culturing for several days, a strong colony is formed for each of the seeded cells. Therefore, the culture is continued until a colony of a sufficient size is formed. [0261] (4) Cells are also collected from each colony, and cultured separately to grow to a sufficient amount to establish a subline of about 100 clones.
[0262] (5)それぞれのサブクローンの細胞を用いてフィーダ一細胞とし、それを用いて ES 細胞の培養を行 、、未分ィ匕状態の支持能の高 、サブラインを選別する。 [0262] (5) One feeder cell is used by using the cells of each subclone, and ES cells are cultured using the cells to select a subline having a high ability to support an undivided state.
[0263] このようにしてサブクローユングした細胞株もまた、フィーダ一細胞として使用できる[0263] The cell line subcloned in this manner can also be used as a feeder cell.
。このようなサブクローンを実施例 3に記載のように前処理した。 . Such subclones were pre-treated as described in Example 3.
[0264] (実施例 5 :フィーダ一細胞の調製) (Example 5: Preparation of feeder single cell)
本実施例では、フィーダ一細胞を調製した。本発明のフィーダ一細胞として、繊維 芽細胞初代培養細胞と、繊維芽細胞株とを単独で使用した場合と、混合した場合と を調製した。フィーダ一細胞の調製は、以下に示すように同様のプロトコールに従つ た。  In this example, one feeder cell was prepared. As a single feeder cell of the present invention, a case where a fibroblast primary culture cell and a fibroblast cell line were used alone and a case where they were mixed were prepared. Preparation of feeder cells followed a similar protocol as shown below.
[0265] (1)フィーダ一を作る培養皿を予めゼラチンコートしておいた。ゼラチンコートは培 養皿の表面をゼラチン溶液で覆い 37°Cで 1時間以上インキュベートして行った。  (1) A culture dish for preparing a feeder was gelatin-coated in advance. The gelatin coating was performed by covering the surface of the culture dish with a gelatin solution and incubating at 37 ° C for 1 hour or more.
[0266] (2)前処理した STO細胞、前処理した MEF細胞を単独でまたは懸濁液を 1: 1に 混合して用いた。  (2) Pretreated STO cells and pretreated MEF cells were used alone or as a 1: 1 mixture of suspensions.
[0267] (3)ゼラチンコートした培養皿力もゼラチン溶液を除き、細胞サンプルを加えた。 35 mmの培養皿に 2mlの細胞懸濁液を加えると培養皿の表面をフィーダ一細胞が隙間 なく覆った。細胞密度はおよそ 2. 5 X 104/cm2になった。 [0267] (3) The gelatin dish was removed from the culture dish coated with gelatin, and a cell sample was added. When 2 ml of the cell suspension was added to a 35 mm culture dish, the feeder cells covered the surface of the culture dish without gaps. The cell density was approximately 2.5 × 10 4 / cm 2 .
[0268] 調製したフィーダ一細胞は 4日以内に使用した。  [0268] The prepared feeder cells were used within 4 days.
[0269] (実施例 6 :幹細胞の榭立)  (Example 6: Establishment of stem cells)
次に、実施例 5で調製したフィーダ一細胞を用いて幹細胞を榭立した。使用した培 養液は以下のとおりである。  Next, stem cells were established using the feeder cells prepared in Example 5. The culture solutions used are as follows.
[0270] (ES細胞用培養液) 培地組成: [0270] (Culture for ES cells) Medium composition:
D M E M/ F 1 2 ( s i g m a D 6 4 2 1 ) 8 0 m  D M E M / F 1 2 (sig ma a D 6 4 2 1) 80m
非必須アミノ酸 (G i b c 0 ) 0 . 8 m  Non-essential amino acids (Gibc0) 0.8 m
2 0 O mM L グルタミン  20 O mM L glutamine
K S R ( G i b c o ) 2 0 m  K S R (G i b c o) 20 m
2 一 M E 0 . 8 μ  2 1 M E 0.8 μ
ヒ ト L I F ( 1 0 ,u g / 1 ) 0 0 μ I ( 1 O ng/ml) 塩基性繊維芽細胞増殖因子( 1 m g /m 1 ) ϋ . 4 /i 1 ( 4 ng/ml)。 Human LIF (10, ug / 1) 00 μI (1 ng / ml) Basic fibroblast growth factor (1 mg / m1) ϋ .4 / i1 (4 ng / ml).
[0271] (細胞解離液) [0271] (Cell Dissociation Solution)
0. 25% トリプシン(PBS)  0.25% trypsin (PBS)
ImM CaCl  ImM CaCl
2  2
20%KSR。  20% KSR.
[0272] (ディッシュ) [0272] (Dish)
フィーダ一をつくる培養ディッシュは予めゼラチンコートしておく。 0. 1%ゼラチン溶 液(swine skin, Type A: Sigma 社製)で覆い、 37°Cで 1 時間以上インキュべ ートする。培養ディッシュのゼラチン溶液を除き、細胞の懸濁液を加える。数時間経 てばフィーダ一細胞として使用可能である。  The culture dish for making the feeder should be gelatin-coated in advance. 0.1 Cover with 1% gelatin solution (swine skin, Type A: Sigma) and incubate at 37 ° C for 1 hour or more. Remove the gelatin solution from the culture dish and add the cell suspension. After several hours, it can be used as a feeder cell.
[0273] フィーダ一細胞を用いた ES細胞の榭立は、以下のように行った。 [0273] ES cells were established using a single feeder cell as follows.
[0274] 1.フィーダ一細胞の培養液を ES細胞用の培養液に交換した。 [0274] 1. The culture solution of one feeder cell was replaced with a culture solution for ES cells.
[0275] 2.免疫手術法もしくは機械的操作により胚盤胞カゝら内部細胞塊を分離した。免疫 手術法は、簡単に述べると、以下のとおりである。 [0275] 2. The inner cell mass of the blastocyst was isolated by immunosurgery or mechanical operation. Briefly, the immunosurgical procedure is as follows.
[0276] 酵素処理などにより透明帯を取り除き、マウス表面抗原に反応する抗体溶液内でィ ンキュペートした。次に補体溶液内でインキュベートすることによって最外層の細胞で ある栄養外胚葉が取り除かれ、内部細胞塊が分離された。 [0276] The zona pellucida was removed by enzyme treatment or the like, and the cells were incubated in an antibody solution that reacts with the mouse surface antigen. The outermost cells, vegetative ectoderm, were then removed by incubation in complement solution, and the inner cell mass was separated.
[0277] ヒト ES細胞の場合は、ヒトの細胞表面抗原に反応する抗体溶液を用いることに留意 して上記手順を行った。力-クイサル((株)ケアリー力も入手)の場合は、サルの細胞 表面抗原に反応する抗体溶液を用いることに留意して上記手順を行った。 [0277] In the case of human ES cells, the above procedure was performed while keeping in mind that an antibody solution that reacts with human cell surface antigens was used. In the case of Riki-Quisal (also available from Cary Power Co., Ltd.), the above procedure was performed while keeping in mind that an antibody solution that reacts with the monkey cell surface antigen was used.
[0278] 3.分離した内部細胞塊をフィーダ一細胞上で培養した。 [0278] 3. The separated inner cell mass was cultured on one feeder cell.
[0279] 4.翌日内部細胞塊がフィーダ一細胞に接着して 、ることを確認して、培養液を交 換した。以後毎日培地交換した。 [0279] 4. Check that the inner cell mass adheres to the feeder cell the next day, and exchange the culture solution. Changed. Thereafter, the medium was changed every day.
[0280] 5. 5— 10日後に内部細胞塊力も増殖してくる ES細胞様の細胞が認められるように なった。細胞塊が 100から 200 mになったら継代を行った。  [0280] After 5 to 10 days, ES cell-like cells in which the inner cell mass also proliferated became evident. Subculture was performed when the cell mass reached 100 to 200 m.
[0281] 6.培養液をのぞき細胞解離液を入れた。 [0281] 6. Except for the culture solution, the cell dissociation solution was added.
[0282] 7. 5から 10分間処理すると ES細胞様細胞が培養皿よりはがれてくるので、これを 細く引 ヽたガラス管で拾 、上げ数回出し入れすることで細胞塊を数個に分けた。  [0282] After treatment for 7.5 to 10 minutes, ES cell-like cells came off the culture dish. The cells were picked up with a thinly drawn glass tube, raised and removed several times to divide the cell mass into several pieces. .
[0283] 8.培養液中で細胞解離液を洗浄して除き、新しいフィーダ一上に細胞塊を移した [0283] 8. The cell dissociation solution was washed away in the culture solution, and the cell mass was transferred onto a new feeder.
