US20150072417A1 - Method of nuclear reprogramming - Google Patents

Method of nuclear reprogramming Download PDF

Info

Publication number
US20150072417A1
US20150072417A1 US14/502,374 US201414502374A US2015072417A1 US 20150072417 A1 US20150072417 A1 US 20150072417A1 US 201414502374 A US201414502374 A US 201414502374A US 2015072417 A1 US2015072417 A1 US 2015072417A1
Authority
US
United States
Prior art keywords
cells
cell
genes
gene
expression vector
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/502,374
Inventor
Shinya Yamanaka
Keisuke Okita
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kyoto University
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 US14/502,374 priority Critical patent/US20150072417A1/en
Publication of US20150072417A1 publication Critical patent/US20150072417A1/en
Priority to US16/702,391 priority patent/US20200172875A1/en
Priority to US18/146,644 priority patent/US20230282445A1/en
Abandoned legal-status Critical Current

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
    • 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]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • HELECTRICITY
    • H01ELECTRIC ELEMENTS
    • H01JELECTRIC DISCHARGE TUBES OR DISCHARGE LAMPS
    • H01J37/00Discharge tubes with provision for introducing objects or material to be exposed to the discharge, e.g. for the purpose of examination or processing thereof
    • H01J37/02Details
    • H01J37/24Circuit arrangements not adapted to a particular application of the tube and not otherwise provided for
    • H01J37/241High voltage power supply or regulation circuits
    • HELECTRICITY
    • H01ELECTRIC ELEMENTS
    • H01JELECTRIC DISCHARGE TUBES OR DISCHARGE LAMPS
    • H01J37/00Discharge tubes with provision for introducing objects or material to be exposed to the discharge, e.g. for the purpose of examination or processing thereof
    • H01J37/32Gas-filled discharge tubes
    • H01J37/32009Arrangements for generation of plasma specially adapted for examination or treatment of objects, e.g. plasma sources
    • H01J37/32073Corona discharge
    • 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
    • C12N2501/605Nanog
    • 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
    • C12N2501/60Transcription factors
    • C12N2501/608Lin28
    • 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
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic 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
    • 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
    • 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
    • C12N2506/1307Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from connective tissue cells, from mesenchymal cells from adult fibroblasts
    • 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
    • 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
    • C12N2510/00Genetically modified cells
    • HELECTRICITY
    • H01ELECTRIC ELEMENTS
    • H01JELECTRIC DISCHARGE TUBES OR DISCHARGE LAMPS
    • H01J2237/00Discharge tubes exposing object to beam, e.g. for analysis treatment, etching, imaging
    • H01J2237/03Mounting, supporting, spacing or insulating electrodes
    • H01J2237/038Insulating
    • HELECTRICITY
    • H01ELECTRIC ELEMENTS
    • H01JELECTRIC DISCHARGE TUBES OR DISCHARGE LAMPS
    • H01J2237/00Discharge tubes exposing object to beam, e.g. for analysis treatment, etching, imaging
    • H01J2237/06Sources
    • H01J2237/063Electron sources
    • H01J2237/06375Arrangement of electrodes

Definitions

  • the present invention relates to a method of reprogramming a somatic cell and producing an induced pluripotent stem cell.
  • embryonic stem cells are capable of being cultured for a long time while maintaining their potential for differentiating into all types of cells found in a living organism.
  • human ES cells are expected to serve for cell transplantation therapies for many diseases, including Parkinson's disease, juvenile diabetes, and leukemia.
  • ES cell transplantation poses the problem of causing rejections as with organ transplantation. Additionally, not a few people oppose the use of ES cells established with the destruction of a human embryo, from an ethical viewpoint.
  • iPS cells induced pluripotent stem cells
  • ES-like cells embryonic stem cell-like cells
  • iPS cells induced pluripotent stem cells
  • the established cells will be useful as ideal pluripotent cells that do not pose the problems of rejections and ethical issues.
  • iPS cells can be produced from mouse and human differentiated cells, arousing great attention (International Patent Application Publication No. WO2007/69666; Cell, 126, pp. 663-676, 2006; Cell, 131, pp. 861-872, 2007; Science, 318, pp. 1917-1920, 2007; Nature, 451, pp. 141-146, 2008).
  • All these methods comprise the step of introducing a plurality of particular nuclear reprogramming factors (e.g., in Cell, 126, pp. 1-14, 2006, 4 factors are used: Oct3/4, Sox2, Klf4, and c-Myc) into a somatic cell to achieve reprogramming, which step involves the use of a retrovirus or a lentivirus for the purpose of introducing the genes that encode the nuclear reprogramming factors into a somatic cell efficiently.
  • a retrovirus or a lentivirus for the purpose of introducing the genes that encode the nuclear reprogramming factors into a somatic cell efficiently.
  • the present inventors extensively investigated to solve the problems described above, and found that an iPS cell can be produced by introducing genes that encode reprogramming factors into a somatic cell by means of a non-viral expression vector such as a plasmid vector, and that a safe iPS cell can be obtained from a somatic cell by the method.
  • the present invention has been developed on the basis of these findings.
  • the present invention provides a method of producing an induced pluripotent stem cell, comprising the step of introducing at least one kind of non-viral expression vector incorporating at least one gene that encodes a reprogramming factor into a somatic cell.
  • the present invention provides the above-described method wherein the vectors are non-viral expression vectors autonomously replicable outside a chromosome; and the above-described method wherein the vector is a plasmid vector.
  • the present invention provides the above-described method wherein the gene that encodes a reprogramming factor is one of genes selected by a method of screening for nuclear reprogramming factors described in WO 2005/80598 or a combination of a plurality of such genes; and the above-described method wherein the gene that encodes a reprogramming factor is one or more kinds of genes selected from the group consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, and the Nanog gene, preferably a combination of two kinds of genes, more preferably a combination of three kinds of genes, particularly preferably a combination of four or more kinds of genes.
  • More preferable combinations are (a) a combination of two kinds of genes consisting of an Oct family gene and a Sox family gene; (b) a combination of three kinds of genes consisting of an Oct family gene, a Klf family gene, and a Sox family gene; (c) a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene; (d) a combination of four kinds of genes consisting of an Oct family gene, a Sox family gene, a Lin family gene, and the Nanog gene; (e) a combination of six kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, and the Nanog gene; and the like. Furthermore, it is also preferable to include the TERT gene and/or the SV40 Large T antigen gene in the combination. As the case may be, it is preferable to exclude Klf family genes.
  • Particularly preferred combinations thereof are a combination of two kinds of genes consisting of Oct3/4 and Sox2; a combination of three kinds of genes consisting of Oct3/4, Klf4, and Sox2; a combination of four kinds of genes consisting of Oct3/4, Klf4, Sox2, and c-Myc; a combination of four kinds of genes consisting of Oct3/4, Sox2, Lin28, and Nanog; and a combination of six kinds of genes consisting of Oct3/4, Klf4, Sox2, c-Myc, Lin28, and Nanog. It is also preferable to include the TERT gene and/or the SV40 Large T antigen gene in these combinations. As the case may be, it is preferable to exclude Klf4.
  • the present invention provides the above-described method wherein the number of kinds of non-viral expression vectors introduced into a somatic cell is 1, 2, 3, or 4; the above-described method wherein the genes that encode reprogramming factors are a combination of three kinds of genes consisting of an Oct family gene, a Klf family gene, and a Sox family gene, and these genes are incorporated in one kind of non-viral expression vector; the above-described method wherein the genes that encode nuclear reprogramming factors are a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene, and the Oct family gene, the Klf family gene, and the Sox family gene are incorporated in one kind of non-viral expression vector; the above-described method wherein the Oct family gene, the Klf family gene, and the Sox family gene are incorporated in one kind of non-viral expression vector in this order in the orientation from the 5′ to 3′ end;
  • the present invention provides the above-described method wherein two or more kinds of the above-described non-viral expression vectors are concurrently introduced into a somatic cell; the above-described method wherein the genes that encode reprogramming factors are a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene, and a first non-viral expression vector incorporating three or less kinds of genes selected from among the four kinds of genes, and a second non-viral expression vector incorporating the remaining gene(s) out of the four kinds of genes are concurrently introduced into a somatic cell; the above-described method wherein the three or less kinds of genes are an Oct family gene, a Klf family gene, and a Sox family gene, and the remaining gene is a Myc family gene; the above-described method wherein the three or less kinds of genes are Oct3/4, Klf4, and Sox2, and the remaining gene is c-Myc; and the above
  • the present invention provides the above-described method wherein a first non-viral expression vector harboring Oct3/4, Klf4, and Sox2, and a second non-viral expression vector harboring c-Myc are introduced into a somatic cell; the above-described method wherein a first non-viral expression vector harboring Oct3/4, Klf4, and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector harboring c-Myc are introduced into a somatic cell; the above-described method wherein Oct3/4, Klf4, and Sox2 are ligated in this order in the orientation from the 5′ to 3′ end with an intervening sequence enabling polycistronic expression and inserted into the first non-viral expression vector; the above-described method wherein the first non-viral expression vector and the second non-viral expression vector are concurrently introduced into a somatic cell; and the above-described method wherein the introduction is repeatedly performed twice or more.
