WO2015098111A1 - PROCÉDÉ D'ÉTABLISSEMENT DE CELLULES iPS ET PROCÉDÉ DE MAINTENANCE À LONG TERME DE CELLULES SOUCHES - Google Patents

PROCÉDÉ D'ÉTABLISSEMENT DE CELLULES iPS ET PROCÉDÉ DE MAINTENANCE À LONG TERME DE CELLULES SOUCHES Download PDF

Info

Publication number
WO2015098111A1
WO2015098111A1 PCT/JP2014/006454 JP2014006454W WO2015098111A1 WO 2015098111 A1 WO2015098111 A1 WO 2015098111A1 JP 2014006454 W JP2014006454 W JP 2014006454W WO 2015098111 A1 WO2015098111 A1 WO 2015098111A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
serum
medium
ips
free
Prior art date
Application number
PCT/JP2014/006454
Other languages
English (en)
Japanese (ja)
Inventor
哲治 岡本
佐知子 山崎
顕 嶋本
栄俊 田原
Original Assignee
国立大学法人広島大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from JP2014087314A external-priority patent/JP6516280B2/ja
Application filed by 国立大学法人広島大学 filed Critical 国立大学法人広島大学
Publication of WO2015098111A1 publication Critical patent/WO2015098111A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/44Thiols, e.g. mercaptoethanol
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/46Amines, e.g. putrescine
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/602Sox-2
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/603Oct-3/4
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/604Klf-4
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/606Transcription factors c-Myc
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/90Polysaccharides
    • C12N2501/91Heparin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/13Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells
    • C12N2506/1346Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells
    • C12N2506/1361Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from mesenchymal stem cells from dental pulp or dental follicle stem cells