[0284] 9.翌日、培養液を交換した。以後毎日培地交換した。 [0284] 9. The next day, the culture solution was replaced. Thereafter, the medium was changed every day.
[0285] 10.フィーダ一上で個々の細胞塊が 100— 200 μ m程度に増殖したら、同様の操 作で継代を行った。  [0285] 10. When individual cell clusters grew to about 100 to 200 µm on the feeder, subculture was performed by the same operation.
[0286] 11.細胞が十分増殖するようになれば(35mm培養皿 1枚に増えるぐらい)サル ES 細胞の通常の継代操作と同様に継代ができるようになった。この段階に達すれば以 後安定した培養が可能であることが確認される。  [0286] 11. If the cells were able to proliferate sufficiently (the number of cells increased to one per 35 mm culture dish), the cells could be passaged in the same manner as the normal passage operation of monkey ES cells. It is confirmed that stable culture is possible after this stage is reached.
[0287] この実施例で榭立された細胞を図 1および 2に示す(図 1はサル、図 2はヒトを示す)  [0287] The cells established in this example are shown in Figures 1 and 2 (Figure 1 shows monkeys, Figure 2 shows humans)
[0288] 本実施例では、基本培地に DMEM/F12を使用した力 DMEMまたは他の基本培地 でも問題なく使用できることが示される。 [0288] This example shows that DMEM / F12 as a basic medium can be used without any problem even with DMEM or another basic medium.
[0289] 本実施例では、補填成分として KSRを使用した力 KSRのかわりに通常使用され ているゥシ胎仔血清でもよいようである。ただし、 KSRのほうがゥシ胎仔血清よりも良 い結果が出るようであった。 [0289] In the present example, the strength of using KSR as a supplementing component It seems that instead of KSR, commonly used fetal serum may be used. However, KSR seemed to give better results than fetal fetal serum.
[0290] さらに必要に応じて 1一 2mg/mlのコラゲナーゼを添カ卩した。コラゲナーゼの添加に より、若干榭立しゃすくなるようである。 [0290] Further, if necessary, 1-2 mg / ml collagenase was added thereto. With the addition of collagenase, it appears to be slightly upright.
[0291] (実施例 7 :マーカーによる染色) (Example 7: Staining with marker)
次に、実施例 6で榭立した細胞の未分化状態を、特異的マーカーによる染色により 調べた。胚性幹細胞を、 4% PFAで固定し標準的な培養細胞の免疫染色法 (Willi ngham, M. C. et. al. , 1985. An Atlas of Immunofluorescence in C ultured Cell, Academic Press, Orlando, FL, pp. 1—13. )【こ従 ヽ免疫 染色を行った。ブロッキングは 0. 1% Triton X/PBS/2% skim milkを用い 室温で一時間、洗いは 0. 1% TritonXZPBSを用い室温で 5分を 4回行った。一 次抗体は SSEA-4、 TRA-1-60のモノクローナル抗体をそれぞれ 200 μ g/ml (CL ONTECH)を 1Z100希釈したものを、二次抗体は FITC標識 goat 抗ー mouse I gG (H + L) (ZYMED LABORATORIES, INC)を 1Z200希釈したものを使 用した。二次抗体の反応後は rhodamine phalloidin (Molecular Probe)、 DAP I (SIGMA)の順に染色を行 、シグナルを検出した。 Next, the undifferentiated state of the cells established in Example 6 was examined by staining with a specific marker. Embryonic stem cells were fixed with 4% PFA and immunostained in standard cultured cells (Willingham, MC et.al., 1985.An Atlas of Immunofluorescence in Cultured Cell, Academic Press, Orlando, FL, pp. 1-13. Staining was performed. Blocking was performed using 0.1% Triton X / PBS / 2% skim milk for 1 hour at room temperature, and washing was performed 4 times using 0.1% Triton XZPBS for 5 minutes at room temperature. The primary antibody was a 200 μg / ml (CL ONTECH) 1Z100 diluted monoclonal antibody of SSEA-4 and TRA-1-60, respectively, and the secondary antibody was FITC-labeled goat anti-mouse IgG (H + L ) (ZYMED LABORATORIES, INC) diluted 1Z200 was used. After the reaction with the secondary antibody, staining was performed in the order of rhodamine phalloidin (Molecular Probe) and DAPI (SIGMA) to detect a signal.
[0292] ALP (アルカリホスファターゼ)の組織ィ匕学的染色は、以下のように行った。培養デ イツシュをダルベッコの PBS (—)で 2回洗浄した後、 4°Cの 95%冷エタノールで 30分 以上固定し、次いで 4 °Cの無水エタノールで 30分以脱水した。固定液を除き、ディ ッシュを室温で 0. 1M Tris-HCl (pH9. 0-9. 5)にて 5 分間 3 回洗浄した後、 1 . 5— 2mlの染色液(ナフトール AS— BIホスフェート(Sigma、カタログ番号 N— 2125 ) 2. 5mg と、ファースト赤 TR塩(Sigma) 15mg [またはファースト紫 B 塩 6mg (Sig ma)、ファースト青 BB塩 12. 5mg (Sigma) ]とを 0. 1M Tris— HC1 緩衝液 25ml に加え、遮光下で 3 — 5 分間撹拌して溶解し、次いで濾過する)を各ディッシュに 加え、遮光下で室温にて ALP を 15— 30分間染色した。 PBS (—)で 2 回洗浄した 後グリセロールを加え位相差立体顕微鏡で赤褐色に染まった EG細胞コロニーを検[0292] ALP (alkaline phosphatase) histologic staining was performed as follows. The culture tissue was washed twice with Dulbecco's PBS (-), fixed with cold 95% ethanol at 4 ° C for 30 minutes or more, and then dehydrated with absolute ethanol at 4 ° C for 30 minutes or more. After removing the fixative, the dishes were washed three times for 5 minutes at room temperature with 0.1 M Tris-HCl (pH 9.0-9.5), and then 1.5--2 ml of the staining solution (Naphthol AS-BI phosphate ( Sigma, Catalog No. N-2125) 2. 5 mg and fast red TR salt (Sigma) 15 mg [or fast purple B salt 6 mg (Sig ma), fast blue BB salt 12.5 mg (Sigma)] in 0.1 M Tris — Add 25 ml of HC1 buffer, dissolve with stirring for 3-5 minutes in the dark, then filter) and stain with ALP for 15-30 minutes at room temperature in the dark. After washing twice with PBS (-), glycerol was added, and a red-brown stained EG cell colony was examined using a phase contrast microscope.
(し/こ。 (Shi / ko.
[0293] 結果を図 3に示す。図 3は、左力ら ALP、 SSEA-4、 TRA-1-60を示す。  [0293] The results are shown in FIG. Fig. 3 shows ALP, SSEA-4, and TRA-1-60 from left power.
[0294] 図 3からも明らかなように、本発明のフィーダ一細胞を用いて榭立した幹細胞は通 常どおり多分化能を保持して ヽることが確認された。  [0294] As is clear from Fig. 3, it was confirmed that the stem cells established using the single feeder cell of the present invention normally retained pluripotency.
[0295] (実施例 8:複数の細胞での再現性) (Example 8: Reproducibility with multiple cells)
実施例 6および 7で行った実験を、力-クイザル ES細胞、ヒト ES細胞について行い、 The experiments performed in Examples 6 and 7 were performed on force-quiz monkey ES cells and human ES cells,
MEFフィーダ一単独、 STO株フィーダ一単独、および混合フィーダ一単独で試験した Tested with MEF feeder alone, STO stock feeder alone, and mixed feeder alone
[0296] その結果を以下の表に表す。 Si 力-クイサル ES J¾ [0296] The results are shown in the following table. Si Force-Quisal ES J¾
サンブル «ί用 敫 «立» ») 効串  Samburu «ί use 敫« standing »»)
MEFtWt : 32 8 25%  MEFtWt: 32 8 25%
STO ¾ : 4 0 0%  STO ¾: 4 0 0%
«合フィーダ一: 5 4 80% 2 ヒ ト  «Mixed feeder: 5 4 80% 2 human
サンブル ¾用 HEBIS* W立数 槲立効率 HEMBIS * W cubes for Samburu
EF^Ut: N. Λ.  EF ^ Ut: N. Λ.
STOHUt: N. Λ.  STOHUt: N. Λ.
合フィーダ一: 3 100%  Combined feeder: 3 100%
[0297] この表からも明らかなように、それぞれのフィーダ一細胞を単独で用いた場合は、 2 5%以下の幹細胞榭立効率しか達成できな力つたのに対して、混合フィーダ一細胞 では、 80%以上で、場合によっては 100%の幹細胞榭立効率を達成することができ るという優れた効果が実証された。ヒト ES細胞は、 MEF単独でも STO単独でも従来 ほとんど榭立することができな力つたことから、本発明の効率は驚くべきものと考えら れる。 [0297] As is clear from this table, when each feeder cell was used alone, only a stem cell establishment efficiency of 25% or less was achieved, whereas in the mixed feeder single cell, An excellent effect was demonstrated in which a stem cell establishment efficiency of 80% or more, and in some cases, 100% stem cell establishment efficiency could be achieved. The efficiency of the present invention is considered to be surprising, since human ES cells have been hardly established by MEF or STO alone.