  • the present invention provides the above-described method wherein the somatic cell is a somatic cell of a mammal, including a human, preferably a human or mouse somatic cell, particularly preferably a human somatic cell; the above-described method wherein the somatic cell is a fetal human cell or a somatic cell derived from an adult human; and the above-described method wherein the somatic cell is a somatic cell collected from a patient.
  • the present invention provides an induced pluripotent stem cell that can be obtained by the above-described method.
  • the present invention also provides an induced pluripotent stem cell wherein all or some of the at least one non-viral expression vector introduced is substantially not integrated in the chromosome.
  • the somatic cell is a somatic cell of a mammal, including a human, preferably a human or mouse somatic cell, particularly preferably a human somatic cell; the above-described induced pluripotent stem cell wherein the somatic cell is a fetal human cell or a somatic cell derived from an adult human; and the above-described induced pluripotent stem cell wherein the somatic cell is a somatic cell collected from a patient.
  • a non-viral expression vector preferably a plasmid vector, for use in the above-described method of producing an induced pluripotent stem cell, incorporating at least one gene that encodes a reprogramming factor, is also provided by the present invention.
  • a somatic cell induced and differentiated from the above-described induced pluripotent stem cell is also provided by the present invention.
  • the present invention also provides a stem cell therapy comprising the step of transplanting to a patient a somatic cell obtained by differentiation induction of an induced pluripotent stem cell obtained by the above-described method using a somatic cell separated from the patient.
  • the present invention further provides a method of evaluating the physiological activities and toxicities of compounds, drugs, poisonous substances and the like using various cells obtained by differentiation induction of an induced pluripotent stem cell obtained by the above-described method.
  • the induced pluripotent stem cell provided by the present invention is advantageous in that tumorigenesis and other problems do not arise in the somatic cells and tissues obtained by differentiating the induced pluripotent stem cell.
  • all or some of the at least one non-viral expression vector introduced is episomally present, substantially not integrated in the chromosome.
  • the method of the present invention makes it possible to prepare a highly safe induced pluripotent stem cell from, for example, a patient's somatic cell, and the cells obtained by differentiating this cell (e.g., myocardial cells, insulin-producing cells, or nerve cells and the like) can be safely used for stem cell transplantation therapies for a broad range of diseases, including heart failure, insulin-dependent diabetes, Parkinson's disease and spinal injury.
  • a highly safe induced pluripotent stem cell from, for example, a patient's somatic cell
  • the cells obtained by differentiating this cell e.g., myocardial cells, insulin-producing cells, or nerve cells and the like
  • stem cell transplantation therapies for a broad range of diseases, including heart failure, insulin-dependent diabetes, Parkinson's disease and spinal injury.
  • FIG. 1 shows a time course protocol for transfecting a somatic cell (MEF) with Oct3/4, Klf4, Sox2, and c-Myc using plasmids according to the method of the present invention, results of seven independent tests (left photographs, 432A-1 to 432A-7: cell density 1 ⁇ 10 6 cells/100 mm dish) and results of another test (right photographs, 432B-1: cell density 2 ⁇ 10 5 cells/100 mm dish). The lowermost panels in the center show control results (no transfection).
  • the Phase columns show phase-contrast images, and the GFP columns show GFP-positive colonies.
  • FIG. 2 shows an expression plasmid for iPS cell production.
  • Three kinds of cDNAs that encode Oct3/4, Klf4, and Sox2 were ligated in this order with sequence encoding the 2A peptide as intervening sequence, and inserted into the pCX plasmid (pCX-2A-mOKS).
  • pCX-2A-mOKS pCX-2A-mOKS
  • a cDNA of c-Myc was inserted into pCX (pCX-c-Myc).
  • the bald lines show the amplification regions used in the PCR analysis for detecting plasmid integration in the genome ( FIG. 6 ).
  • FIG. 3 shows the time schedules for IFS cell induction using plasmids.
  • the solid arrows indicate the time points of transfection of the respective plasmids.
  • FIG. 4 shows the morphology of non-virus mediated IFS cells established.
  • FIG. 5 shows results of PCR analysis for the genetic expression of ES cell markers, obtained using total RNAs isolated from ES cells, iPS cells induced using retroviruses (clone 20D-17: Nature, 448, pp. 313-317, 2007), iPS cells induced using plasmids (clones 440A-3, 4, 7, 8, 10 and 11; clone 432A-1), and MEF cells.
  • FIG. 6 shows the detection of plasmid integration by PCR.
  • Genomic DNAs were extracted from a C57BL/6 mouse, iPS cell induced using retroviruses (clone 20D-17), iPS cells induced with plasmids (clone 432A-1; clones 440A-1 to 11) and MEF cells, and analyzed by PCR using the primers shown in FIGS. 2 , 13 and 14 .
  • the bands derived from endogenous genes are indicated by the outlined arrowheads
  • the bands derived from integrated plasmids are indicated by the solid arrowheads.
  • the lower band indicates wild-type alleles
  • the upper band indicates knocked-in alleles.
  • FIG. 7 shows results of teratoma formation.
  • FIG. 8 shows chimeric mice derived from iPS cells without integration (clones 440A-3 and -8).
  • FIG. 9 shows the detection of integration of plasmids by PCR.
  • Genomic DNAs were extracted from an ICR mouse, iPS cell (clone 432A-1), and chimeric mice derived from iPS cells induced using plasmids (clone 432A-1; clones 0.440A-3, 8), and the O-1, K and M regions shown in FIG. 2 were amplified by PCR.
  • the bands derived from endogenous genes are indicated by the outlined arrowheads, and the bands derived from integrated plasmids are indicated by the solid arrowheads.
  • the presence of the Nanog reporter and Fbx15 reporter was also detected by PCR.
  • FIG. 10 shows the probes used in Southern blot analysis and the positions of the restriction endonuclease recognition sites.
  • E indicates EcoRI
  • B indicates BamHI.
  • FIG. 11 shows results of Southern blot analysis.
  • Genomic DNAs (6 ⁇ g) were extracted from RF8 ES cells and iPS cells (clones 440A-3, 4, 7, 8, 10, and 11; clone 432A-1), and cleaved with BamHI and EcoRI.
  • the outlined arrowheads indicate the bands derived from endogenous genes, and the solid arrowhead indicates the band derived from the Oct3/4 pseudogene (estimated size 2049 bp) on chromosome 3.
  • the arrows indicate the bands derived from transgenes.
  • FIG. 12 shows results of SSLP analysis.
  • genomic DNAs each 50 ng
  • RFB ES cell iPS cells without integration
  • MEF cells MEF cells
  • FIGS. 13 and 14 show the primers used for PCR in Examples 1 to 3.
  • FIG. 15 shows a time course protocol for transfecting human dental pulp stem cells with Oct3/4, Klf4, Sox2, c-Myc, Lin28, Nanog and the SV40 Large T antigen using plasmids according to the method of the present invention, and 16 independent IFS cell colonies.
  • FIGS. 16 and 17 show photographs of IFS cells established from fetal HDF (5 clones: 203A-1 to 203A-5, of which 203A-4 was picked up as a negative control) on day 31 after transfection ( FIG. 16 ) and in the 2nd subculture ( FIG. 17 ).
  • FIG. 18 shows the results of genomic-PCR analysis of 5 iPS cell clones (203A-1 to 203A-5).
  • FIGS. 19 and 20 show photographs of IFS cells established from human dental pulp stem cells (5 clones: 217A-1 to -4 and -6) on day 35 after transfection ( FIG. 19 ) and in the 2nd subculture ( FIG. 20 ).
  • FIG. 21 shows the results of genomic-PCR analysis of 5 iPS cell clones (217A-1 to -4 and -6).
  • FIGS. 22 and 23 show photographs of IFS cells established from young female HDF (2 clones: 279A-1 and -2) on day 35 after the first electroporation ( FIG. 22 ) and clone 279A-2 after passage culture ( FIG. 23 ; the right panel is a closeup picture of the boxed area in the left panel).
  • FIG. 24 shows the results of genomic-PCR analysis of iPS cell clone 279A-2 demonstrating the integration of the transgenes.
  • FIG. 25 shows photographs of iPS cells (8 clones: 497A-1 to A-8) after the selection (colonies were selected on day 25 after transfection).
  • the upper panels show phase-contrast images, and the lower panels show GFP-positive colonies.
  • FIG. 26 shows the results of genomic-PCR analysis of 5 iPS cell clones (497A-1 to A-5). In 497A-2 and 497A-5, no exogenous gene was not integrated into the genome.
  • the method of the present invention is intended to produce an induced pluripotent stem cell, comprising the step of introducing at least one kind of non-viral expression vector incorporating at least one gene that encodes a reprogramming factor into a somatic cell.