Definitions

  • the present invention relates to a method for establishing induced pluripotent stem cells (iPS cells) and a method for long-term maintenance of the undifferentiated state and differentiation pluripotency of stem cells. Specifically, the present invention relates to a method for inducing iPS cells under serum-free culture conditions without using feeder cells, and passage of stem cells by adding tumor growth factor- ⁇ (TGF- ⁇ ) to the medium. The present invention relates to a method for long-term maintenance of the undifferentiated state and differentiation pluripotency of stem cells, characterized by culturing.
  • TGF- ⁇ tumor growth factor- ⁇
  • embryonic stem cells embryonic stem cell: hereinafter referred to as ES cells
  • induced pluripotent stem cells induced pluripotent stem cell: hereinafter referred to as iPS cells
  • Stem cells are self-replicating and multipotent cells and have the ability to differentiate into various cell lineages. Since Yamanaka et al. Of Kyoto University reported the establishment of mouse iPS cells in 2006, it has attracted a great deal of attention as an alternative to ES cells that remain ethical problems using fertilized eggs.
  • iPS cells are also referred to as induced pluripotent stem cells.
  • Reprogramming genes (Oct3 / 4, Sox2, Klf4, c-Myc) are introduced into terminally differentiated cells such as skin using viruses, plasmid vectors, etc. By programming, it becomes a universal cell that can be differentiated into all cell types except for extraembryonic tissues.
  • These stem cells generally use an alternative serum-supplemented medium containing serum or animal-derived components, and increase feeder cells such as inactivated mouse embryonic fibroblasts (MEF) on the dish and support them. It has been cultured as. By the day before the passage of iPS cells, MEF cells were treated with mitomycin C or ⁇ -irradiated in advance to prepare feeder cells with suppressed proliferation (inactivated), and then seeded on gelatin-coated dishes. After sufficient adhesion and extension on the dish, iPS cells are seeded thereon and co-cultured. In addition, it is necessary to separate feeder cells and stem cells such as ES cells and iPS cells as much as possible during passage.
  • feeder cells such as ES cells and iPS cells
  • somatic differentiated cells before being induced by iPS cells are also cultured in a medium supplemented with fetal bovine serum, human serum or human autoserum.
  • somatic differentiated cells somatic cells
  • fetal bovine serum human serum or human autoserum.
  • the feeder cells to be used have a great variety, kind of treatment, and lot difference, and the state of the stem cells is considerably influenced. Therefore, it was impossible to culture the stem cells under stable conditions.
  • Stem cells cultured under conditions with many indeterminate elements contain many unknown factors in the medium, making standardization of basic research such as tissue / organ development and regeneration difficult, and maintaining undifferentiation. It is difficult to compare the functions of necessary growth factors and differentiation-inducing factors. In addition, due to the action of various components contained in the medium, it is difficult to induce differentiation into specific cells. Furthermore, considering clinical application to regenerative medicine, there are problems with safety such as contamination of unknown factors and risk factors such as pathogens. It becomes. Therefore, in order to eliminate these elements, it is necessary to standardize the culture method under the culture conditions whose composition is clear. In particular, development of a method for producing iPS cells using a serum-free medium with a clear composition has been desired.
  • Non-Patent Document 1 a tumor (transforming) growth factor (TGF) is also added to the medium.
  • TGF tumor growth factor
  • the serum-free medium used in Patent Document 1 does not contain TGF, and that used in Non-Patent Document 1 is a serum-containing medium, which is different from the present invention.
  • Stem cells cultured under conditions with many indefinite elements such as serum-containing medium contain many unknown factors in the medium, making standardization of basic research such as tissue and organ development and regeneration difficult and undifferentiated It is difficult to compare and examine the functions of growth factors and differentiation-inducing factors that are necessary for maintenance of cancer. In addition, it is difficult to induce differentiation into specific cells due to the action of various components contained in the medium. In addition, when considering clinical application to regenerative medicine, it is necessary to eliminate risk factors such as unknown factors and pathogens from the viewpoint of safety. There is. Therefore, in order to solve these problems, it is necessary to establish and standardize a culture method using a medium whose composition is clear and does not contain risk factors.
  • a specific problem to be solved by the present invention is to cultivate somatic cells before induction into iPS cells under a condition that does not include an uncertain element and a risk factor, and iPS under a condition that does not include an uncertain element and a risk factor. It was to produce cells and to maintain stem cells such as iPS cells in culture. Specifically, specific problems to be solved by the present invention include culturing somatic cells under serum-free culture conditions, producing iPS cells under serum-free culture conditions without using feeder cells, and feeders. It was to provide a culture method that can maintain the undifferentiation and differentiation pluripotency of stem cells such as iPS cells for a long period of time without using cells.
  • the inventors of the present invention have made extensive studies to solve the above-mentioned problems, a method for serum-free culture of somatic cells before induction into iPS cells, and iPS cells by culturing without using feeder cells in a serum-free medium. Established the establishment method. In particular, it has been found preferable to use fibronectin as an adhesion factor. According to the method of the present invention, the induction efficiency of human iPS cells was 10 times or more that of the conventional method (culture conditions with serum). In addition, the present inventors have identified TGF- ⁇ as a cell growth factor capable of maintaining stem cell undifferentiation and pluripotency even after long-term passage in the above culture system. Based on these matters, the present invention has been completed.
  • the present invention provides the following: (1) Production of iPS cells characterized by inducing induced pluripotent stem cells (iPS cells) by culturing reprogrammed somatic cells without using feeder cells in a serum-free medium. Method. (2) The method according to (1), wherein fibronectin is used as an adhesion factor. (3) The method according to (1) or (2), wherein the reprogramming treatment of somatic cells is performed in a serum-free medium. (4) The method according to any one of (1) to (3), wherein the somatic cells before reprogramming are cultured from a primary culture in a serum-free medium. (5) The method according to any one of (1) to (4), wherein the iPS cell is a human iPS cell.
  • a serum-free culture substrate for inducing iPS cells containing fibronectin as an adhesion factor (however, the culture substrate does not contain feeder cells).
  • An undifferentiated state of a stem cell characterized in that a stem cell is subcultured by adding a protein belonging to the tumor growth factor- ⁇ (TGF- ⁇ ) family to a serum-free medium without using a feeder cell And methods of maintaining pluripotency.
  • TGF- ⁇ tumor growth factor- ⁇
  • the iPS cells obtained by the method according to any one of (1) to (5) are subcultured without using feeder cells in a serum-free medium to which a protein belonging to the TGF- ⁇ family is added.
  • a method for maintaining the undifferentiated state and pluripotency of iPS cells (12) Serum-free culture substrate for subculture for maintaining the undifferentiated state and differentiation pluripotency of stem cells, including proteins belonging to the TGF- ⁇ family (however, the culture substrate does not include feeder cells) .
  • the culture substrate according to (12) or (13), wherein the stem cells are iPS cells.
  • a kit for producing a serum-free culture substrate for subculture for maintaining the undifferentiated state and differentiation pluripotency of stem cells comprising a serum-free medium component and a TGF- ⁇ family protein (however, The culture substrate does not contain feeder cells).
  • somatic cells are cultured using a serum-free medium with a clear component, and in the culture environment using a serum-free medium with a clear component, without using feeder cells,