[0298] (実施例 9:神経細胞への分化)  (Example 9: Differentiation into nerve cells)
本発明の上記実施例において調製した上記 ES細胞を、 3回無血清培地にて洗浄 し、血清を完全に取り除いた。 KSR (Knockout Serum Replacement; GIBCO /Invitrogen Cat. No.10828— 028)を含む ES細胞培地を用いて、 PA6フィー ダー細胞上で未分化細胞を 8—11日間培養した(Kawasaki et al. , 2000, Neur on 28; 31— 40、Tada et al. , 2003; Dev. Dyn. , 227; 504—510.参照 The ES cells prepared in the above Examples of the present invention were washed three times with a serum-free medium to completely remove serum. Undifferentiated cells were cultured on PA6 feeder cells for 8 to 11 days using ES cell culture medium containing KSR (Knockout Serum Replacement; GIBCO / Invitrogen Cat. No. 10828-028) (Kawasaki et al., 2000, Neur on 28; 31-40; see Tada et al., 2003; Dev. Dyn., 227; 504-510.
)oこれにより、神経細胞に分化させた。 ) o This differentiated into neurons.
[0299] その結果を図 4に示す。  [0299] The results are shown in Figure 4.
[0300] 図 4の結果力 も明らかなように、本発明の方法で榭立した ES細胞は、神経への分 化能を保持していたことが明らかになった。  [0300] As is clear from the results in Fig. 4, it was revealed that the ES cells established by the method of the present invention retained the ability to differentiate into nerves.
[0301] (実施例 10:テラトーマ形成) (Example 10: Teratoma formation)
次に、本発明の ES細胞のテラトーマ形成能を確認した。 SCIDマウス(日本クレア、 東京)の皮下および腹腔内に本発明の ES細胞を約 107個移植し、 1一 3ヶ月様子を 見ノ ο Next, the teratoma-forming ability of the ES cells of the present invention was confirmed. SCID mouse (Clear Japan, Approximately 10 7 ES cells of the present invention were transplanted subcutaneously and intraperitoneally in (Tokyo) and observed for 13 months.
[0302] 1一 3力月後に得られるテラトーマを回収し、組織学的解析を行った。その一例を図 [0302] Teratoma obtained after one to three months was collected and histologically analyzed. Figure of an example
5に示す。図 5から明らかなように、本発明の ES細胞は、外胚葉、内胚葉、中胚葉な どに由来する種々の組織が観察された。したがって、本発明で榭立された ES細胞はSee Figure 5. As apparent from FIG. 5, various tissues derived from the ectoderm, endoderm, mesoderm and the like were observed in the ES cells of the present invention. Therefore, the ES cells established in the present invention
、多分ィ匕能を保持していることが実証された。 Probably, it was proved that it had retained the ability.
[0303] (実施例 11 :造血系細胞への分化) (Example 11: Differentiation into hematopoietic cells)
次に、本発明の ES細胞の造血系細胞への分化を観察する。分化誘導培養液とし て、 a MEM (Gibco BRL、 Cat # 11900— 016)に 10%ゥシ胎仔血清を加え、 5 X Next, differentiation of the ES cells of the present invention into hematopoietic cells is observed. Add 10% fetal calf serum to MEM (Gibco BRL, Cat # 11900-016) as a
10— 5M メルカプトエタノールを加えた培養液を使用した。 Using broth plus 10- 5 M mercaptoethanol.
[0304] まず、 ES細胞は、中胚葉細胞へと分化させた。未分化 ES細胞を、上述の分化誘 導培養液で懸濁し、 IV型コラーゲンコート 6プレートに 1 X 104Zゥエルの密度で播種 する。これを 37°C、 5% COで 4日間培養する。 [0304] First, ES cells were differentiated into mesodermal cells. Undifferentiated ES cells are suspended in the above-mentioned differentiation-inducing culture medium, and seeded on a type IV collagen-coated 6 plate at a density of 1 × 10 4 Z-well. Incubate this at 37 ° C, 5% CO for 4 days.
2  2
[0305] 培養上清を回収し、ゥエルを PBS (—)で一回洗う。次に細胞剥離液 (Cell Dissoci ation Buffer (Gibco BRL, # 13150— 016)を入れて 10分間静置(37°C、 5%C O )し、細胞を剥離して回収する。細胞塊をほぐし、ほぐれないものはメッシュで除き、 [0305] The culture supernatant is collected, and the wells are washed once with PBS (-). Next, add a cell dissociation solution (Cell Dissociation Buffer (Gibco BRL, # 13150-016) and let stand for 10 minutes (37 ° C, 5% CO 2) to detach and collect the cells. If you can't loose it, remove it with a mesh,
2 2
細胞数を計数する。 1, 200rpmで 5分間遠心分離し、上清を除いた。  Count the number of cells. After centrifugation at 1,200 rpm for 5 minutes, the supernatant was removed.
[0306] 次に、 10mlの HBSSZBSAで細胞を懸濁し、 1, 200rpmで 5分間遠心分離して 上清を除いた。 1 X 107細胞あたり 0. 1mlの正常マウス血清に懸濁し、 10分間静置 した。適切な量の E—力ドヘリン抗体および抗 FLK1抗体をカ卩ぇ 20分間氷水上に配 置した。 HBSSZBSAで 2回細胞を洗浄した後、 FLK1 +E—力ドヘリン一中胚葉細 胞を F ACSによりソートする。 Next, the cells were suspended in 10 ml of HBSSZBSA, and centrifuged at 1,200 rpm for 5 minutes to remove the supernatant. The cells were suspended in 0.1 ml of normal mouse serum per 1 × 10 7 cells and allowed to stand for 10 minutes. Appropriate amounts of E-force doherin antibody and anti-FLK1 antibody were placed on ice water for 20 minutes. After washing the cells twice with HBSSZBSA, the FLK1 + E-forcedherin monomesoderm cells are sorted by FACS.
[0307] ここで選択した中胚葉細胞を、分化誘導用培養液で懸濁し、 IV型コラーゲンコート プレートに 3 X 105の密度で播種する。培養液は 1ゥエルあたり 3mlカ卩え、 37°C、 5%[0307] The mesodermal cells selected here are suspended in a culture medium for inducing differentiation, and seeded on a type IV collagen-coated plate at a density of 3 x 10 5 . The culture solution is 3ml per liter, 37 ° C, 5%
COで 3日間培養する。 Incubate for 3 days in CO.
2  2
[0308] 細胞の回収およびブロッキングを行う。回収およびブロッキングは、上述のように行 う。適切な量の VE—力ドヘリン抗体をカ卩え、 20分間氷温に静置する。細胞を HBSS ZBSAで洗浄した後、 VE—力ドヘリン +血管内皮細胞を FACSによりソートして血管 内皮細胞を調製する。 [0308] The cells are collected and blocked. Recovery and blocking are performed as described above. Add an appropriate amount of VE-doherin antibody and allow to stand at ice temperature for 20 minutes. After washing cells with HBSS ZBSA, VE-forcedherin + vascular endothelial cells were sorted by FACS Prepare endothelial cells.
[0309] このようにして、本発明の ES細胞が造血系細胞へも分ィ匕することが実証される。  [0309] In this way, it is demonstrated that the ES cells of the present invention are also divided into hematopoietic cells.
[0310] (実施例 12 :心筋への分化誘導) (Example 12: Induction of differentiation into myocardium)
上述の実施例で調製される ES細胞を、 KSRを含む基本培地 (例えば、 DMEM) 中で浮遊培養することによって胚様体を形成させる。胚様体形成後、 4一 7日経過後 に胚様体を集めゼラチンコート下培養皿に播種する。 KSRを含む基本培地中で 1一 Embryoid bodies are formed by suspending and culturing the ES cells prepared in the above Examples in a basic medium (eg, DMEM) containing KSR. Four to seven days after the formation of embryoid bodies, the embryoid bodies are collected and seeded on a culture dish under gelatin coating. 1 in a basic medium containing KSR
3週間培養することによって心筋細胞が分ィ匕する。 By culturing for 3 weeks, the cardiomyocytes are divided.
[0311] (実施例 13:他の幹細胞 (例えば組織幹細胞)での実験) (Example 13: Experiment with other stem cells (eg, tissue stem cells))