  • the non-viral expression vector is preferably an expression vector autonomously replicable outside a chromosome, more preferably a plasmid expression vector.
  • a nuclear reprogramming factor screening method described in WO 2005/80598 can be utilized. All disclosures therein are incorporated herein by reference. Those skilled in the art are able to screen for nuclear reprogramming factors, and to utilize them for the method of the present invention, by referring to the aforementioned publication. It is also possible to identify nuclear reprogramming factors using a method modified or altered from the above-described screening method.
  • genes that encode reprogramming factors are disclosed in WO2007/69666. All disclosures therein are incorporated herein by reference. Those skilled in the art are able to choose genes that can suitably be used in the method of the present invention as appropriate by referring to the aforementioned publication. Other examples of combinations of genes that encode reprogramming factors are given in Science, 318, pp. 1917-1920, 2007, WO2008/118820 and the like.
  • genes that encode reprogramming factors include one or more kind of genes selected from the group consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, and the Nanog gene, preferably a combination of two kinds of genes, more preferably of three kinds of genes, and particularly preferably of four kinds of genes.
  • Oct family genes examples include Klf family genes, Sox family genes, and Myc family genes.
  • Lin family genes those skilled in the art are likewise able to extract a family gene.
  • Lin28 and Lin28B may be included.
  • More preferable combinations include, but are not limited to,
  • genes are present in common in mammals, including humans. Genes derived from optionally chosen mammals (e.g., humans, mice, rats, bovines, sheep, horses, monkeys) can be used in the present invention.
  • mutant genes whose translation products have several (e.g., 1 to 10, preferably 1 to 6, more preferably 1 to 4, more preferably 1 to 3, particularly preferably 1 or 2) amino acids substituted, inserted, and/or deleted, and possess a function similar to that of the wild type gene product, can also be utilized.
  • mutant genes whose translation products have several (e.g., 1 to 10, preferably 1 to 6, more preferably 1 to 4, more preferably 1 to 3, particularly preferably 1 or 2) amino acids substituted, inserted, and/or deleted, and possess a function similar to that of the wild type gene product, can also be utilized.
  • c-Myc genes the wild type, a gene encoding stable type mutant (T58A) and the like may be used. The same applies to other gene products.
  • a gene that encodes a factor that induces cell immortalization may further be combined.
  • the TERT gene and one or more kind of genes selected from the group consisting of the following genes: SV40 Large T antigen, HPV16 E6, HPV16 E7, and Bmil, can be used singly, or in combination as appropriate.
  • the Klf family gene may be excluded from the aforementioned combinations.
  • one or more kind of genes selected from the group consisting of Fbx15, ERas, ECAT15-2, Tcl1, and ⁇ -catenin may be combined, and/or one or more kind of genes selected from the group consisting of ECAT1, Esg1, Dnmt3L, ECAT8, Gdf3, Sox15, ECAT15-1, Fth117, Sal14, Rex1, UTF1, Stella, Stat3, and Grb2 may also be combined. These combinations are specifically described in WO2007/69666.
  • the gene(s) can be excluded from the genes to be introduced.
  • the remaining one or more genes can be introduced using a non-viral expression vector according to the method of the present invention.
  • the remaining one or more genes can be introduced using a non-viral expression vector according to the method of the present invention.
  • the remaining one or more genes can be introduced using a non-viral expression vector according to the method of the present invention.
  • a gene that encodes a factor that induces cell immortalization may further be combined.
  • genes selected from the group consisting of the TERT gene, and the following genes: HPV16 E6, HPV16 E7, and Bmil can be used singly, or in combination as appropriate.
  • L-Myc can be used in place of c-Myc.
  • genes are not limited thereto.
  • the scope of the present invention includes a method wherein one or more genes selected from among the above-described genes are introduced into a somatic cell using a non-viral expression vector, and the remaining gene or gene product is introduced into the somatic cell by another means.
  • a viral vector such as retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector.
  • genes that encode reprogramming factors for example, a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene can be used.
  • a combination of three kinds of genes consisting of an Oct family gene, a Klf family gene, and a Sox family gene, or a combination of two kinds of genes selected from among the aforementioned three kinds of genes can also be used.
  • the above-described four kinds, three kinds, or two kinds of genes be concurrently introduced into a somatic cell.
  • one kind of non-viral expression vector incorporating all these genes may be used.
  • several kinds of non-viral expression vectors may be used in combination as appropriate, so as to cover all the combinations of these genes.
  • non-viral expression vectors may be combined as appropriate, so as to cover all the combinations of these genes.
  • These non-viral expression vectors are preferably concurrently introduced-into a somatic cell.
  • An example of a preferable method is a method wherein one non-viral expression vector harboring an Oct family gene, a Klf family gene, and a Sox family gene, and one non-viral expression vector harboring a Myc family gene are introduced into a somatic cell concurrently or at different times; in this method, it is preferable that the two kinds of non-viral expression vectors be concurrently introduced into the somatic cell.
  • a combination of four kinds of genes consisting of an Oct3/4, Klf4, Sox2, and c-Myc, or an optionally chosen combination of three kinds or two kinds selected from among these four kinds of genes, preferably the combination or three kinds or two kinds of genes, wherein said combination does not contain c-Myc, can be used.
  • This preferred embodiment is hereinafter described specifically, to which the scope of the present invention is never limited.
  • (a1) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4, Sox2 and c-Myc, is introduced into a somatic cell.
  • (b1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4, Sox2 and c-Myc, and a second non-viral expression vector, more preferably a plasmid vector, harboring the remaining two kinds of genes selected from among Oct3/4, Klf4, Sox2 and c-Myc, are introduced into a somatic cell.
  • the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • a first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • a first non-viral expression vector more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2
  • a second non-viral expression vector more preferably a plasmid vector, harboring c-Myc
  • the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • (f1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector, more preferably a plasmid vector, harboring c-Myc and any one gene out of Oct3/4, Klf4 and Sox2 not contained in the first non-viral expression vector, are introduced into a somatic cell.
  • a first non-viral expression vector more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end
  • a second non-viral expression vector more preferably a plasmid vector, harboring c-Myc and any one gene out of Oct3/4, Klf4 and Sox2 not contained in the first non-viral expression vector
  • a first non-viral expression vector preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, or (iii) Oct3/4 and Sox2 in this order in the orientation from the 5′ to 3′ end
  • the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • (g1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector, more preferably a plasmid vector, harboring c-Myc are introduced into a somatic cell.
  • a first non-viral expression vector more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end
  • a second non-viral expression vector more preferably a plasmid vector, harboring c-Myc
  • the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • the method of (f1) or (g1) can be preferably used when the somatic cell is derived from mouse.
  • a viral vector e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like
  • retroviral vector e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like
  • L-Myc in (a1) to (f2) above, L-Myc can be used in place of c-Myc.
  • (a2) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2, is introduced into a somatic cell.
  • (b2) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end are introduced into a somatic cell.
  • (c2) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2, and a second non-viral expression vector, more preferably a plasmid vector, harboring the remaining one kind of gene selected from among Oct3/4, Klf4 and Sox2, are introduced into a somatic cell.
  • a first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • (d2) A method wherein a first non-viral expression vector; more preferably a plasmid Vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector, more preferably a plasmid vector, harboring any one gene out of Oct3/4, Klf4 and Sox2 not contained in the first non-viral expression vector are introduced into a somatic cell.
  • a first non-viral expression vector more preferably a plasmid Vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end
  • a second non-viral expression vector more preferably a plasmid vector, harboring any one gene out of Oct3/4, Klf4 and Sox2 not contained in the first non-viral expression vector are introduced into a somatic cell.
  • a first non-viral expression vector preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, or (iii) Oct3/4 and Sox2 in this order in the orientation from the 5′ to 3′ end
  • the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • the method of (b2) or (d2) can be preferably used when the somatic cell is derived from mouse.
  • a viral vector e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like
  • retroviral vector e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like
  • a viral vector can also be used in place of the non-viral vector.
  • (a3) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2, is introduced into a somatic cell.
  • (b3) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, or (iii) Oct3/4 and Sox2 in this order in the orientation from the 5′ to 3′ end, is introduced into a somatic cell.
  • the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • the method of (b3) can be preferably used when the somatic cell is derived from mouse.
  • a viral vector e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like
  • retroviral vector e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like
  • the first, second and third non-viral expression vectors can be concurrently introduced into a somatic cell.
  • a first non-viral expression vector more preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, (iii) Oct3/4 and Sox2 or (iv) Sox2 and Klf4 in this order in the orientation from the 5′ to 3′ end
  • a second non-viral expression vector more preferably a plasmid vector, harboring the remaining one kind of gene selected from among Oct3/4, Klf4 and Sox2
  • a third non-viral expression vector more preferably a plasmid vector, harboring c-Myc, Lin28 and Nanog genes in this order in the orientation from the 5′ to 3′ end are introduced into a somatic cell.