  • a method for producing iPS cells by gene transfer and induction, etc., and a culture substrate material and a culture solution capable of maintaining and culturing stem cells such as ES cells and iPS cells while maintaining an undifferentiated state and differentiation pluripotency A method for culturing stem cells using the above is provided.
  • the method for producing iPS cells of the present invention it was found that iPS cells can be induced with an efficiency 10 times or more that of the conventional method using a serum-containing medium.
  • iPS cells can be induced with high efficiency in the present invention. And the undifferentiated state and differentiation pluripotency of a stem cell can be maintained over a long period of time.
  • the culture medium there are almost no unknown factors in the culture medium, and it is easy to standardize basic research such as tissue and organ development and regeneration, and the functions of growth factors and differentiation-inducing factors necessary for maintaining undifferentiation are compared. It is also easy to do.
  • the present invention since the types of components contained in the medium are relatively few and known, differentiation induction into specific cells is easy, and highly safe cells can be obtained considering clinical application to regenerative medicine. can get.
  • FIG. 1 is a microscopic image showing the morphology of TIG-3 cell-derived hiPS cells induced on various extracellular matrices using serum-free medium hESF9.
  • FIG. 1A shows the results of examining the influence of adhesion factors when inducing human iPS cells using a serum-free medium.
  • FIG. 1B shows the form of human iPS cells (hiPS cells) 2 days after seeding on each extracellular matrix using serum-free medium hESF9.
  • FIG. 1C is an ALP-stained image (36 days after infection) of hiPS cells on each extracellular matrix.
  • the scale bar in FIGS. 1A-C is 200 ⁇ m.
  • FIG. 2 is a microscopic image showing cell morphology on various extracellular matrices after induction.
  • FIG. 3 is a micrograph showing the examination results of serum-free culture conditions when virus infection is established and a graph showing virus infection efficiency. The results of transfection into PLAT-A using EGFP and infection into TIG-3 cells are shown.
  • FIG. 4 is a scheme and microscopic image showing the induction of hiPS cells using dental pulp-derived cells under serum-free culture conditions without using feeder cells.
  • FIG. 4A is a scheme showing an outline up to hiPS cell induction.
  • Day-7-7 Primary culture of dental pulp cells on type I collagen-coated dish using serum-free medium RD6F.
  • FIG. 4B is a phase contrast microscopic image of dental pulp cells in serum-free medium RD6F before introduction of reprogramming gene.
  • FIG. 4C is a phase-contrast microscope image of pulp cells that have been passaged 4 times on a type I collagen-coated dish using serum-free medium RD6F.
  • FIG. 4D is an ALP-stained image 43 days after infection.
  • FIG. 5 shows the results of analyzing the characteristics of human iPS cells derived from dental pulp-derived cells under culture conditions using serum-free medium hESF9 without using feeder cells.
  • FIG. 5A is a phase contrast image of human iPS cells maintained in serum-free medium hESF9 or hESF9T medium (serum-free medium obtained by adding TGF- ⁇ 1 to hESF9).
  • DP-A-iPS-CL1 Phase contrast image of cells maintained for 2 generations (passage 2) or 21 generations (passage 21) on fibronectin-coated dishes using serum-free medium hESF9.
  • the right side shows a phase contrast image of cells maintained for 5 generations (passage 5) on feeder cells using a serum-supplemented medium for human ES cells.
  • DP-F-iPS-CL4, -CL6, -CL16 Cells maintained in the 58s (passage 58), 59s (passage 59) or 21s (passage 21) on the fibronectin-coated dish using the serum-free medium hESF9T Phase contrast image.
  • the right side shows a phase contrast image of a cell (CL31) maintained for 19 generations (passage 19) on a feeder cell using a serum-supplemented medium for human ES cells.
  • the scale bar is 200 ⁇ m.
  • FIG. 5B shows flow cytometry using cells that have been passaged and maintained on a fibronectin-coated dish for a long time using serum-free medium hESF9 or hESF9T, and antibodies against Oct3 / 4 and SSEA-4 that are undifferentiated markers. The result of having analyzed by is shown.
  • FIG. 5C shows the results of comprehensive gene expression analysis. Cluster analysis using a microarray was performed. DP cell (dental pulp cell), DP-iPS cells (dental pulp-derived human iPS cells) maintained on a fibronectin-coated dish using serum-free medium hESF9 or hESF9T, and feeder medium using serum medium for human ES cells The maintained human iPS cells (Tic) were analyzed.
  • FIG. 6 shows the results of comprehensive gene expression analysis (scatter-plot analysis).
  • FIG. 7 shows the results of examining the effect of TGF- ⁇ 1 under serum-free culture conditions.
  • FIG. 7A shows the cell morphology when various concentrations of TGF- ⁇ 1 (0, 0.1, 1, 2, 5, 10 ng / ml) are added.
  • FIG. 7B shows the results of gene expression analysis of human iPS cells using Droplet® Digital-PCR when TGF- ⁇ 1 was added. Each expression intensity was corrected by GAPDH.
  • FIG. 8 shows the results of pluripotency analysis of human iPS cells induced and maintained under serum-free culture conditions.
  • FIG. 8A shows the results of studies on the expression of various differentiation markers.
  • FIG. 8B shows the results of examination of teratoma formation ability.
  • the scale bar is 100 ⁇ m.
  • FIG. 9 shows the results of cell proliferation ability and karyotype analysis.
  • FIG. 9A shows the results of examining the cell doubling time of human iPS cells induced under serum-free culture conditions and passaged and maintained for 21 passages with hESF9T.
  • FIG. 9B shows the result of karyotype simplified analysis of human iPS cells induced under serum-free culture conditions and maintained for 20 passages in serum-free medium hESF9T.
  • FIG. 10 shows the results of studies on universality in passage and maintenance of human iPS cells using serum-free culture conditions.
  • the scale bar is 200 ⁇ m.
  • the upper part of FIG. 10 shows a phase contrast microscopic image of human iPS cells maintained on fibronectin using hESF9T medium.
  • the lower panel shows a phase contrast microscopic image of human iPS cells maintained on fibronectin using DF8T medium. It is shown that both serum-free media maintain human ES cell-like morphology.
  • the present invention is characterized in that induced pluripotent stem cells (iPS cells) are induced by culturing somatic cells subjected to reprogramming treatment in a serum-free medium without using feeder cells.
  • iPS cells induced pluripotent stem cells
  • a method for producing iPS cells is provided.
  • an iPS cell refers to a cell that has been subjected to reprogramming by introducing a plurality of genes into a somatic cell to have differentiation pluripotency and self-replication ability.
  • iPS cells may be derived from somatic cells of any animal, and iPS cells derived from somatic cells such as humans, monkeys, mice, rats, dogs, cats, horses, pigs and the like are exemplified. However, the present invention is not limited to these.
  • Reprogramming is dedifferentiation of differentiated somatic cells to change them into undifferentiated cells, which is also called initialization. Differentiated cells become iPS cells by the reprogramming process. In general, four types of genes (Oct3 / 4, Sox2, Klf4, c-Myc (or L-Myc)), or some of these genes (for example, Nanog, Lin28, etc.) plus differentiated cells
  • the reprogramming process is performed by introducing into the above.
  • a vector is generally used for gene introduction, and for example, a retrovirus vector is used.
  • the reprogramming process is not limited to the above method, and can be performed using a method known to those skilled in the art.
  • the medium on which the reprogramming process is performed is not particularly limited, but in the present invention, the reprogramming process is preferably performed in a serum-free medium.
  • the feature of the present invention is to induce iPS cells by culturing somatic cells subjected to reprogramming treatment in a serum-free medium.
  • a serum-free medium concerns that uncertain elements, unknown factors, or risk factors exist in the medium can be eliminated, and various analyzes can be easily performed with good reproducibility.
  • a harmful factor derived from a serum-containing medium can be eliminated by using a serum-free medium, the safety of the obtained iPS cells is ensured.
  • Serum-free medium is a medium that does not contain serum.
  • a serum-free medium is a basic culture solution containing amino acids, inorganic salts, vitamins, trace elements, sugars, etc., with known hormones and protein factors such as insulin and iron-binding protein.
  • the basal medium for animal cell culture that does not contain serum include Dulbecco's modified Eagle medium (DMEM), minimum essential medium (MEM), Eagle basal medium (BME), RPMI 1640 medium, F12 medium, MCDB medium, In addition, mixed media thereof and the like are known.