組織幹細胞 (神経幹細胞、間葉系幹細胞など)を含む組織を取り出し、混合フィー ダー細胞上で培養する。増殖する幹細胞の集団を分離し、本発明上記実施例 5のよ うに調製されるフィーダ一細胞を用いて培養することによって安定して継代維持する ことができる細胞株が樹立される。  Remove tissue containing tissue stem cells (neural stem cells, mesenchymal stem cells, etc.) and culture on mixed feeder cells. By isolating the proliferating stem cell population and culturing it using the feeder cell prepared as described in Example 5 of the present invention, a cell line that can be stably maintained and maintained is established.
[0312] (実施例 14 :サルでの再生治療) (Example 14: Regenerative treatment in monkeys)
次に、力-クイサルから、 ES細胞を採取し、実施例 9と同様の処置を行い、神経細 胞に分ィ匕させる。この神経細胞の移植物を神経疾患を有する力-クイサル被検体に 移植すると、移植物が機能できることがわかる。  Next, ES cells are collected from the force-quisar, and the same treatment as in Example 9 is performed to divert the cells to nerve cells. Transplanting this transplant of nerve cells into a force-quisar subject with neurological disease shows that the transplant can function.
[0313] (実施例 15 :ヒトでの再生治療) (Example 15: Regenerative treatment in human)
次に、ヒトから、実施例 6に記載されるように、同意を得た患者力も ES細胞を採取し Next, ES cells were collected from humans with consent from the patient, as described in Example 6.
、実施例 14に記載するように実施例 9と同様の手順に基づいて処理する。この神経 細胞の移植物を、神経疾患を有するヒト被検体に予め同意を得た後に移植すると、 移植物が機能できることがわかる。 The processing is performed according to the same procedure as in Example 9 as described in Example 14. It can be seen that transplantation of this transplant of nerve cells into a human subject having a neurological disease after obtaining consent in advance has enabled the transplant to function.
[0314] (実施例 16 :フィーダ一効果の検討) (Example 16: Examination of feeder-one effect)
次に、実施例 1で調製した初代培養細胞と、実施例 3— 4で調製した細胞株とを適 宜混合して、フィーダ一細胞調製物を調製し、フィーダ一効果に差異が現れるかどう か検証する。  Next, the primary cultured cells prepared in Example 1 and the cell line prepared in Example 3-4 were appropriately mixed to prepare a feeder-cell preparation, and whether or not a difference in the feeder effect was exhibited. Verify that
[0315] 使用する比率を以下に示す。 初代^養細胞 (%) 細胞株 (%) [0315] The ratios used are shown below. Primary ^ cultured cells (%) Cell line (%)
1 00 0  1 00 0
95 5  95 5
90 10  90 10
80 20  80 20
75 2 δ  75 2 δ
70 0  70 0
60 40  60 40
50 50  50 50
40 60  40 60
30 70  30 70
25 7 δ  25 7 δ
20 80  20 80
1 0 90  1 0 90
5 9 δ  5 9 δ
0 100  0 100
[0316] 上記条件のうち、 10 :90— 90: 10まででは、フィーダ一細胞として単独で用いたと きよりも改善された効果が示される。 5 :95および 95 :5では、単独使用と同じ程度の 効果が観察される。特に、 20 :80— 80 :20では、混入する分化細胞が 30%未満程 度に低減され、ほぼ等量で観察すると、混入する分化細胞が 1%程度に低減すること が観察される。 [0316] Of the above conditions, up to 10:90 to 90:10, an improved effect is shown as compared to the case of using the cells alone as a feeder cell. At 5:95 and 95: 5, the same effect is observed as when used alone. In particular, at 20: 80-80: 20, the number of contaminating differentiated cells is reduced to less than 30%, and when observed in almost the same amount, the amount of contaminating differentiated cells is reduced to about 1%.
[0317] 従って、好ましい比率としては、例えば、 1:4一 4:1、あるいは 1:3— 3:1を使用す ることが有利であり、より好ましくは、ほぼ等量 (例えば、 3:7— 7:3、 6:4— 4:6など) を使用することが有利であり得る。  [0317] Therefore, it is advantageous to use, for example, a preferable ratio of 1: 4 to 4: 1, or 1: 3 to 3: 1, and more preferably, an approximately equivalent amount (for example, 3: 7—7: 3, 6: 4—4: 6, etc.).
[0318] (実施例 17:他の幹細胞での検討)  (Example 17: Examination with other stem cells)
次に、実施例 16と同様の実験を、幹細胞として力-クイサルを用いて行う。  Next, an experiment similar to that in Example 16 is performed using force-quisar as a stem cell.
[0319] プロトコ一ノレは、実施例 16に準じる。  [0319] The protocol is the same as in Example 16.
[0320] その結果、実施例 16と同様に、 10 :90— 90: 10まででは、フィーダ一細胞として単 独で用いたときよりも改善された効果が示される。 5 :95および 95 :5では、単独使用 と同じ程度の効果が観察される。特に、 20 :80— 80 :20では、混入する分化細胞が 30%未満程度に低減され、ほぼ等量で観察すると、混入する分化細胞が 1%程度に 低減することが観察される。 [0320] As a result, as in Example 16, the effect improved from 10:90 to 90:10 as compared to the case where the feeder cell was used alone was shown. At 5:95 and 95: 5, the same degree of effect is observed as when used alone. In particular, at 20: 80—80: 20, contaminating differentiated cells When observed at about the same amount, it is observed that the number of mixed differentiated cells is reduced to about 1%.
[0321] 従って、好ましい比率としては、例えば、 1:4一 4:1、あるいは 1:3— 3:1を使用す ることが有利であり、より好ましくは、ほぼ等量 (例えば、 3:7— 7:3、 6:4— 4:6など) を使用することが有利であり得る。 [0321] Therefore, as a preferable ratio, for example, it is advantageous to use 1: 4 to 4: 1, or 1: 3 to 3: 1, and more preferably, approximately equal amounts (eg, 3: 7—7: 3, 6: 4—4: 6, etc.).
[0322] (実施例 18:ヒト細胞での検証) (Example 18: Verification in human cells)
次に、実施例 16と同様の実験を、幹細胞としてヒトの細胞を用いて行う。  Next, an experiment similar to that in Example 16 is performed using human cells as stem cells.
[0323] プロトコールは、実施例 16に準じる。ただし、ヒトの ES細胞は、上記のように、提供 者の同意の下で行う。 The protocol is according to Example 16. However, human ES cells are used with the consent of the provider as described above.
[0324] その結果、実施例 16と同様に、 10 :90— 90: 10まででは、フィーダ一細胞として単 独で用いたときよりも改善された効果が示される。 5 :95および 95 :5では、単独使用 と同じ程度の効果が観察される。特に、 20 :80— 80 :20では、混入する分化細胞が 30%未満程度に低減され、ほぼ等量で観察すると、混入する分化細胞が 1%程度に 低減することが観察される。  [0324] As a result, as in Example 16, up to 10:90-90:10, an improved effect was obtained as compared to the case where the single feeder cell was used alone. At 5:95 and 95: 5, the same degree of effect is observed as when used alone. In particular, at 20: 80-80: 20, the amount of contaminated differentiated cells is reduced to less than about 30%, and when observed in almost equal amounts, the amount of contaminated differentiated cells is reduced to about 1%.
[0325] 従って、好ましい比率としては、例えば、 1:4一 4:1、あるいは 1:3— 3:1を使用す ることが有利であり、より好ましくは、ほぼ等量 (例えば、 3:7— 7:3、 6:4— 4:6など) を使用することが有利であり得る。  [0325] Therefore, as a preferable ratio, for example, it is advantageous to use 1: 4 to 4: 1, or 1: 3 to 3: 1, and more preferably, to an approximately equivalent amount (for example, 3: 3). 7—7: 3, 6: 4—4: 6, etc.).
[0326] (実施例 19:他のフィーダ一細胞での検証)  (Example 19: Verification with another feeder cell)
次に、実施例 16と同様の実験を、幹細胞としてヒトの細胞を用いて行い、フィーダ 一細胞として、初代培養細胞として、マウス繊維芽初代培養細胞およびヒト表皮繊維 芽細胞(Clonetics, a division of BioWhittakerゝ USA ; Cambrex社、 US A )を用い、細胞株として、 SL10および MRC5(ATCC number CCL— 171)を用 いて行う。  Next, the same experiment as in Example 16 was performed using human cells as stem cells, and as primary feeder cells, mouse fibroblast primary cultured cells and human epidermal fibroblasts (Clonetics, a division of BioWhittaker® USA; Cambrex, USA) using SL10 and MRC5 (ATCC number CCL-171) as cell lines.