  • a gene encoding a factor that induces cell immortalization such as TERT, SV40 large T antigen, HPV16 E6, HPV16 E7 or Bmil, is further combined with the two, three, four or six genes mentioned above, it can be preferably incorporated into another non-viral expression vector.
  • a plurality of genes e.g., Oct family gene, Klf family gene, and Sox family gene
  • these genes can preferably be inserted into the non-viral expression vector with an intervening sequence enabling polycistronic expression.
  • an intervening sequence enabling polycistronic expression it is possible to more efficiently express a plurality of genes incorporated in one kind of non-viral expression vector.
  • Useful sequences enabling polycistronic expression include, for example, the 2A sequence of foot-and-mouth disease virus (SEQ ID NO:61, sometimes referred to as FMDV 2A-self-processing sequence) (PLoS ONE 3, e2532, 2008; Stem Cells 25, 1707, 2007), IRES sequence and the like, preferably the 2A sequence.
  • the present invention also provides a use of the 2A sequence for preparing a non-viral expression vector for iPS cell induction, harboring two or more kinds of reprogramming factors.
  • the number of repeats of the manipulation to introduce a non-viral expression vector into a somatic cell is not particularly limited, as far as the effect of the present invention of reprogramming a somatic cell to produce an induced pluripotent stem cell can be accomplished, the transfection can be performed once or more optionally chosen times (e.g., once to 10 times, once to 5 times or the like).
  • the transfection can be performed once or more optionally chosen times (e.g., once to 10 times, once to 5 times or the like), preferably the transfection can be repeatedly performed twice or more (e.g., 3 times or 4 times).
  • the time interval is exemplified by, but not limited to, 12 hours to 1 week, preferably 12 hours to 4 days, for example, 1 day to 3 days.
  • iPS cell induced pluripotent stem cell
  • iPS cell refers to a cell possessing properties similar to that of ES cells, more specifically including undifferentiated cells reprogrammed from somatic cells possessing pluripotency and proliferating (self-renewal) capability. It should be noted, however, that this term must not be construed as limiting in any sense, and must be construed in the broadest sense.
  • a method of preparing an induced pluripotent stem cell by means of hypothetical nuclear reprogramming factors is described in WO2005/80598 (in this publication, the term ES-like cell is used), and a method of isolating an induced pluripotent stem cell is also described specifically.
  • WO2007/69666 discloses specific examples of reprogramming factors and methods of somatic cell reprogramming using the same. Therefore, it is desirable that in embodying the present invention, those skilled in the art refer to these publications.
  • a regulatory sequence required for transcription e.g., promoter, enhancer, and/or terminator and the like
  • promoter, enhancer, and/or terminator and the like is preferably operably linked to the gene in the non-viral expression vector.
  • a DNA sequence exhibiting transcription activity in somatic cells can be used, and the promoter can be chosen as appropriate according to animal species and kind of somatic cell.
  • useful promoters that can be expressed in mammalian cells include a promoter of the IE (immediate early) gene of cytomegalovirus (human CMV), initial promoter of SV40, promoter of retrovirus, metallothionein promoter, heat shock promoter, SR ⁇ promoter and the like.
  • An enhancer of the IE gene of human CMV may be used along with a promoter.
  • a useful promoter is the CAG promoter (comprising cytomegalovirus enhancer, chicken ⁇ -actin promoter and ⁇ -globin gene polyA signal site).
  • the non-viral expression vector may incorporate a DNA sequence that allows the autonomous replication of the expression vector in a mammalian somatic cell.
  • An example of the DNA sequence is the SV40 replication origin.
  • the non-viral expression vector is preferably an expression vector autonomously replicable outside the chromosome, and the non-viral expression vector is preferably one that is not integrated in the chromosome. More preferable examples include plasmid vectors. Examples of the plasmid vector include, but are not limited to, Escherichia coli -derived plasmids (ColE-series plasmids such as pBR322, pUC18, pUC19, pUC118, pUC119, and pBluescript, and the like), Actinomyces -derived plasmids (pIJ486 and the like), Bacillus subtilis -derived plasmids (e.g., pUB110, pSH19 and others), yeast-derived plasmids (YEp13, YEp 24, Ycp50 and the like) and the like, as well as artificial plasmid vectors and the like.
  • non-viral expression vectors examples include, but are not limited to, pCMV6-XL3 (OriGene Technologies Inc.), EGFP-C1 (Clontech), pGBT-9 (Clontech), pcDNAI (FUNAKOSHI), pcDMB (FUNAKOSHI), pAGE107 (Cytotechnology, 3,133, 1990), pCDM8 (Nature, 329, 840, 1987), pcDNAI/AmP (Invitrogen), pREP4 (Invitrogen), pAGE103 (J. Blochem., 101, 1307, 1987), pAGE210 and the like.
  • the non-viral expression vector may incorporate a selectable marker as required.
  • selectable marker include genes that are deficient in the host cell, such as the dihydrofolate reductase (DHFR) gene or the Schizosaccaromyces pombe TPI gene, and genes for resistance to drugs such as ampicillin, kanamycin, tetracycline, chloramphenicol, neomycin, or hygromycin.
  • DHFR dihydrofolate reductase
  • Schizosaccaromyces pombe TPI genes for resistance to drugs such as ampicillin, kanamycin, tetracycline, chloramphenicol, neomycin, or hygromycin.
  • the non-viral expression vector can preferably contain a sequence enabling the excicion of transgenes, such as loxP sequence (Chang et al., STEM CELLS Published Online: 12 Feb. 2009 (doi: 10.1002/stem.39)), piggyback transposon (Kaji et al., Nature advance online publication 1 Mar.
  • a method of ligating a gene that encodes a reprogramming factor, a promoter, an enhancer, and/or a terminator and the like, used in the present invention, in an appropriate order to construct a non-viral expression vector capable of expressing the reprogramming factor in the somatic cell, is obvious to those skilled in the art.
  • the genes may be incorporated in one non-viral expression vector.
  • two or more kinds of non-viral expression vectors incorporating different genes may be used.
  • one non-viral expression vector incorporating two or more kinds of genes and a non-viral expression vector incorporating one or more kind genes different therefrom can be combined as appropriate.
  • Any method of expression vector introduction into an animal cell available to those skilled in the art can be used to introduce a non-viral expression vector into a somatic cell.
  • useful methods include the use of a transfection reagent such as the FuGENE 6 transfection reagent (Roche), the use of a microporator, the electroporation method, the calcium phosphate method, the lipofection method, the DEAE-dextran-mediated transfection method, the transfection method, the microinjection method, the cationic lipid-mediated transfection method, and the like. Nucleofection can also be used to introduce a gene. These methods may be used in combination.
  • the expression vector may be introduced into the somatic cell being cultured on feeder cells, and may be introduced only into the somatic cell.
  • the feeder cells used may be those for cultivation of embryonic stem cells; for example, primary culture fibroblasts from a 14- to 15-day mouse embryo, STO (fibroblast-derived cell line) and the like, treated with a chemical agent such as mitomycin C or exposed to radiation, and the like can be used.
  • culturing a somatic cell incorporating a non-viral expression vector under appropriate conditions it is possible to allow nuclear reprogramming to progress autonomically, and to produce an induced pluripotent stem cell from the somatic cell.
  • the step of culturing a somatic cell incorporating a non-viral expression vector to obtain an induced pluripotent stem cell can be performed in the same manner as a conventional method using a retrovirus; for example, this can be achieved as described in publications such as Cell, 126, pp. 1-14, 2006; Cell, 131, pp. 1-12, 2007; and Science, 318, pp. 1917-1920, 2007.
  • the cell culture density after expression vector introduction be set at a level lower than that for ordinary animal cell culture. For example, it is preferable to continue the cultivation at a cell density of 10,000 to 100,000 cells, preferably about 50,000 cells per cell culture dish. Any medium can be used for the cultivation, chosen as appropriate by those skilled in the art; for example, in producing a human induced pluripotent stem cell, it is sometimes preferable to use a medium suitable of human ES cell culture. Regarding the choice of medium and culturing conditions, the aforementioned publications serve for references.
  • the resulting induced pluripotent stem cells can be identified using various markers characteristic of undifferentiated cells; means for this identification are also described in the aforementioned publications specifically and in detail.
  • Various media allowing the maintenance of undifferentiated state and pluripotency of ES cells or media not allowing the maintenance of these properties are known in the art; by using appropriate media in combination, an induced pluripotent stem cell can be isolated efficiently.
  • the differentiation potential and proliferation potential of the isolated induced pluripotent stem cells are easily confirmable for those skilled in the art by utilizing a method of identification in common use for ES cells.
  • a colony of induced pluripotent stem cells is obtained; it is possible to identify the presence of an induced pluripotent stem cell on the basis of the shape of the colony.