  • Examples of preferable serum-free medium used in the iPS cell induction method of the present invention include hESF9 medium, hESF9T medium, RD8F medium, DME / F12-8F medium, RDF8F medium, and modified media thereof.
  • the serum-free medium used in the present invention is not limited to these media, and those skilled in the art can appropriately select and use known serum-free media, or can modify and use known serum-free media as appropriate.
  • fibronectin is used as an adhesion factor in the induction of iPS cells in a serum-free medium without feeder cells.
  • fibronectin as an adhesion factor, the induction efficiency of iPS cells can be dramatically increased, and iPS cell induction efficiency compared to the case of inducing iPS cells by a conventional method (in a serum-containing medium, using feeder cells). Rises about 10 times.
  • Fibronectin is a protein known to those skilled in the art. Various biological sources of fibronectin are known and can be used in the present invention.
  • the fibronectin used in the present invention may be isolated from cells or may be produced by a genetic engineering method such as a recombinant method.
  • the fibronectin used in the present invention may be full length or a fragment thereof.
  • the somatic cells that have undergone the reprogramming treatment are seeded in a culture vessel coated with fibronectin, and cultured to induce iPS cells.
  • reprogramming is performed in a medium other than a serum-free medium
  • somatic cells are seeded in a culture vessel coated with fibronectin together with the medium, and then cultured while exchanging the medium with the serum-free medium.
  • somatic cells are seeded in a culture vessel coated with fibronectin together with the serum-free medium, and then cultured while changing the medium to the same or different serum-free medium. Cells can be induced.
  • the culture temperature can be 30 to 40 ° C., preferably about 37 ° C., and for example, the culture can be performed in an incubator filled with air containing 5 to 10% carbon dioxide.
  • a method for coating fibronectin on a culture vessel is known to those skilled in the art.
  • the surface of the culture container is treated with poly-L-ornithine or poly-L-lysine, washed, and then the surface of the culture container is treated with a buffer containing fibronectin to coat fibronectin on the culture container. can do.
  • a person skilled in the art can easily determine the amount of fibronectin to be used.
  • an adhesion factor other than fibronectin may be used in combination with fibronectin.
  • Methods for coating culture vessels with fibronectin are known.
  • An example of a preferable coating method is as follows.
  • the culture vessel is coated with fibronectin at a concentration of 2 ⁇ g / cm 2 and allowed to stand at 37 ° C. for 3 hours or longer and overnight. If not used immediately, seal with parafilm to prevent the coating solution from drying, store at 4 ° C., and use within 1 week.
  • the fibronetin solution is aspirated and washed once with PBS before use.
  • subculture of somatic cells to be reprogrammed may be performed from primary culture in a serum-free medium, and somatic cell reprogramming may be performed in a serum-free medium.
  • Such embodiments are described in the Examples herein by conducting subculture of somatic cells from primary culture.
  • Primary culture and subculture of somatic cells can be performed by those skilled in the art by appropriately selecting the medium composition and culture conditions according to the type of somatic cells.
  • the present invention provides a serum-free culture substrate for inducing iPS cells, which contains fibronectin as an adhesion factor.
  • the culture substrate of this aspect of the invention includes a serum-free medium and fibronectin. Therefore, one specific example of the culture substrate of this aspect of the present invention is a serum-free medium containing fibronectin. Further embodiments of the culture substrate of this aspect of the invention may include a culture vessel coated with fibronectin and a serum-free medium contained therein. However, the culture substrate does not contain feeder cells.
  • the shape of the culture container includes a dish, a bottle, a tube, a flask, a bag and the like, and is not particularly limited.
  • the present invention provides a kit for producing a serum-free culture substrate for inducing iPS cells, comprising a serum-free medium component and fibronectin as essential components.
  • the culture substrate does not contain feeder cells.
  • the serum-free medium examples include those exemplified above and those obtained by modifying them.
  • the components are also known or can be appropriately selected by those skilled in the art.
  • the form of the serum-free medium component contained in the kit is not particularly limited. For example, it may be a liquid that can be transferred to a culture vessel as it is, may be in the form of a concentrate, or may be a solid such as a powder.
  • the serum-free medium components included in the kit may be included in the kit, for example, divided into vitamins, minerals, amino acids, saccharides and the like.
  • fibronectin contained in the kit of the present invention is not particularly limited.
  • the fibronectin may be a solid such as a powder or a liquid such as an aqueous fibronectin solution containing an appropriate buffer.
  • a culture vessel may be included in the kit of the present invention.
  • the culture vessel may be made of any material such as glass or plastic. There are no particular limitations on the shape of the plate, bottle, tube, flask, bag, or the like.
  • the culture container contained in the kit of the present invention may be coated with fibronectin in advance.
  • an instruction manual is attached to the kit.
  • the stem cell is subcultured by adding a tumor (transforming) growth factor- ⁇ (TGF- ⁇ ) superfamily protein to a serum-free medium without using feeder cells.
  • TGF- ⁇ tumor growth factor- ⁇
  • a method for maintaining the undifferentiated state and differentiation pluripotency of stem cells is provided.
  • the cells by adding a protein belonging to the TGF- ⁇ superfamily, particularly a protein belonging to the TGF- ⁇ family, to a serum-free medium, the cells can be passaged over a long period of time while maintaining the undifferentiated state and differentiation pluripotency of stem cells. It becomes possible to culture.
  • Proteins belonging to the TGF- ⁇ superfamily include TGF- ⁇ family, activin family, and bone morphogenetic protein (BMP) family proteins.
  • Preferred in the present invention are proteins belonging to the TGF- ⁇ family. Proteins belonging to the TGF- ⁇ family are known to those skilled in the art, and TGF- ⁇ forms a family with activin and BMP and is currently composed of 33 types of family molecules in humans, including TGF- ⁇ 1, Activin- A, BMP-2 and the like are exemplified.
  • a particularly preferred protein belonging to TGF- ⁇ used in the above method of the present invention is TGF- ⁇ 1.
  • the concentration of the TGF- ⁇ family protein in the serum-free medium is usually 1 ng / ml to 10 ng / ml, preferably 1 ng / ml to 5 ng / ml, but may be changed as appropriate. Can do.
  • the TGF- ⁇ family protein used may be one type or two or more types.
  • TGF- ⁇ family proteins are known to those skilled in the art. Various biological TGF- ⁇ family proteins are known and can be used in the present invention.
  • the TGF- ⁇ family protein used in the present invention may be isolated from cells or may be produced by a genetic engineering method such as a recombinant method. Further, the TGF- ⁇ family protein used in the present invention may be a full-length protein or a fragment thereof.
  • Preferred serum-free media used in the method for maintaining the undifferentiated state and differentiation pluripotency of the stem cells of the present invention include media such as hESF9 media, hESF9T media, RD8F media, DME / F12-8F media, RDF8F media, ESF7 media, etc. As well as their modified media.
  • fibronectin as an adhesion factor.
  • the method for coating fibronectin on the culture container and other explanations regarding fibronectin are as described above.
  • the stem cell to which the above-described method of the present invention can be applied is not particularly limited, and includes all stem cells including those exemplified below.
  • Stem cells are cells that have the ability to differentiate into cells of multiple lineages (differentiation pluripotency) and the ability to maintain differentiation pluripotency even after cell division (self-renewal ability).
  • Stem cells include embryonic stem cells (ES cells) produced from fertilized eggs, somatic stem cells present in in vivo tissues, and induced pluripotent stem cells (iPS cells) created by introducing a specific gene.
  • ES cells and iPS cells have the property of being able to differentiate into all types of cells (totipotency).