[0327] プロトコールは、実施例 16に準じる。ただし、ヒトの ES細胞は、上記のように、提供 者の同意の下で行う。  [0327] The protocol is according to Example 16. However, human ES cells are used with the consent of the provider as described above.
[0328] その結果、実施例 16と同様に、 10 :90— 90: 10まででは、フィーダ一細胞として単 独で用いたときよりも改善された効果が示される。 5 :95および 95 :5では、単独使用 と同じ程度の効果が観察される。特に、 20 :80— 80 :20では、混入する分化細胞が 30%未満程度に低減され、ほぼ等量で観察すると、混入する分化細胞が 1%程度に 低減することが観察される。 [0328] As a result, as in Example 16, up to 10:90-90:10, an improved effect was exhibited as compared to the case where the single feeder cell was used alone. At 5:95 and 95: 5, the same degree of effect is observed as when used alone. In particular, at 20: 80—80: 20, contaminating differentiated cells When observed at about the same amount, it is observed that the number of mixed differentiated cells is reduced to about 1%.
[0329] 従って、好ましい比率としては、例えば、 1 :4一 4 : 1、あるいは 1 : 3— 3 : 1を使用す ることが有利であり、より好ましくは、ほぼ等量 (例えば、 3 : 7— 7 : 3、 6 :4— 4 : 6など) を使用することが有利であり得る。  [0329] Therefore, as a preferable ratio, for example, it is advantageous to use 1: 4 to 4: 1, or 1: 3 to 3: 1, and more preferably, approximately equal amounts (for example, 3: 1: 7—7: 3, 6: 4—4: 6, etc.) may be advantageous.
[0330] 従って、フィーダ一細胞として使用される他の正常細胞および他の細胞株を用いて も、同様に混合した場合のフィーダ一効果が顕著に増強されることが認められる。  [0330] Accordingly, it can be seen that the feeder effect when mixed as well with other normal cells and other cell lines used as feeder cells is significantly enhanced.
[0331] (実施例 20:フィーダ一細胞調製物のパッケージング)  (Example 20: Packaging of feeder-cell preparation)
実施例 5などで調製したフィーダ一細胞調製物が販売可能なルートでも活性を保 持することができるかどうかを以下に確認する。  It is confirmed below whether the feeder-cell preparation prepared in Example 5 or the like can maintain the activity even through a marketable route.
[0332] フィーダ一細胞として使用する調製物にマイトマイシン C処理を行う。マイトマイシン Cは、 Sigmaから入手可能である。マイトマイシン C処理をすることによって、フィーダ 一細胞が増殖しなくなり、長時間の保存が可能で、販売することが可能となる。このよ うなマイトマイシン C処理した細胞は、凍結保存して販売することができる。このような 凍結保存は、従来の初代培養細胞で行われている方法と同じものを利用でき、例え ば、そのような方法は、「ES細胞の遺伝子操作」分子生物学プロトコール (南江堂) p 471— 479に記載されている(このほかに、非特許文献 1を参照。 ) o  [0332] The preparation to be used as a feeder cell is treated with mitomycin C. Mitomycin C is available from Sigma. By treating with mitomycin C, feeder cells do not proliferate, can be stored for a long time, and can be sold. Such mitomycin C-treated cells can be cryopreserved and sold. For such cryopreservation, the same method as used in conventional primary culture cells can be used. For example, such a method is described in “Genetic Manipulation of ES Cells” Molecular Biology Protocol (Nan-Edo) p 471 — It is described in 479 (In addition, refer to Non-Patent Document 1.) o
[0333] 具体的には以下のように処理することが可能である。  [0333] Specifically, the processing can be performed as follows.
[0334] 本実施例において調製されるフィーダ一細胞の販売時のパッケージは、例えば、 凍結チューブの形態で提供する。その処理は以下の通りである。  [0334] The package at the time of sale of the feeder cells prepared in this example is provided, for example, in the form of a cryotube. The processing is as follows.
[0335] マイトマイシン C処理までは通常の調製時 (実施例 2および 3を参照)と同様で、その 後、以下の操作を行う。  [0335] Up to the treatment with mitomycin C, the procedure is the same as that for normal preparation (see Examples 2 and 3), and then the following operation is performed.
[0336] マイトマイシン C処理した培養皿から培地を除き PBSで 3回洗 、、培地をカ卩え、数時 間から一晩培養する。  [0336] The medium is removed from the culture dish treated with mitomycin C, washed three times with PBS, the medium is cultivated, and cultured for several hours to overnight.
[0337] 培地を除き PBSで 3回洗い、トリプシン処理により細胞を解離し、培地を加え細胞を よく懸濁した後、遠心して上清を除く。  [0337] Remove the medium, wash three times with PBS, dissociate the cells by trypsinization, add the medium, suspend the cells well, and centrifuge to remove the supernatant.
[0338] 細胞を細胞凍結培地(90%FCS + 10%DMSO)にたとえば 5 X 106Zmlに懸濁 する。 [0339] 細胞懸濁液をクライオチューブ(NUNC 377224)〖こ lmlいれ、しつ力りとふたを する。 The cells are suspended in, for example, 5 × 10 6 Zml in a cell freezing medium (90% FCS + 10% DMSO). [0339] Add the cell suspension to a cryotube (NUNC 377224) and place the lid tightly.
[0340] 凍結チューブを細胞凍結用容器 (Nulgene 5100)へ入れ、容器を- 80°Cフリー ザ一へ入れる。  [0340] Put the freezing tube in a cell freezing container (Nulgene 5100), and put the container in a -80 ° C freezer.
[0341] 4時間から一晩したら、凍結チューブを液体窒素容器に移す。  [0341] After 4 hours to overnight, transfer the cryotube to a liquid nitrogen container.
[0342] このように調製した販売用フィーダ一細胞調製物は、好ましくは凍結状態で、販売 先で提供される。提供先では、通常の凍結細胞と同様に解凍作業を行い、フィーダ 一細胞として使用することができる。  [0342] The feeder cell preparation for sale prepared in this manner is provided to the seller, preferably in a frozen state. At the provider, the cells can be thawed in the same manner as normal frozen cells and used as a single feeder cell.
[0343] 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、この実施形態に限定して解釈されるべきものではない。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。当業者は、 本発明の具体的な好ましい実施形態の記載から、本発明の記載および技術常識に 基づいて等価な範囲を実施することができ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こ とが理解される。 As described above, the present invention which has exemplified the present invention using the preferred embodiment of the present invention should not be construed as being limited to this embodiment. It is understood that the scope of the present invention should be construed only by the appended claims. It is understood that those skilled in the art can implement an equivalent range based on the description of the present invention and common technical knowledge from the description of the specific preferred embodiments of the present invention. Patents, patent applications, and references cited herein should be incorporated by reference in their entirety, as if the content itself were specifically described herein. Is understood.
産業上の利用可能性  Industrial applicability
[0344] 本発明は、再生医療において、多大な有用性を有する。好ましい実施形態では特 に、従来達成不可能であった高い榭立効率で幹細胞 (特に、ヒトを含む霊長類の ES 細胞)を榭立することができたことから、その有用性は高い。したがって、本発明は、 再生医療およびその治療用医薬などを製造する業において利用可能性がある。 [0344] The present invention has tremendous utility in regenerative medicine. The usefulness of the preferred embodiment is particularly high, since stem cells (especially ES cells of primates including humans) can be established with a high efficiency of establishment that was previously impossible. Therefore, the present invention has applicability in the field of manufacturing regenerative medicine and pharmaceuticals for treating it.