  • mouse induced pluripotent stem cells form raised colonies
  • human induced pluripotent stem cells form flat colonies
  • the shapes of these colonies are extremely similar to those of mouse ES cell and human ES cell colonies, respectively; therefore, it is possible for those skilled in the art to identify the resulting induced pluripotent stem cell on the basis of the shape of the colony.
  • somatic cell having a gene incorporating a marker gene such as GFP downstream of a promoter of gene specifically expressing in ES cells, it is possible to identify an induced pluripotent stem cell if the cell becomes positive for the marker (GFP).
  • GFP marker
  • Somatic cells to be reprogrammed by the method of the present invention refers to any cells except totipotent and pluripotent cells such as early embryos and ES cells, and the choice thereof is not limited.
  • somatic cells in the fetal stage neonatal somatic cells and mature somatic cells may be used.
  • somatic cells derived from mammals, including humans are used; more preferably human- or mouse-derived somatic cells are used.
  • tissue stem cells such as nerve stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells
  • tissue progenitor cells or (3) differentiated cells such as lymphocytes, epithelial cells, muscle cells, fibroblasts (dermal cells and the like), hair cells, liver cells, and gastromucosal cells
  • somatic stem cells separated from a patient to be treated or from another person sharing the same type of HLA as that of the patient; for example, somatic cells involved in disease and somatic cells involved in disease treatment and the like can be used.
  • tPS cell establishment efficiency improvers include, but are not limited to, histone deacetylase (HDAC) inhibitors [e.g., valproic acid (VPA) (Nat.
  • HDAC histone deacetylase
  • VPA valproic acid
  • low-molecular inhibitors such as trichostatin A, sodium butyrate, MC 1293, and M344, nucleic acid-based expression inhibitors such as siRNA and shRNA against HDAC (e.g., HDAC1 siRNA Smartpool® (Millipore), HuSH 29mer shRNA Constructs against HDAC1 (OriGene) and the like), and the like], G9a histone methyltransferase inhibitors [e.g., low-molecular inhibitors such as BIX-01294 (Cell Stem Cell, 2: 525-528 (2008)), nucleic acid-based expression inhibitors such as siRNA and shRNA against G9a (e.g., G9a siRNA (human) (Santa Cruz Biotechnology) and the like) and the like], L-channel calcium agonist (e.g., Bayk8644) (Cell Stem Cell, 3, 568-574 (2008)), UTF1 (Cell Stem Cell, 3, 4
  • the nucleic acid-based expression inhibitors may be in the form of expression vectors harboring a DNA that encodes siRNA or shRNA.
  • the DNA that encodes siRNA or shRNA may be inserted into a non-viral expression vector of the present invention, together with reprogramming factors.
  • the induced pluripotent stem cell produced by the method of the present invention is not subject to limitations concerning the use thereof, and can be used for all types of studies and investigations with the use of ES cells and for the treatment of diseases using ES cells, in place of ES cells.
  • desired differentiated cells e.g., nerve cells, myocardial cells, blood cells and the like
  • stem cell therapy by autologous cell transplantation can be accomplished.
  • the use of an induced pluripotent stem cell of the present invention is not limited to the above-described particular embodiment.
  • the present invention also provides a non-viral expression vector for use in the above-described method of producing an induced pluripotent stem cell, i.e., a non-viral expression vector (preferably a plasmid vector) incorporating at least one gene that encodes a reprogramming factor.
  • a non-viral expression vector preferably a plasmid vector
  • the structure of the vector is as described in detail in the section of a method of producing an induced pluripotent stem cell of the present invention.
  • An example is a non-viral expression vector incorporating an Oct family gene, a Klf family gene, and a Sox family gene, preferably incorporated in this order in the orientation from the 5′ to 3′ end.
  • a more preferable example is a non-viral expression vector incorporating these genes with an intervening sequence enabling polycistronic expression, particularly preferably a non-viral expression vector wherein OCT3/4, Klf4 and Sox 2 are incorporated with an intervening sequence enabling polycistronic expression, preferably FMDV 2A-self-processing sequence, in this order in the orientation from the 5′ to 3′ end.
  • the present invention provides an induced pluripotent stem cell wherein transgenes are not integrated into the genome. Since such iPS cell reduces a risk causing tumorigenesis in tissues or organs differentiated therefrom and/or disturbance (e.g., disruption or activation) of an endogenous gene, it can preferably be used for regenerative medicine such as cell transplantation therapy.
  • the present invention provides an induced pluripotent stem cell wherein transgenes are integrated into the genome in the form of plasmid.
  • iPS cell can reduce a risk causing tumorigenesis in tissues or organs differentiated therefrom as compared to an iPS cell induced by retroviral infection.
  • transgenes can be excised from the genome as necessary using a Cre/loxP system (Chang et al., 2009 (supra)) or a piggyback transposon vector and piggyback transposon (Kaji et al., 2009 (supra); Woltjen et al., 2009 (supra)) or tetracycline dependent gene induction.
  • a Cre recombinase or transposase for the excision can be introduced into and expressed in the iPS cell using a plasmid vector or adenoviral vector.
  • Tet-repressor protein or mutated Tet-repressor protein is concomitantly expressed.
  • mice having a Nanog reporter were used as an experimental system (Okita et al. Nature, Vol. 448, pp. 313-317, 2007). These mice were prepared by incorporating EGFP and a puromycin resistance gene into the Nanog gene locus of a BAC (bacterial artificial chromosome) purchased from BACPAC Resources. The mouse Nanog gene is expressed specifically in pluripotent cells such as ES cells and early embryos. Mouse iPS cells positive for this reporter have been shown to possess a differentiation potential nearly equivalent to that of ES cells. These Nanog reporter mice were mated with Fbx15 reporter mice (Tokuzawa et al. Mol Cell Biol, Vol. 23, 2699-2708 (2003)), whereby mutant mice having both the Nanog reporter and the Fbx15 reporter were generated.
  • Fbx15 reporter mice Yamazawa et al. Mol Cell Biol, Vol. 23, 2699-2708 (2003)
  • the plasmid used for reprogramming was prepared by treating pCX-EGFP (a plasmid supplied by Dr. Masaru Okabe at Osaka University: FEBS Letters, 407, 313-319, 1997) with EcoRI, and inserting a construct wherein the coding regions of Oct3/4, Sox2, and Klf4 (all mouse-derived genes) are ligated via the 2A sequence of foot-and-mouth disease virus in the order of Oct3/4, Klf4, and Sox2, in place of EGFP (pCX-2A-mOKS; FIG. 2 ).
  • pCX-2A-mOKS a plasmid with the coding region of c-Myc inserted thereinto was prepared (pCX-c-Myc; FIG. 2 ).
  • oligonucleotides comprising the 2A sequence of foot-and-mouth disease virus (SEQ ID NO:61), upstream restriction endonuclease sites (XbaI and BglII), and downstream restriction endonuclease sites (BspHI, Mfel and PstI), were annealed and inserted into pBluescript II KS ( ⁇ ) vector digested with the XbaI and PstI (pBS-2A).
  • a mouse cDNA that encodes Oct3/4 or Klf4 was amplified by PCR, the translation termination codon was replaced with a BamHI site, and each cDNA was cloned into pCR2.1. Subsequently, the cDNAs of Oct3/4 and Klf4 were ligated with pBS-2A using an appropriate restriction endonuclease to yield pBS-Oct3/4-2A and pBS-Klf4-2A.
  • Klf4-2A was inserted into pBS-Oct3/4-2A in frame using an appropriate restriction endonuclease, whereby pBS-Oct3/4-2A-Klf4-2A was produced.
  • the resulting Oct3/4-2A-Klf4-2A construct was ligated with a cDNA of Sox2 having a translation termination codon in frame, using an appropriate restriction endonuclease.
  • MEF Fibroblasts
  • the culture medium was replaced with a fresh supply (DMEM/10% FCS) and an expression vector (pCX-2A-mOKS) was introduced as described above; the day after, the culture medium was replaced with an ES cell culture medium (DMEM (Nacalai Tesque) supplemented with 15% fetal calf serum, 2 mM L-glutamine (Invitrogen), 100 ⁇ M non-essential amino acids (Invitrogen), 100 ⁇ M 2-mercaptoethanol (Invitrogen), 50 U/mL penicillin (Invitrogen) and 50 mg/mL streptomycin (Invitrogen)), and an expression vector (pCX-c-Myc) was introduced using the FuGene6 transfection reagent as described above.
  • DMEM Necalai Tesque
  • the medium was replaced with an ES cell culture medium.
  • the MEF culture medium was removed, and the cells were washed by the addition of PBS 2 mL.
  • PBS 0.25% Trypsin/1 mM EDTA (Invitrogen) was added, and the reaction was carried out at 37° C. for about 5 minutes.
  • an ES cell culture medium was added, the cells were suspended, and 1 ⁇ 10 5 (Exp432A) or 2 ⁇ 10 5 (Exp432B) cells were sown onto a 100 mm dish with feeder cells sown thereto previously.
  • the feeder cells used were SNL cells that had been treated with mitomycin C to terminate their cell division.