  • somatic stem cells include hematopoietic stem cells, neural stem cells, hepatic stem cells, skin stem cells, and reproductive stem cells.
  • the stem cells may be derived from any animal, and examples thereof include, but are not limited to, stem cells derived from humans, monkeys, mice, rats, dogs, cats, horses, pigs, and the like.
  • the methods for producing and obtaining these stem cells are known to those skilled in the art, and some are stored in research institutions and others are commercially available.
  • ES cells can be established by taking an inner cell mass from a blastocyst of a fertilized egg of a subject animal and culturing it on feeder cells such as fibroblasts. The method for producing iPS cells is as described above.
  • Subculture refers to the growth and maintenance of transferred cultured cells by transferring them to a new culture vessel.
  • Methods for subculturing stem cells are known to those skilled in the art.
  • a part of the cells can be detached from the culture vessel using a digestive enzyme such as trypsin, and subculture can be performed by culturing using a new culture vessel.
  • Selection of medium components and other culture conditions, timing of medium replacement, stripping conditions, culture time per passage, etc., techniques and techniques for subculture are appropriately determined by those skilled in the art according to the cells to be subcultured, You can choose.
  • the above method of the present invention is characterized in that stem cells are subcultured by adding TGF- ⁇ family protein in a serum-free medium without using feeder cells. Preparation and selection of a serum-free medium is easy for those skilled in the art. According to the method of the present invention, by adding a TGF- ⁇ family protein to the medium, even when many passages are repeated over a long period of time, the undifferentiated state and the differentiation level of the stem cells are stably maintained. Performance can be maintained.
  • Confirmation of the undifferentiated state of stem cells can be performed by examining the expression of undifferentiated markers such as Oct3 / 4, Nanog, Sox2, and SSEA-4 in addition to morphological observation.
  • the method for confirming the undifferentiated state of stem cells is not limited to the above method, and can be performed by methods known to those skilled in the art.
  • Confirmation of the differentiation pluripotency of stem cells can be performed by forming embryonic rods from passaged and maintained stem cells, inducing differentiation, and examining the expression of various differentiation markers.
  • the method for confirming the pluripotency of stem cells is not limited to the above method, and can be performed by methods known to those skilled in the art.
  • iPS cells obtained by the method for producing iPS cells of the present invention are passaged without using feeder cells in a serum-free medium supplemented with a protein belonging to the TGF- ⁇ family. By culturing, the undifferentiated state and pluripotency of iPS cells can be maintained.
  • the induction efficiency may decrease. It is preferred not to use family proteins.
  • the present invention provides a serum-free culture substrate for subculture for maintaining the undifferentiated state and differentiation pluripotency of stem cells, which comprises a TGF- ⁇ family protein.
  • the culture substrate of this aspect of the invention includes a serum-free medium containing a TGF- ⁇ family protein.
  • a serum-free medium containing a TGF- ⁇ family protein may be provided as a liquid that can be used as it is by adding it to a culture vessel, or may be provided as a solid such as a concentrate or powder that can be prepared at the time of use. .
  • the culture substrate of this aspect of the invention may include a culture vessel and a serum-free medium containing the TGF- ⁇ family protein contained therein.
  • the kind of TGF- ⁇ family protein and the concentration in the serum-free medium are as described above.
  • the shape of the culture container includes a dish, a bottle, a tube, a flask, a bag and the like, and is not particularly limited.
  • the culture vessel is coated with fibronectin.
  • the culture substrate does not contain feeder cells.
  • the present invention provides a serum-free culture substrate for subculture for maintaining the undifferentiated state and differentiation pluripotency of stem cells, comprising a serum-free medium component and a TGF- ⁇ family protein.
  • a serum-free culture substrate for subculture for maintaining the undifferentiated state and differentiation pluripotency of stem cells, comprising a serum-free medium component and a TGF- ⁇ family protein.
  • Providing a kit for The concentration of the TGF- ⁇ family protein contained in the culture medium in the culture substrate is as described above. However, the culture substrate does not contain feeder cells.
  • the serum-free medium examples include those exemplified above and those obtained by modifying them.
  • the components are also known or can be appropriately selected by those skilled in the art.
  • the form of the serum-free medium component contained in the kit is not particularly limited. For example, it may be a liquid that can be transferred to a culture vessel as it is, may be in the form of a concentrate, or may be a solid such as a powder.
  • the serum-free medium components included in the kit may be included in the kit, for example, divided into vitamins, minerals, amino acids, saccharides and the like.
  • fibronectin is included as an adhesion factor.
  • the form of fibronectin contained in the kit of the present invention is not particularly limited.
  • the fibronectin may be a solid such as a powder or a liquid such as an aqueous fibronectin solution containing an appropriate buffer.
  • a culture vessel may be included in the kit of the present invention.
  • the culture vessel may be made of any material such as glass or plastic. There are no particular limitations on the shape of the plate, bottle, tube, flask, bag, or the like.
  • the culture container contained in the kit of the present invention may be coated with fibronectin in advance.
  • an instruction manual is attached to the kit.
  • Serum-free medium hESF9 (Furue MK, Na J, Okamoto T, et al. (2008) Heparin promotes the growth of human embryonic stem cells in a defined serum-free medium. Proc Natl Acad Sci USA -13414), each process until induction of iPS cells was examined under serum-free culture conditions. Furthermore, using human dental pulp-derived cells, we attempted to establish and maintain hiPS by performing the entire process from primary culture to viral infection and iPS cell establishment in a complete serum-free culture system.
  • Cell culture method 1-1 Cell culture solution 1) Serum-free medium The serum-free medium used contains hESF-GRO medium (Nipro) (L-ascorbic acid-2-phosphate (100 ⁇ g / ml)) developed for human ES cells.
  • hESF-GRO medium Nipro
  • Lipro L-ascorbic acid-2-phosphate (100 ⁇ g / ml)
  • the serum-free medium RD6F used for dental pulp cell culture was prepared by mixing RPMI1640 medium (Sigma) and DMEM medium (Sigma) at a ratio of 1: 1, bixillin (90 mg / ml) (Meiji, Japan), kanamycin (90 mg / day).
  • TIG-3 cells Human fetal lung-derived normal fibroblasts (TIG-3 cells) TIG-3 cells, which are Japanese fetal lung-derived fibroblasts established by the Tokyo Metropolitan Institute of Health and Longevity Medical Center, have 4 genes (Oct3 / 4, Sox2, Klf4, c) using retrovirus. -Myc) was introduced and human iPS cells were induced.
  • TIG-3 cells were prepared by adding 10% fetal bovine serum (Hyclone® Thermo Scientific, US) and 1% penicillin-streptomycin (GIBCO) to DMEM medium (Sigma) at a ratio of 1: 4 every 2-3 days. The cells were seeded on a 10 cm dish (Falcon) at a split ratio of 5% CO 2 /95% in a gas phase and cultured in a 37 ° C. incubator.
  • Retroviral Packaging that is capable of producing retrovirus structural proteins (gag, pol, env) stably for a long period of time. Retrovirus production was performed using Cell, amphotropic (PLAT-A: Cell Biolabs Inc. CA, USA).
  • transfection When transfection is performed, a medium in which 10% FBS is added to DMEM medium the day before is used, and 2 ⁇ 10 6 cells are seeded in a collagen-coated 25 cm 2 flask (BioCoat Collagen I Cellware, Falcon) After 16 to 24 hours, when the cells became 70% to 80% confluent, the cells were used for gene transfer (transfection), and the virus supernatant 48 to 72 hours after the start of transfection was collected and infected with target cells.
  • a medium in which 10% FBS is added to DMEM medium the day before the day before is used, and 2 ⁇ 10 6 cells are seeded in a collagen-coated 25 cm 2 flask (BioCoat Collagen I Cellware, Falcon) After 16 to 24 hours, when the cells became 70% to 80% confluent, the cells were used for gene transfer (transfection), and the virus supernatant 48 to 72 hours after the start of transfection was collected and infected with target cells.
  • Tic human iPS cell line
  • JCRB1331 its culturing method Tic