Claims

請求の範囲 The scope of the claims
[I] 正常細胞と、細胞株とを含む、幹細胞を調製するためのフィーダ一細胞調製物。  [I] A feeder-one cell preparation for preparing a stem cell, comprising a normal cell and a cell line.
[2] 前記正常細胞は、初代培養細胞を含む、請求項 1に記載のフィーダ一細胞調製物。  [2] The feeder-one cell preparation according to claim 1, wherein the normal cells include primary cultured cells.
[3] 前記正常細胞は、不死化して ヽな 、細胞である、請求項 1に記載のフィーダ一細胞 調製物。 [3] The feeder-one cell preparation according to claim 1, wherein the normal cells are cells that have not been immortalized.
[4] 前記正常細胞は、繊維芽細胞を含む、請求項 1に記載のフィーダ一細胞調製物。  [4] The feeder-one cell preparation according to claim 1, wherein the normal cells include fibroblasts.
[5] 前記正常細胞は、繊維芽細胞初代培養細胞である、請求項 1に記載のフィーダ一細 胞調製物。 [5] The feeder-cell preparation according to claim 1, wherein the normal cells are primary cultured fibroblast cells.
[6] 前記正常細胞は、マウス由来である、請求項 1に記載のフィーダ一細胞調製物。  [6] The feeder-one cell preparation according to claim 1, wherein the normal cells are derived from a mouse.
[7] 前記正常細胞は、胎児由来である、請求項 1に記載のフィーダ一細胞調製物。 [7] The feeder-one cell preparation according to claim 1, wherein the normal cells are derived from a fetus.
[8] 前記正常細胞は、継代数が 5回以下である、請求項 1に記載のフィーダ一細胞調製 物。 [8] The feeder-one cell preparation according to claim 1, wherein the normal cells have a passage number of 5 or less.
[9] 前記正常細胞は、マウス 11一 16日齢胎児から酵素処理により得られた繊維芽細胞 である、請求項 1に記載のフィーダ一細胞調製物。  [9] The feeder-one cell preparation according to claim 1, wherein the normal cells are fibroblasts obtained by enzymatic treatment from a mouse 11-16 days old fetus.
[10] 前記正常細胞は、継代数が 5回以下の初代培養細胞である、請求項 1に記載のフィ ーダー細胞調製物。 [10] The feeder cell preparation according to claim 1, wherein the normal cell is a primary cultured cell having a passage number of 5 or less.
[II] 前記細胞株は、不死化している、請求項 1に記載のフィーダ一細胞調製物。  [II] The feeder-one cell preparation according to claim 1, wherein the cell line is immortalized.
[12] 前記細胞株は、繊維芽細胞株である、請求項 1に記載のフィーダ一細胞調製物。 [12] The feeder-one cell preparation according to claim 1, wherein the cell line is a fibroblast cell line.
[13] 前記細胞株は、継代された期間が 2ヶ月以下である、請求項 1に記載のフィーダ一細 胞調製物。 [13] The feeder-cell preparation according to claim 1, wherein the subculture period of the cell line is 2 months or less.
[14] 前記細胞株は、継代された期間が 1ヶ月以下である、請求項 1に記載のフィーダ一細 胞調製物。  [14] The feeder-cell preparation according to claim 1, wherein the cell line has been passaged for one month or less.
[15] 前記細胞株は、マウス由来である、請求項 1に記載のフィーダ一細胞調製物。  [15] The feeder-one cell preparation according to claim 1, wherein the cell line is derived from a mouse.
[16] 前記細胞株は、胎児由来である、請求項 1に記載のフィーダ一細胞調製物。  [16] The feeder-one cell preparation according to claim 1, wherein the cell line is derived from a fetus.
[17] 前記細胞株は、 STO株である、請求項 1に記載のフィーダ一細胞調製物。  [17] The feeder-one cell preparation according to claim 1, wherein the cell line is an STO line.
[18] 前記細胞株は、 neo抵抗性である、請求項 1に記載のフィーダ一細胞調製物。  [18] The feeder-cell preparation according to claim 1, wherein the cell line is neo-resistant.
[19] 前記細胞株は、 SL10株である、請求項 1に記載のフィーダ一細胞調製物。  [19] The feeder-one cell preparation according to claim 1, wherein the cell line is a SL10 line.
[20] 前記正常細胞と、前記細胞株とは、約 1: 10— 10 : 1である、請求項 1に記載のフィー ダー細胞調製物。 [20] The feed according to claim 1, wherein the normal cells and the cell line are about 1:10 to 10: 1. Dar cell preparation.
[21] 前記正常細胞と、前記細胞株とは、約 1 : 3— 3 : 1である、請求項 1に記載のフィーダ 一細胞調製物。  [21] The feeder one-cell preparation according to claim 1, wherein the normal cell and the cell line are about 1: 3-3: 1.
[22] 前記正常細胞と、前記細胞株とは、ほぼ等量で存在する、請求項 1に記載のフィー ダー細胞調製物。  [22] The feeder cell preparation according to claim 1, wherein the normal cell and the cell line are present in approximately equal amounts.
[23] 前記幹細胞は、胚性幹細胞である、請求項 1に記載のフィーダ一細胞調製物。  [23] The feeder-one cell preparation according to claim 1, wherein the stem cells are embryonic stem cells.
[24] 前記幹細胞は、ヒト胚性幹細胞である、請求項 1に記載のフィーダ一細胞調製物。 [24] The feeder-one cell preparation according to claim 1, wherein the stem cells are human embryonic stem cells.
[25] 正常細胞と、細胞株とを含むフィーダ一細胞調製物の上で、幹細胞を培養する工程 を包含する、 [25] culturing stem cells on a feeder-one cell preparation containing normal cells and a cell line,
幹細胞を調製するための方法。  A method for preparing a stem cell.
[26] 前記フィーダ一細胞調製物は、請求項 1一 24のいずれ力 1項に記載のフィーダ一細 胞調製物である、請求項 25に記載の方法。 26. The method according to claim 25, wherein the feeder-cell preparation is the feeder-cell preparation according to any one of claims 1 to 24.
[27] 前記フィーダ一細胞が配置される培養ディッシュはゼラチンコーティングされて 、る、 請求項 25に記載の方法。 [27] The method according to claim 25, wherein the culture dish in which the feeder cells are arranged is coated with gelatin.
[28] 前記フィーダ一細胞調製物は、約 1 X 104細胞 /cm2—約 1 X 105細胞 /cm2で播 種される、請求項 25に記載の方法。 [28] The method of claim 25, wherein the feeder single cell preparation is seeded at about 1 × 10 4 cells / cm 2 to about 1 × 10 5 cells / cm 2 .
[29] 前記フィーダ一細胞調製物は、混合して播種された後 5日以内に使用される、請求 項 25に記載の方法。 [29] The method according to claim 25, wherein the feeder-one cell preparation is used within 5 days after being mixed and inoculated.
[30] 前記幹細胞は、ノックアウト血清代替添加物 (KSR)を含む培地中で培養される、請 求項 25に記載の方法。  [30] The method according to claim 25, wherein the stem cells are cultured in a medium containing a knockout serum replacement additive (KSR).
[31] 前記方法は、さらに、前記幹細胞を継代する工程を包含し、該継代工程にぉ ヽて、 コラゲナーゼが使用される、請求項 25に記載の方法。 31. The method according to claim 25, wherein the method further comprises a step of subculturing the stem cell, wherein collagenase is used in the subculturing step.
[32] 請求項 25に記載の方法によって調製される、幹細胞調製物。 [32] A stem cell preparation prepared by the method according to claim 25.
[33] 請求項 25に記載の方法によって調製される、胚性幹細胞調製物。 [33] An embryonic stem cell preparation prepared by the method according to claim 25.
[34] 請求項 25に記載の方法によって調製される、霊長類胚性幹細胞調製物。 [34] A primate embryonic stem cell preparation prepared by the method according to claim 25.
[35] 請求項 25に記載の方法によって調製される、ヒト胚性幹細胞調製物。 [35] A human embryonic stem cell preparation prepared by the method according to claim 25.