  • iPS cells obtained as described in Example 2 were subcutaneously transplanted to nude mice. All clones tested (440A-3, -4, -8 and -10) produced tumors, which included a broad range of cell types, including cells derived from all the three germ layers ( FIG. 7 ). Furthermore, iPS cells without integration were injected into ICR mouse blastocysts. Judging from the coat colors, adult chimeras were obtained from all clones injected (440A-3, -4, -6, -8, -9 and -10) ( FIG. 8 ). In these chimeric mice, PCR analysis did not detect the integration of any of the transgenes ( FIG. 9 ).
  • Human dental pulp stem cells (clone name; DP31, PCT/JP2008/068320, J. Dent. Res., 87(7):676-681 (2008)) were used as an experimental system.
  • the DP31 was allowed to express the mouse ecotropic virus receptor Slc7a1 gene using a lentivirus as described in Cell, 131, 861-872 (2007). These cells were cultured using the MSCGM bullet kit (Lonza).
  • the plasmids used for reprogramming were prepared from pCX-EGFP (supplied by Dr. Masaru Okabe at Osaka University, FEBS Letters, 407, 313-319, 1997) in the same manner as Example 1. Specifically, the pCX-EGFP was treated with EcoRI, and a construct with the coding regions of SOX2 and KLF4 ligated via the 2A sequence of foot-and-mouth disease virus therein was inserted in place of EGFP, whereby the plasmid pCX-hSK was prepared.
  • a plasmid with c-Myc, Lin28, and Nanog ligated via the 2A sequence pCX-hMLN
  • a plasmid with the OCT3/4 coding region inserted therein pCX-hOCT3/4
  • a plasmid with the SV40 Large T antigen inserted therein pCX-SV40LT
  • the DP31 cultured in a 100 mm dish was washed with PBS, 0.25% Trypsin/1 mM EDTA (Invitrogen) was added, and the reaction was carried out at 37° C. for about 5 minutes. After cells rose, MSCGM was added, the cells were suspended, and 6 ⁇ 10 5 cells were recovered in a 15 mL tube. The cells were centrifuged at 800 rpm for 5 minutes; after the supernatant was removed, and the expression plasmids were introduced using the Human Dermal Fibroblast Nucleofector Kit (Amaxa).
  • the amounts of plasmids used were 0.5 ⁇ g for pCX-hOCT3/4, 1.0 ⁇ g for pCX-hSK, 1.5 ⁇ g for pCX-hMLN, and 0.5 ⁇ g for pCX-SV40LT.
  • the cells were sown to a 6-well plate.
  • the cells were again washed with PBS, 0.25% Trypsin/1 mM EDTA (Invitrogen) was added, and the reaction was carried out at 37° C. for about 5 minutes. After cells rose, MSCGM was added, the cells were suspended, and 1 ⁇ 10 5 cells were sown onto a 100 mm dish with feeder cells sown thereto previously.
  • the feeder cells used were SNL cells that had been treated with mitomycin C to terminate their cell division. Thereafter, until a colony began to be observed, the medium was replaced with a fresh supply every two days.
  • the medium used was prepared by mixing equal volumes of a primate ES cell culture medium (ReproCELL) supplemented with MSCGM and bFGF (4 ng/mL), respectively. Colonization began around day 19, confirming the establishment of human iPS cell ( FIG. 15 ).
  • fetal human HDF (Cell applications, INC) was transfected with the same seven kinds of genes as described above. After the transfection, the cells were cultured using a primate ES cell culture medium (ReproCELL) supplemented with 4 ng/ml recombinant human bFGF (WAKO). MSTO cells served as feeder cells. Photographs of cells on day 31 after transfection (5 clones: 203A-1 to 203A-5, of which 203A-4 was picked up as a negative control) are shown in FIG. 16 , and photographs of cells in the 2nd subculture are shown in FIG. 17 . The 203A-1 to 203A-3 and 203A-5 clones exhibited a typical ES cell-like morphology, confirming the establishment of human iPS cells.
  • Dental pulp stem cells DP31 used in Example 4, were transfected with six kinds of genes, excluding the SV40 Large T antigen (pCX-hSK, pCX-hMLN, pCX-hOCT3/4), in the same manner as Example 4. Photographs of cells on day 35 after the transfection (5 clones: 217A-1 to -4 and -6) are shown in FIG. 19 . Photographs of cells in the 2nd subculture are shown in FIG. 20 . All clones exhibited a typical ES cell-like morphology, confirming the establishment of human iPS cells.
  • HDF cell line derived from a 6-year-old Japanese female (HDF-120; JCRB) was allowed to express the Slc7a1 gene.
  • the resulting cells (HDF-120-Slc) were transfected with the aforementioned six kinds of genes and an shRNA against p53 (shRNA2: SEQ ID NO:62) (vectors introduced: pCX-hOCT3/4, pCX-hSK, pCX-hMLN-shp53).
  • Each of pCX-hOCT3/4 (0.5 ⁇ g), pCX-hSK (1.0 ⁇ g), and pCX-hMLN-shp53 (1.5 ⁇ g) was electrically introduced into 6.0 ⁇ 10 5 cells of HDF-120-Slc using Microporator (100 ⁇ L tip, 1600 V, 10 ms, 3 times). Ten days later, each vector was once again electrically introduced under the same conditions, and the cells were sown onto MSTO (100 mm dish). These cells were cultured using DMEM/10% FCS until day 10, thereafter using a primate ES cell culture medium (ReproCELL) supplemented with 4 ng/ml recombinant human bFGF (WAKO).
  • ReproCELL a primate ES cell culture medium supplemented with 4 ng/ml recombinant human bFGF
  • Photographs of cells on day 35 after the first electroporation are shown in FIG. 22 .
  • Photographs of cells after passage culture are shown in FIG. 23 .
  • a typical ES cell-like morphology was exhibited, confirming the establishment of human iPS cells.
  • Genomic-PCR analysis demonstrated the integration of the transgenes (lane 279A-2 in FIG. 24 ).
  • Expression vectors separately incorporating the four kinds of genes Oct3/4, Klf4, Sox2 and c-Myc were introduced into MEF cells derived from a Nanog reporter mouse (Okita et al. Nature, Vol. 448, pp. 313-317, 2007) per the protocol in Example 2.
  • Nanog reporter MEF cells were sown onto a gelatin-coated 6-well plate (1.3 ⁇ 10 5 cells/well), and transfected with each of pCX-Oct4 (0.37 ⁇ g), pCX-Sox2 (0.36 ⁇ g), pCX-Klf4 (0.39 ⁇ g), and pCX-c-Myc (0.38 ⁇ g) using FuGene6 on days 1, 3, 5, and 7.
  • 1 ⁇ 10 6 cells (1.0) or 0.2 ⁇ 10 6 cells (0.2) were sown onto MSTO-PH or gelatin (100-mm dish), and colonies were selected on day 25. Photographs of cells after the selection are shown in FIG. 25 .
  • mice iPS cell clones established (497A-1 to A-5) were subjected to genomic-PCR analysis. The results are shown in FIG. 26 . Both 497A-2 and 497A-5 were shown to be iPS cells without integration of any of the exogenous genes.
  • a highly safe induced pluripotent stem cell from, for example, a patient's somatic cell.
  • the cells obtained by differentiating the induced pluripotent stem cell e.g., myocardial cells, insulin-producing cells, nerve cells and the like
  • myocardial cells, insulin-producing cells, nerve cells and the like can be safely used for stem cell transplantation therapy for a broad range of diseases, including heart failure, insulin-dependent diabetes, Parkinson's disease and spinal injury.

Abstract

A method of producing an induced pluripotent stem cell includes introducing into a somatic cell one or more non-viral expression vectors. The vectors include one or more of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, and Nanog gene. The somatic cell is then cultured in a medium that supports pluripotent stem cells. At least a portion of the one or more introduced non-viral expression vectors is not substantially integrated in the chromosome.

Description

    TECHNICAL FIELD
  • The present invention relates to a method of reprogramming a somatic cell and producing an induced pluripotent stem cell.
  • BACKGROUND ART
  • Established from human or mouse early embryos, embryonic stem cells (ES cells) are capable of being cultured for a long time while maintaining their potential for differentiating into all types of cells found in a living organism. With this feature, human ES cells are expected to serve for cell transplantation therapies for many diseases, including Parkinson's disease, juvenile diabetes, and leukemia. However, ES cell transplantation poses the problem of causing rejections as with organ transplantation. Additionally, not a few people oppose the use of ES cells established with the destruction of a human embryo, from an ethical viewpoint.