  • Tic was used as a control iPS cell. Inoculate Tic with 0.1% gelatin in advance on inactivated mouse fetal fibroblasts (Millipore: PMEF-H) seeded as feeder cells and culture using serum-supplemented ES cell medium. went. The cell dispersion was subcultured using Despase II (1 mg / ml) (Roche 4942078, Basel, Switzerland).
  • retrovirus 1-2 A retrovirus was prepared by introducing the reprogramming 4 gene into PLAT-A cells passaged and maintained according to the method of 2).
  • the plasmids used were pMXs- (hOct3 / 4), pMXs- (hSox2), pMXs- (hKlf4), pMXs- (hc-Myc) (Cell Biolabs Inc. CA, USA) prepared in Yamanaka Laboratory, Kyoto University. Was used.
  • TIG-3 cells were infected. In order to attenuate the toxicity of polybrene 4 hours after the infection, the same amount of medium as the virus supernatant was added. The medium was changed 24 hours after infection (Day 1), and culture was performed for 4 days (until Day 4). The medium was changed every 2 days. Four days after infection (Day 4), TIG-3 cells were dispersed into single cells by 0.05% trypsin / EDTA treatment.
  • PLAT-A cells were treated with pMXs- (hOct3 / 4), pMXs- (hSox2), pMXs- (hKlf4), pMXs- ( hc-Myc) each transfected virus supernatant mixture (equal to pMXs- (4F)), or pMXs- (4F) and pMXs- (EGFP) virus supernatant in a ratio of 3: 1 Infect the TIG-3 cells with each of the virus mixtures mixed in 1.
  • Retrovirus supernatant is collected in serum-free medium hESF9, and 24 hours after virus infection is established as serum-free medium hESF9. The effect was examined (Figure 3).
  • PLAT-A cells were transfected with pMXs- (EGFP) or pMXs-( ⁇ ) under serum-added culture conditions with 10% FBS added to DMEM medium, and serum medium conditions with 10% FBS added to DMEM medium ( The virus supernatant 24 to 48 hours after transfection was collected under each condition of condition A) or hESF9 serum-free medium condition (hereinafter referred to as condition B), and infected with TIG-3 cells as target cells. I let you.
  • the medium is exchanged every 2 to 3 days, and after cell growth by treatment with 0.05% trypsin / EDTA at the stage of growth to sub-confluence, 0.1% trypsin inhibitor is added to neutralize trypsin action, and the cells on the dish are neutralized. And subcultured every 3-4 days.
  • RD6F shown below was used as a serum-free medium. Specifically, DMEM medium (Sigma) and RPMI 1640 medium (Sigma) were mixed at a ratio of 1: 1, and bivicillin (90 mg / ml) (Meiji, Japan), kanamycin (90 mg / ml) (GIBCO), sodium pyruvate (110 mg / ml).
  • serum-free medium hESF9 An equal amount of serum-free medium hESF9 was added 4 hours after infection, the medium was changed 24 hours later, and each cell was dispersed into single cells by 0.05% trypsin-EDTA treatment 5 days after infection (Day 5). On a dish coated with fibronectin (2 ⁇ g / cm 2 ), seeding was performed so that the number of cells was 1.0 ⁇ 10 5 cells / 10 cm. After re-seeding, the medium was changed every 2 days using a serum-free medium hESF9 medium (FIG. 4).
  • iPS cells maintained in serum-free medium hESF9T containing TGF- ⁇ 1 could be subcultured while maintaining undifferentiation (FIG. 7A).
  • various concentrations (0, 0.1, 1, 2, 5, 10 ng / ml) of TGF- ⁇ 1 were added to hESF9 medium, and total RNA was recovered after 4 days of culture.
  • QX100 TM Droplet Digital Gene expression analysis was performed using TM PCR system (Bio-RAD).
  • Droplet Digital TM PCR uses ddPCR TM supermix (Bio Rad) according to the attached protocol, creates droplets with QX100 TM Droplet Generator (Bio Rad), amplifies it with PCR reaction, then QX100 TM Droplet Reader (Bio Rad) ) was used for analysis.
  • the number of cells that remained undifferentiated increased depending on the TGF- ⁇ 1 concentration, and the expression of Oct3 / 4 and Nanog undifferentiated marker genes was enhanced (FIG. 7B).
  • the TGF- ⁇ 1 concentration was 2 to 10 ng / ml, and the most undifferentiated marker was highly expressed. However, in the absence of TGF- ⁇ 1, the expression of the undifferentiated marker gene was remarkably reduced, and the mesoderm differentiation marker was positive.
  • differentiation markers such as minogen activator inhibitor-1 (PAI-1) and GATA-binding protein 4 (GATA4), which is an endoderm differentiation marker such as cardiac muscle.
  • PAI-1 minogen activator inhibitor-1
  • GATA4 GATA-binding protein 4
  • illustra RNA spin Mini Isolation kit (GE Healthcare UK Ltd, England) was used to extract total RNA of the cells cultured as described above according to the attached protocol. Nucleic acid quantification was performed using Nano Drop® (Nano Drop Technologies, Inc., USA).
  • RNA extracted from cells (1 ⁇ g) using High Capacity RNA-to-cDNA Master Mix (Applied Biosystems, CA, USA) and thermal cycler (PTC-0220 DNA Engine Dyad: MJ Japan, Tokyo)
  • a reverse transcription reaction was performed by incubation at 25 ° C. for 5 minutes, 42 ° C. for 30 minutes, and 85 ° C. for 5 minutes to synthesize cDNA.
  • RT-PCR is performed using KOD FX Neo (Toyobo, Osaka, Japan) under the conditions of denaturation reaction 98 ° C., 10 seconds, annealing 62 ° C., 30 seconds, extension reaction 68 ° C., 30 seconds. 35 cycles were performed to obtain a PCR product.
  • This PCR product was electrophoresed on a 1.5% agarose gel (Invitrogen) and then visualized with SYBR Safe DNA gel stain (Invitrogen).
  • the expression of undifferentiated marker genes of human ES cells, Sox2, Nanog, Oct3 / 4, Esg1, Rex-1 (Reduced-expression 1; Zfp42) was examined.
  • expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene was examined (FIG. 7C).
  • Human iPS cells induced and maintained under serum-free culture conditions expressed undifferentiated markers, Sox2, Nanog, Oct3 / 4, Esg1, and Rex-1. On the other hand, these undifferentiated markers were not expressed in the pulp-derived cells before gene introduction (FIG. 7C).
  • the cells were seeded on a gelatin-coated plate, and differentiation induction was further performed for 10 days. Fluorescent immunostaining was performed on the cells 14 days after induction using various differentiation marker antibodies. Each antibody was visualized with Alexa Fluor (R) 594 and combined with nuclear staining with DAPI. As a result, neural stem cell line marker nestin, nerve cell marker ⁇ III-tubulin, mesoderm marker ⁇ -smooth muscle actin (SMA), Expression was observed for ⁇ -fetoprotein (AFP), an endoderm marker. On the other hand, the undifferentiated marker Oct3 / 4 was not expressed (FIG. 8A).
  • the sections showed ectoderm tissues such as epidermis and nerves, mesoderm tissues such as cartilage, muscle and connective tissue, and tissues that differentiated into endoderm such as the digestive tract and liver, and the excised tumor was shown to be teratoma. (FIG. 8B).
  • ectoderm tissues such as epidermis and nerves
  • mesoderm tissues such as cartilage, muscle and connective tissue
  • endoderm such as the digestive tract and liver
  • STR Short Tandem Repeat analysis Genomic DNA was extracted from dental pulp-derived cells collected from patients and human iPS cells prepared by introducing reprogramming genes under serum-free culture conditions. Powerplex 16 system (Promega Corporation , Madison, Wis.) And STR analysis using ABI PRISM® 3100 Genetic analyzer (Applied Biosystems) and Gene Mapper v3.5, allele patterns of 16 loci matched.
  • hypotonic solution was treated with a 0.075 M potassium chloride solution at 37 ° C. for 10 minutes, fixed with a Carnoy fixative, centrifuged, and fixed again with a Carnoy fixative. It was dropped on a slide glass, air-dried, stained with 4% Giemsa staining solution (Mudo Chemical, Tokyo, Japan), and then examined with an optical microscope for karyotype analysis.
  • stem cells such as ES cells and iPS cells are cultured under conditions containing inactivated feeder cells and animal-derived components such as serum.
  • animal-derived components such as serum.
  • various foreign antigens may be mixed, and it is difficult to apply to regenerative medicine. It is also very difficult to clarify the growth / differentiation control mechanism and its control factors of these stem cells.
  • human iPS cells can be induced for the first time by performing the entire process from primary culture to viral infection and human iPS cell establishment in a complete serum-free culture system.
  • the method of subculture while maintaining undifferentiation and pluripotency of the present invention is applicable not only to human iPS cells but also to stem cells of all animals.
  • the serum-free culture system of the present invention comprises only known factors, it is easy to identify and examine various factors that control the maintenance of proliferation and differentiation of stem cells, and to develop development, tissue, and organ regeneration. It becomes possible to elucidate the mechanism, apply it to drug discovery screening, and realize safe and reliable regenerative medicine.
  • the present invention can be used in the development of pharmaceuticals and medical materials, biochemical research fields, livestock industry, and the like.