[36] 臓器、組織または細胞を再生するための移植物を調製するための方法であって: [36] A method for preparing a transplant for regenerating an organ, tissue or cell, comprising:
A)所望の臓器、組織または細胞に分ィ匕し得る幹細胞を提供する工程; B)該幹細胞を、請求項 1に記載のフィーダ一細胞調製物とともに培養する工程;お よび A) a step of providing a stem cell that can be divided into a desired organ, tissue or cell; B) culturing the stem cells with the feeder-cell preparation of claim 1; and
C)該幹細胞を所望の臓器、組織または細胞を再生するための移植物へと分化さ せる工程、  C) a step of differentiating the stem cells into a transplant for regenerating a desired organ, tissue or cell,
を包含する、方法。  A method comprising:
[37] 前記所望の臓器、組織または細胞は、神経、血球、骨、軟骨、心臓、心膜、血管、筋 肉、眼、肝臓、脾臓、腸、胃、肺、気管、毛および皮膚力もなる群より選択される、請 求項 36に記載の方法。  [37] The desired organ, tissue or cell also includes nerve, blood cell, bone, cartilage, heart, pericardium, blood vessel, muscle, eye, liver, spleen, intestine, stomach, lung, trachea, hair and skin power. 37. The method of claim 36, wherein the method is selected from a group.
[38] 前記所望の臓器、組織または細胞と前記フィーダ一細胞調製物とは、同じ種である、 請求項 36に記載の方法。  38. The method of claim 36, wherein the desired organ, tissue or cell and the feeder-cell preparation are of the same species.
[39] 前記所望の臓器、組織または細胞は霊長類のものであり、前記フィーダ一細胞調製 物はマウス由来である、請求項 36に記載の方法。  [39] The method according to claim 36, wherein the desired organ, tissue or cell is a primate, and the feeder-cell preparation is derived from a mouse.
[40] 前記培養は、ェキソビボで行われる、請求項 36に記載の方法。  [40] The method according to claim 36, wherein the culturing is performed ex vivo.
[41] 前記幹細胞は、被検体力 摘出されてすぐのものである力、または凍結保存されたも のである、請求項 36に記載の方法。 [41] The method according to claim 36, wherein the stem cell is a force that is immediately obtained after the subject force has been removed, or has been cryopreserved.
[42] 前記培養は、 37°C、飽和湿度中で 5%CO下で行われる、請求項 36に記載の方法  42. The method according to claim 36, wherein the culturing is performed at 37 ° C. in a saturated humidity under 5% CO.
[43] 前記分化は、 DNA脱メチル化剤、ヒストン脱ァセチル化剤、核内レセプターリガンド 、細胞増殖因子、サイト力イン、へキサメチレンビスァセトアミド、ジメチルァセトアミド、 ジブチル cAMP、ジメチォルスルホキシド、ョードデォキシゥリジン、ヒドロキシル尿素 、シトシンァラビノシド、マイトマイシン C、酪酸ナトリウム、ァフイディコリン、フルォロデ ォキシゥリジン、ポリプレンおよびセレン力もなる群より選択される少なくとも 1つの分 化因子を含む培養液にぉ ヽて行われる、請求項 36に記載の方法。 [43] The differentiation is carried out by DNA demethylating agent, histone deacetylating agent, nuclear receptor ligand, cell growth factor, cytokin, hexamethylenebisacetamide, dimethylacetamide, dibutyl cAMP, dimethoate Including at least one factor selected from the group consisting of rusulfoxide, ododeoxyperidine, hydroxyl urea, cytosine arabinoside, mitomycin C, sodium butyrate, aphidicolin, fluorodeoxyperidine, polypropylene and selenium. 37. The method according to claim 36, wherein the method is performed on a culture solution.
[44] 請求項 36に記載の方法によって調製される、臓器、組織または細胞。  [44] An organ, tissue or cell prepared by the method of claim 36.
[45] 臓器、組織または細胞を再生するためのシステムであって: [45] A system for regenerating an organ, tissue or cell, comprising:
A)容器;および  A) container; and
B)該容器上に播種される、繊維芽細胞初代培養細胞と、繊維芽細胞株とを含むフ ィーダ一細胞調製物、 を備える、システム。 B) a feeder-one cell preparation comprising a fibroblast primary culture cell and a fibroblast cell line, seeded on the container; A system comprising:
[46] 臓器、組織または細胞を再生するための方法であって:  [46] A method for regenerating an organ, tissue or cell, comprising:
A)所望の臓器、組織または細胞に分ィ匕し得る幹細胞を提供する工程; A) a step of providing a stem cell that can be divided into a desired organ, tissue or cell;
B)該幹細胞を、請求項 1に記載されるフィーダ一細胞調製物とともに培養する工程 ;および B) culturing said stem cells with the feeder-cell preparation of claim 1; and
C)該培養された該幹細胞を被検体の処置されるべき部位に移植する工程、 を包含する、方法。  C) transplanting the cultured stem cells to a site to be treated in a subject.
[47] D)前記幹細胞を分ィ匕させる工程をさらに包含する、請求項 46に記載の方法。  [47] The method according to claim 46, further comprising: D) separating the stem cells.
[48] フィーダ一細胞調製物としての、正常細胞と、細胞株とを含む細胞調製物の使用。 [48] Use of a cell preparation containing normal cells and a cell line as a feeder-cell preparation.
[49] 前記フィーダ一細胞調製物は、増殖しな!ヽように処理されて!ヽる、請求項 48に記載 の使用。 [49] The use according to claim 48, wherein the feeder-cell preparation is treated so as not to proliferate.
[50] 前記フィーダ一細胞調製物は、マイトマイシン Cにより処理されている、請求項 48に 記載の使用。  [50] The use according to claim 48, wherein the feeder-cell preparation has been treated with mitomycin C.
[51] 幹細胞の使用が適切である疾患、障害または状態の処置または予防のための、幹細 胞を含む医薬の製造における、請求項 1に記載されるフィーダ一細胞調製物の使用  [51] Use of a feeder-cell preparation according to claim 1 in the manufacture of a medicament comprising stem cells for the treatment or prevention of a disease, disorder or condition for which use of stem cells is appropriate.
[52] 前記フィーダ一細胞調製物は、増殖しな!、ように処理されて!、る、請求項 51に記載 の使用。 [52] The use of claim 51, wherein the feeder-cell preparation is not proliferated!
[53] 前記フィーダ一細胞調製物は、マイトマイシン Cにより処理されている、請求項 51に 記載の使用。 [53] The use of claim 51, wherein the feeder-cell preparation has been treated with mitomycin C.
1/6 1/6
図 1AFigure 1A
Figure imgf000064_0001
Figure imgf000064_0001
差替 え 用 紙(規則 26) 2/6 Replacement paper (Rule 26) 2/6
図 1BFigure 1B
Figure imgf000065_0001
Figure imgf000065_0001
差替え用紙(規則 26》 Replacement Form (Rule 26)
3/6 3/6
図 2A Figure 2A
2;,『:' 5μΓΠ  2 ; 、 「: '5μΓΠ
差替 え 用 紙(規則 26) 4/6 Replacement paper (Rule 26) 4/6
< <
図 2BFigure 2B
Figure imgf000067_0001
Figure imgf000067_0001
図 3  Fig 3
趣薩Hosatsu
: :
.纖
Figure imgf000067_0002
. Fiber
Figure imgf000067_0002
ALP SSEA-4 TRA-1-60  ALP SSEA-4 TRA-1-60
差眷ぇ用 ·紙 (規則 26)
Figure imgf000068_0001
Paper (Rule 26)
Figure imgf000068_0001
6/66/6
]  ]
胚^胞 免疫手術
Figure imgf000069_0001
'ーダ一 »肐 培養
Embryo surgery
Figure imgf000069_0001
'ー daichi »肐 Culture
Figure imgf000069_0002
Figure imgf000069_0002
35 mm培猱皿  35 mm culture dish
PCT/JP2004/015854 2003-10-28 2004-10-26 Method of simply preparing stem cell and feeder cell to be used therein WO2005040361A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2005515006A JPWO2005040361A1 (en) 2003-10-28 2004-10-26 Simple preparation method of stem cells and feeder cells used therefor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2003-368162 2003-10-28
JP2003368162 2003-10-28