  • If the dedifferentiation of a patient's somatic cells is induced to establish cells possessing pluripotency and proliferating capability similar to those of an ES cell (herein these cells are referred to as “induced pluripotent stem cells” (iPS cells), and sometimes referred to as “embryonic stem cell-like cells” or “ES-like cells”), the established cells will be useful as ideal pluripotent cells that do not pose the problems of rejections and ethical issues. In recent years, it has been reported that iPS cells can be produced from mouse and human differentiated cells, arousing great attention (International Patent Application Publication No. WO2007/69666; Cell, 126, pp. 663-676, 2006; Cell, 131, pp. 861-872, 2007; Science, 318, pp. 1917-1920, 2007; Nature, 451, pp. 141-146, 2008).
  • All these methods comprise the step of introducing a plurality of particular nuclear reprogramming factors (e.g., in Cell, 126, pp. 1-14, 2006, 4 factors are used: Oct3/4, Sox2, Klf4, and c-Myc) into a somatic cell to achieve reprogramming, which step involves the use of a retrovirus or a lentivirus for the purpose of introducing the genes that encode the nuclear reprogramming factors into a somatic cell efficiently. However, since gene delivery using a viral vector involves safety issues, there is a demand for developing a method of producing iPS cells without using a viral vector.
  • SUMMARY OF THE INVENTION Technical Problem
  • It is an object of the present invention to provide a method of producing an iPS cell by reprogramming a somatic cell without using a viral vector such as a retrovirus.
  • Solution to Problem
  • The present inventors extensively investigated to solve the problems described above, and found that an iPS cell can be produced by introducing genes that encode reprogramming factors into a somatic cell by means of a non-viral expression vector such as a plasmid vector, and that a safe iPS cell can be obtained from a somatic cell by the method. The present invention has been developed on the basis of these findings.
  • Accordingly, the present invention provides a method of producing an induced pluripotent stem cell, comprising the step of introducing at least one kind of non-viral expression vector incorporating at least one gene that encodes a reprogramming factor into a somatic cell.
  • In a preferred embodiment, the present invention provides the above-described method wherein the vectors are non-viral expression vectors autonomously replicable outside a chromosome; and the above-described method wherein the vector is a plasmid vector.
  • In another preferred embodiment, the present invention provides the above-described method wherein the gene that encodes a reprogramming factor is one of genes selected by a method of screening for nuclear reprogramming factors described in WO 2005/80598 or a combination of a plurality of such genes; and the above-described method wherein the gene that encodes a reprogramming factor is one or more kinds of genes selected from the group consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, and the Nanog gene, preferably a combination of two kinds of genes, more preferably a combination of three kinds of genes, particularly preferably a combination of four or more kinds of genes.
  • More preferable combinations are (a) a combination of two kinds of genes consisting of an Oct family gene and a Sox family gene; (b) a combination of three kinds of genes consisting of an Oct family gene, a Klf family gene, and a Sox family gene; (c) a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene; (d) a combination of four kinds of genes consisting of an Oct family gene, a Sox family gene, a Lin family gene, and the Nanog gene; (e) a combination of six kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, and the Nanog gene; and the like. Furthermore, it is also preferable to include the TERT gene and/or the SV40 Large T antigen gene in the combination. As the case may be, it is preferable to exclude Klf family genes.
  • Particularly preferred combinations thereof are a combination of two kinds of genes consisting of Oct3/4 and Sox2; a combination of three kinds of genes consisting of Oct3/4, Klf4, and Sox2; a combination of four kinds of genes consisting of Oct3/4, Klf4, Sox2, and c-Myc; a combination of four kinds of genes consisting of Oct3/4, Sox2, Lin28, and Nanog; and a combination of six kinds of genes consisting of Oct3/4, Klf4, Sox2, c-Myc, Lin28, and Nanog. It is also preferable to include the TERT gene and/or the SV40 Large T antigen gene in these combinations. As the case may be, it is preferable to exclude Klf4.
  • In another preferred embodiment, the present invention provides the above-described method wherein the number of kinds of non-viral expression vectors introduced into a somatic cell is 1, 2, 3, or 4; the above-described method wherein the genes that encode reprogramming factors are a combination of three kinds of genes consisting of an Oct family gene, a Klf family gene, and a Sox family gene, and these genes are incorporated in one kind of non-viral expression vector; the above-described method wherein the genes that encode nuclear reprogramming factors are a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene, and the Oct family gene, the Klf family gene, and the Sox family gene are incorporated in one kind of non-viral expression vector; the above-described method wherein the Oct family gene, the Klf family gene, and the Sox family gene are incorporated in one kind of non-viral expression vector in this order in the orientation from the 5′ to 3′ end; and the above-described method wherein the Oct family gene, the Klf family gene, and the Sox family gene are incorporated in one kind of non-viral expression vector with an intervening sequence enabling polycistronic expression.
  • In another preferred embodiment, the present invention provides the above-described method wherein two or more kinds of the above-described non-viral expression vectors are concurrently introduced into a somatic cell; the above-described method wherein the genes that encode reprogramming factors are a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene, and a first non-viral expression vector incorporating three or less kinds of genes selected from among the four kinds of genes, and a second non-viral expression vector incorporating the remaining gene(s) out of the four kinds of genes are concurrently introduced into a somatic cell; the above-described method wherein the three or less kinds of genes are an Oct family gene, a Klf family gene, and a Sox family gene, and the remaining gene is a Myc family gene; the above-described method wherein the three or less kinds of genes are Oct3/4, Klf4, and Sox2, and the remaining gene is c-Myc; and the above-described method wherein introduction of the non-viral expression vector into a somatic cell is repeatedly performed twice or more.
  • In a particularly preferred embodiment, the present invention provides the above-described method wherein a first non-viral expression vector harboring Oct3/4, Klf4, and Sox2, and a second non-viral expression vector harboring c-Myc are introduced into a somatic cell; the above-described method wherein a first non-viral expression vector harboring Oct3/4, Klf4, and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector harboring c-Myc are introduced into a somatic cell; the above-described method wherein Oct3/4, Klf4, and Sox2 are ligated in this order in the orientation from the 5′ to 3′ end with an intervening sequence enabling polycistronic expression and inserted into the first non-viral expression vector; the above-described method wherein the first non-viral expression vector and the second non-viral expression vector are concurrently introduced into a somatic cell; and the above-described method wherein the introduction is repeatedly performed twice or more. Also provided is the above-described method wherein whole or prat of the at least one non-viral expression vector introduced is substantially not integrated in the chromosome.
  • In another preferred embodiment, the present invention provides the above-described method wherein the somatic cell is a somatic cell of a mammal, including a human, preferably a human or mouse somatic cell, particularly preferably a human somatic cell; the above-described method wherein the somatic cell is a fetal human cell or a somatic cell derived from an adult human; and the above-described method wherein the somatic cell is a somatic cell collected from a patient.
  • In another aspect, the present invention provides an induced pluripotent stem cell that can be obtained by the above-described method. In a preferred embodiment, the present invention also provides an induced pluripotent stem cell wherein all or some of the at least one non-viral expression vector introduced is substantially not integrated in the chromosome.
  • Also provided are the above-described induced pluripotent stem cell wherein the somatic cell is a somatic cell of a mammal, including a human, preferably a human or mouse somatic cell, particularly preferably a human somatic cell; the above-described induced pluripotent stem cell wherein the somatic cell is a fetal human cell or a somatic cell derived from an adult human; and the above-described induced pluripotent stem cell wherein the somatic cell is a somatic cell collected from a patient.
  • A non-viral expression vector, preferably a plasmid vector, for use in the above-described method of producing an induced pluripotent stem cell, incorporating at least one gene that encodes a reprogramming factor, is also provided by the present invention.
  • A somatic cell induced and differentiated from the above-described induced pluripotent stem cell is also provided by the present invention.
  • The present invention also provides a stem cell therapy comprising the step of transplanting to a patient a somatic cell obtained by differentiation induction of an induced pluripotent stem cell obtained by the above-described method using a somatic cell separated from the patient.
  • The present invention further provides a method of evaluating the physiological activities and toxicities of compounds, drugs, poisonous substances and the like using various cells obtained by differentiation induction of an induced pluripotent stem cell obtained by the above-described method.
  • Advantageous Effects of Invention
  • Produced without using a vector to be integrated into a chromosome, such as a retrovirus, the induced pluripotent stem cell provided by the present invention is advantageous in that tumorigenesis and other problems do not arise in the somatic cells and tissues obtained by differentiating the induced pluripotent stem cell. In a preferred embodiment of the present invention, in the induced pluripotent stem cell produced by the method of the present invention, all or some of the at least one non-viral expression vector introduced is episomally present, substantially not integrated in the chromosome. Therefore, the method of the present invention makes it possible to prepare a highly safe induced pluripotent stem cell from, for example, a patient's somatic cell, and the cells obtained by differentiating this cell (e.g., myocardial cells, insulin-producing cells, or nerve cells and the like) can be safely used for stem cell transplantation therapies for a broad range of diseases, including heart failure, insulin-dependent diabetes, Parkinson's disease and spinal injury.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 shows a time course protocol for transfecting a somatic cell (MEF) with Oct3/4, Klf4, Sox2, and c-Myc using plasmids according to the method of the present invention, results of seven independent tests (left photographs, 432A-1 to 432A-7: cell density 1×106 cells/100 mm dish) and results of another test (right photographs, 432B-1: cell density 2×105 cells/100 mm dish). The lowermost panels in the center show control results (no transfection). In FIG. 1, the Phase columns show phase-contrast images, and the GFP columns show GFP-positive colonies.