Landscapes

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

Abstract

 Le problème est la production de cellules iPS dans des conditions de culture sans sérum sans utiliser des cellules nourricières, et la description d'un procédé de culture capable de maintenir l'état non différencié et la capacité de différenciation pluripotente de cellules iPS et d'autres cellules souches sur une période prolongée dans des conditions de culture sans sérum sans utiliser de cellules nourricières. La présente invention concerne : un procédé de production de cellules souches pluripotentes induites (cellules iPS), caractérisé en ce que des cellules iPS sont induites par culture de cellules somatiques reprogrammées dans du milieu sans sérum sans utiliser des cellules nourricières, de préférence en utilisant de la fibronectine en tant que facteur d'adhésion; et un procédé de maintenance de l'état non différencié et de la capacité de différenciation pluripotente de cellules souches, caractérisé en ce qu'une protéine appartenant à la famille du facteur de croissance tumoral β (TGF-β) est ajoutée et les cellules souches sont sous-cultivées dans du milieu sans sérum sans utiliser de cellules nourricières.
PCT/JP2014/006454 2013-12-26 2014-12-25 PROCÉDÉ D'ÉTABLISSEMENT DE CELLULES iPS ET PROCÉDÉ DE MAINTENANCE À LONG TERME DE CELLULES SOUCHES WO2015098111A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361920868P 2013-12-26 2013-12-26
US61/920,868 2013-12-26
JP2014087314A JP6516280B2 (ja) 2013-12-26 2014-04-21 iPS細胞の樹立方法および幹細胞の長期維持方法
JP2014-087314 2014-04-21