Publications (1)

Publication Number Publication Date
WO2005040361A1 true WO2005040361A1 (en) 2005-05-06

Family

ID=34510333

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2004/015854 WO2005040361A1 (en) 2003-10-28 2004-10-26 Method of simply preparing stem cell and feeder cell to be used therein

Country Status (2)

Country Link
JP (1) JPWO2005040361A1 (en)
WO (1) WO2005040361A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010047510A (en) * 2008-08-21 2010-03-04 Nippon Dental Univ Method for forming organ-anlage, and organ-anlage
JP5092124B2 (en) * 2005-05-24 2012-12-05 国立大学法人 熊本大学 ES cell differentiation induction method
US9289312B2 (en) 2011-07-13 2016-03-22 Vivex Biomedical, Inc. Spinal implants with stem cells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003029443A1 (en) * 2001-09-28 2003-04-10 Es Cell International Pte Ltd Methods of derivation and propagation of undifferentiated human embryonic stem (hes) cells on feeder-free matrices and human feeder layers

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003029443A1 (en) * 2001-09-28 2003-04-10 Es Cell International Pte Ltd Methods of derivation and propagation of undifferentiated human embryonic stem (hes) cells on feeder-free matrices and human feeder layers

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KAWASE E. ET AL.: "Effect of feeder cells on the establishment of ES cells from mice and rats: the difference between allogenic and xenogenic feeders", JOURNAL OF REPRODUCTION AND DEVELOPMENT, vol. 46, no. 1, 2000, pages 15 - 22, XP002994471 *
SUEMORI H. ET AL.: "Haikan saibo (ES SAibo) no iji", PROTEIN, NUCLEIC ACID AND ENZYME, vol. 36, no. 13, 1991, pages 2061 - 2063 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5092124B2 (en) * 2005-05-24 2012-12-05 国立大学法人 熊本大学 ES cell differentiation induction method
JP2010047510A (en) * 2008-08-21 2010-03-04 Nippon Dental Univ Method for forming organ-anlage, and organ-anlage
US9289312B2 (en) 2011-07-13 2016-03-22 Vivex Biomedical, Inc. Spinal implants with stem cells
US9757223B2 (en) 2011-07-13 2017-09-12 Vivex Biomedical, Inc. Spinal implants with stem cells
US9814558B2 (en) 2011-07-13 2017-11-14 Vivex Biomedical, Inc. Spinal implants with stem cells

Also Published As

Publication number Publication date
JPWO2005040361A1 (en) 2007-03-22

Similar Documents

Publication Publication Date Title
EP0701608B1 (en) Embryonic stem cells for making chimeric and transgenic ungulates
Petitte et al. Avian pluripotent stem cells
JP5456467B2 (en) Pluripotent cells from rats and other species
Sofikitis et al. Efforts to create an artificial testis: culture systems of male germ cells under biochemical conditions resembling the seminiferous tubular biochemical environment
CN109486751B (en) Method for feeder cells-free culture of bovine and porcine spermatogonial stem cells
Vaags et al. Derivation and characterization of canine embryonic stem cell lines with in vitro and in vivo differentiation potential
IL126416A (en) Cultured inner cell mass cell lines derived from ungulate embryos
Gerfen et al. Isolation of embryonic cell‐lines from porcine blastocysts
JP2002511732A (en) Compositions and methods for generating transgenic animals
US6194635B1 (en) Embryonic germ cells, method for making same, and using the cells to produce a chimeric porcine
AU721375B2 (en) Porcine embryonic stem-like cells, methods of making and using the cells to produce transgenic pigs
Wang et al. Derivation and characterization of primordial germ cells from Guangxi yellow-feather chickens
EP0774510A1 (en) Ungulate eg cell
US20070250943A1 (en) Construction of Chimera Using En Cells
Hornen et al. Production of viable pigs from fetal somatic stem cells
US20070053890A1 (en) Canine embryonic stem cells
WO2005040361A1 (en) Method of simply preparing stem cell and feeder cell to be used therein
WO2005045007A1 (en) Method and system for storing stem cell in frozen state
US20070204357A1 (en) Process for producing normal parenchymal cells, tissues or organs by bioincubator
JP2002176973A (en) Mammalian embryonic stem cell and method for establishing the same and subculture method for the same
Sarmah et al. Towards human organ generation using interspecies blastocyst complementation: Challenges and perspectives for therapy
조호연 Studies on application of spermatogonial stem cell for quail genome modification
Gurer et al. Therapeutic use of cloning: Osmangazi turk identical embryonic stem cells and embryonic stem cell transfer to diabetic mice
MXPA98008103A (en) Cellular lines of internal cellular mass cultivated derivatives of embryos ungula
KR20140043260A (en) Method for preparing avian somatic chimera by injection of bone marrow cell into fetilized egg

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005515006

Country of ref document: JP

122 Ep: pct application non-entry in european phase