Publications (1)

Publication Number Publication Date
WO2015098111A1 true WO2015098111A1 (fr) 2015-07-02

Family

ID=53478007

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2014/006454 WO2015098111A1 (fr) 2013-12-26 2014-12-25 PROCÉDÉ D'ÉTABLISSEMENT DE CELLULES iPS ET PROCÉDÉ DE MAINTENANCE À LONG TERME DE CELLULES SOUCHES

Country Status (1)

Country Link
WO (1) WO2015098111A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114939097A (zh) * 2017-05-02 2022-08-26 田边刚士 医药品组合物及化妆品组合物

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06169761A (ja) * 1992-12-04 1994-06-21 Noevir Co Ltd 動物由来線維芽細胞の無血清培養用培地及び培養方法
JP2013510567A (ja) * 2009-11-12 2013-03-28 テクニオン リサーチ アンド ディベロップメント ファウンデーション リミテッド 多能性幹細胞を未分化状態で培養する培地、細胞培養および方法

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH06169761A (ja) * 1992-12-04 1994-06-21 Noevir Co Ltd 動物由来線維芽細胞の無血清培養用培地及び培養方法
JP2013510567A (ja) * 2009-11-12 2013-03-28 テクニオン リサーチ アンド ディベロップメント ファウンデーション リミテッド 多能性幹細胞を未分化状態で培養する培地、細胞培養および方法

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CHEN, G. ET AL.: "Chemically defined conditions for human iPSC derivation and culture.", NAT. METHODS, vol. 8, no. 5, 2011, pages 424 - 429 *
HAYASHI, Y. ET AL.: "Reduction of N- Glycolylneuraminic Acid in Human Induced Pluripotent Stem Cells Generated or Cultured under Feeder- and Serum-Free Defined Conditions.", PLOS ONE, vol. 5, no. 11, 2010, pages E14099, PAGES 1 - 11 *
YAMASAKI, S. ET AL.: "Generation of Human Induced Pluripotent Stem (iPS) Cells in Serum- and Feeder-Free Defined Culture and TGF-beta1 Regulation of Pluripotency.", PLOS ONE, vol. 9, no. 1, 29 January 2014 (2014-01-29), pages E87151, PAGES 1 - 13 *
YAMASAKI, S. ET AL.: "Generation of Human Induced Pluripotent Stem (iPS) Cells in Serum- and Feeder-free Defined Culture from Fetal Lung Fibroblasts and Dental Pulp Cells Derived from a Patient with Cleidocranial Dysplasia.", VITRO CELL . DEV. BIOL. ANIM., vol. 48, no. SUPPL., 2012, pages 48 - 49 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114939097A (zh) * 2017-05-02 2022-08-26 田边刚士 医药品组合物及化妆品组合物

Similar Documents

Publication Publication Date Title
US20180355313A1 (en) Pluripotent stem cell that can be isolated from body tissue
CA2768238C (fr) Cellule souche pluripotente isolee a partir de tissue organique
KR20080056181A (ko) 전구세포주의 유도 방법
KR101861171B1 (ko) 투석된 혈청이 있는 심근세포 배지
WO2006091766A2 (fr) Cellules souches du trophoblaste humain et utilisation associee
CN103237886A (zh) 细胞集合体的非静态悬浮培养
KR101656388B1 (ko) 다능성 줄기세포의 제작, 유지, 증식을 증진하는 대사산물 및 이를 포함하는 조성물과 배양방법
CN101978047A (zh) 干细胞聚集体及制备和使用方法
JP2022513355A (ja) 条件付き不死化に関する制御可能な導入遺伝子を含む人工多能性細胞
JP5751548B2 (ja) イヌiPS細胞及びその製造方法
CN110713973A (zh) 一种诱导多能干细胞分化为间充质干细胞的培养基组合及方法
WO2012133942A1 (fr) Cellule souche pluripotente pouvant être isolée de tissu adipeux ou de cordon ombilical de corps biologique
RU2399667C1 (ru) Способ получения плюрипотентных клеток
KR20160063066A (ko) 유전자 전달용 그래핀 옥사이드―폴리에틸렌이민 복합체 및 이의 용도
Yamasaki et al. Long-term serial cultivation of mouse induced pluripotent stem cells in serum-free and feeder-free defined medium.
WO2015098111A1 (fr) PROCÉDÉ D'ÉTABLISSEMENT DE CELLULES iPS ET PROCÉDÉ DE MAINTENANCE À LONG TERME DE CELLULES SOUCHES
JP6516280B2 (ja) iPS細胞の樹立方法および幹細胞の長期維持方法
EP1775340A1 (fr) Procede de transdifferenciation cellulaire
CN109852587B (zh) 一种人科凯恩氏综合征特异性成体干细胞的制备方法
KR102137884B1 (ko) 타우로우루소디옥시콜린산을 포함하는 고효율 세포전환용 배지 첨가제
JP2005095138A (ja) 多分化能を有する細胞の分化誘導方法
KR20190070254A (ko) 페닐부틸산나트륨을 포함하는 고효율 세포전환용 배지 첨가제
CN117487743B (zh) 一种诱导鸡胚成纤维细胞为鸡多能干细胞的化学诱导剂及诱导方法
CN117467599B (zh) 一种重编程鸡性腺体细胞为鸡多能干细胞的化学诱导剂及重编程方法
KR102137885B1 (ko) 부틸화하이드록시아니솔을 포함하는 고효율 세포전환용 배지 첨가제

Legal Events

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

Ref document number: 14874069

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14874069

Country of ref document: EP

Kind code of ref document: A1