  • FIG. 2 shows an expression plasmid for iPS cell production. Three kinds of cDNAs that encode Oct3/4, Klf4, and Sox2 were ligated in this order with sequence encoding the 2A peptide as intervening sequence, and inserted into the pCX plasmid (pCX-2A-mOKS). Furthermore, a cDNA of c-Myc was inserted into pCX (pCX-c-Myc). The bald lines show the amplification regions used in the PCR analysis for detecting plasmid integration in the genome (FIG. 6).
  • FIG. 3 shows the time schedules for IFS cell induction using plasmids. The solid arrows indicate the time points of transfection of the respective plasmids.
  • FIG. 4 shows the morphology of non-virus mediated IFS cells established. The upper panels show phase-contrast images, and the lower panels show GFP-positive colonies (scale bar=200 μm).
  • FIG. 5 shows results of PCR analysis for the genetic expression of ES cell markers, obtained using total RNAs isolated from ES cells, iPS cells induced using retroviruses (clone 20D-17: Nature, 448, pp. 313-317, 2007), iPS cells induced using plasmids (clones 440A-3, 4, 7, 8, 10 and 11; clone 432A-1), and MEF cells.
  • FIG. 6 shows the detection of plasmid integration by PCR. Genomic DNAs were extracted from a C57BL/6 mouse, iPS cell induced using retroviruses (clone 20D-17), iPS cells induced with plasmids (clone 432A-1; clones 440A-1 to 11) and MEF cells, and analyzed by PCR using the primers shown in FIGS. 2, 13 and 14. In the PCR for O-1, K and M, the bands derived from endogenous genes are indicated by the outlined arrowheads, and the bands derived from integrated plasmids are indicated by the solid arrowheads. For the Fbx15 reporter, the lower band indicates wild-type alleles, and the upper band indicates knocked-in alleles.
  • FIG. 7 shows results of teratoma formation. iPS cells without integration of plasmids (clones 440A-3, -4, and -8) were subcutaneously transplanted to nude mice. Four weeks later, tumors were resected and stained with hematoxylin and eosin. Shown from above are the results for gut-like epithelial tissue, epidermal tissue, striated muscles, and nerve tissue, respectively (scale bar=50 μm).
  • FIG. 8 shows chimeric mice derived from iPS cells without integration (clones 440A-3 and -8).
  • FIG. 9 shows the detection of integration of plasmids by PCR. Genomic DNAs were extracted from an ICR mouse, iPS cell (clone 432A-1), and chimeric mice derived from iPS cells induced using plasmids (clone 432A-1; clones 0.440A-3, 8), and the O-1, K and M regions shown in FIG. 2 were amplified by PCR. The bands derived from endogenous genes are indicated by the outlined arrowheads, and the bands derived from integrated plasmids are indicated by the solid arrowheads. The presence of the Nanog reporter and Fbx15 reporter was also detected by PCR.
  • FIG. 10 shows the probes used in Southern blot analysis and the positions of the restriction endonuclease recognition sites. E indicates EcoRI, and B indicates BamHI.
  • FIG. 11 shows results of Southern blot analysis. Genomic DNAs (6 μg) were extracted from RF8 ES cells and iPS cells (clones 440A-3, 4, 7, 8, 10, and 11; clone 432A-1), and cleaved with BamHI and EcoRI. A mixture of the pCX-2A-mOKS and pCX-c-Myc plasmids (each 20 pg) served for control. The outlined arrowheads indicate the bands derived from endogenous genes, and the solid arrowhead indicates the band derived from the Oct3/4 pseudogene (estimated size 2049 bp) on chromosome 3. The arrows indicate the bands derived from transgenes. Although the identities of the many bands observed in clone 432A-1 are unclear, this may suggest the integration of multiple transgenes. The GFP probe was used to detect Nanog reporter alleles.
  • FIG. 12 shows results of SSLP analysis. On genomic DNAs (each 50 ng) from C57BL/6 mouse, RFB ES cell, iPS cells without integration (clones 440A-3 to 11) and MEF cells, SSLP analysis was performed. These iPS cells derive from a mixture of five MEF cell clones ( clones 1, 2, 3, 5, and 6).
  • FIGS. 13 and 14 show the primers used for PCR in Examples 1 to 3.
  • FIG. 15 shows a time course protocol for transfecting human dental pulp stem cells with Oct3/4, Klf4, Sox2, c-Myc, Lin28, Nanog and the SV40 Large T antigen using plasmids according to the method of the present invention, and 16 independent IFS cell colonies.
  • FIGS. 16 and 17 show photographs of IFS cells established from fetal HDF (5 clones: 203A-1 to 203A-5, of which 203A-4 was picked up as a negative control) on day 31 after transfection (FIG. 16) and in the 2nd subculture (FIG. 17).
  • FIG. 18 shows the results of genomic-PCR analysis of 5 iPS cell clones (203A-1 to 203A-5).
  • FIGS. 19 and 20 show photographs of IFS cells established from human dental pulp stem cells (5 clones: 217A-1 to -4 and -6) on day 35 after transfection (FIG. 19) and in the 2nd subculture (FIG. 20).
  • FIG. 21 shows the results of genomic-PCR analysis of 5 iPS cell clones (217A-1 to -4 and -6).
  • FIGS. 22 and 23 show photographs of IFS cells established from young female HDF (2 clones: 279A-1 and -2) on day 35 after the first electroporation (FIG. 22) and clone 279A-2 after passage culture (FIG. 23; the right panel is a closeup picture of the boxed area in the left panel).
  • FIG. 24 shows the results of genomic-PCR analysis of iPS cell clone 279A-2 demonstrating the integration of the transgenes.
  • FIG. 25 shows photographs of iPS cells (8 clones: 497A-1 to A-8) after the selection (colonies were selected on day 25 after transfection). The upper panels show phase-contrast images, and the lower panels show GFP-positive colonies.
  • FIG. 26 shows the results of genomic-PCR analysis of 5 iPS cell clones (497A-1 to A-5). In 497A-2 and 497A-5, no exogenous gene was not integrated into the genome.
  • DESCRIPTION OF EMBODIMENTS
  • The method of the present invention is intended to produce an induced pluripotent stem cell, comprising the step of introducing at least one kind of non-viral expression vector incorporating at least one gene that encodes a reprogramming factor into a somatic cell. The non-viral expression vector is preferably an expression vector autonomously replicable outside a chromosome, more preferably a plasmid expression vector.
  • As an example of a means for identifying a nuclear reprogramming factor, a nuclear reprogramming factor screening method described in WO 2005/80598 can be utilized. All disclosures therein are incorporated herein by reference. Those skilled in the art are able to screen for nuclear reprogramming factors, and to utilize them for the method of the present invention, by referring to the aforementioned publication. It is also possible to identify nuclear reprogramming factors using a method modified or altered from the above-described screening method.
  • Some examples of combinations of genes that encode reprogramming factors are disclosed in WO2007/69666. All disclosures therein are incorporated herein by reference. Those skilled in the art are able to choose genes that can suitably be used in the method of the present invention as appropriate by referring to the aforementioned publication. Other examples of combinations of genes that encode reprogramming factors are given in Science, 318, pp. 1917-1920, 2007, WO2008/118820 and the like. Therefore, those skilled in the art are able to understand the diversity of combinations of genes that encode nuclear reprogramming factors; by utilizing a nuclear reprogramming factor screening method described in WO 2005/80598, appropriate combinations of genes other than the combinations described in WO2007/69666 and Science, 2007 (supra) can be utilized in the method of the present invention.
  • Preferable genes that encode reprogramming factors include one or more kind of genes selected from the group consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, and the Nanog gene, preferably a combination of two kinds of genes, more preferably of three kinds of genes, and particularly preferably of four kinds of genes.
  • Examples of Oct family genes, Klf family genes, Sox family genes, and Myc family genes are given in WO2007/69666. Likewise, for Lin family genes, those skilled in the art are likewise able to extract a family gene. For example, as examples of Lin family genes, Lin28 and Lin28B may be included.
  • More preferable combinations include, but are not limited to,
  • (a) a combination of two kinds of genes consisting of an Oct family gene and a Sox family gene;
    (b) a combination of three kinds of genes consisting of an Oct family gene, a Klf family gene, and a Sox family gene;
    (c) a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene;
    (d) a combination of four kinds of genes consisting of an Oct family gene, a Sox family gene, a Lin family gene, and the Nanog gene;
    (e) a combination of six kinds of genes consisting of an Oct family gene, a Sox family gene, a Klf family gene, a Myc family gene, a Lin family gene, and the Nanog gene; and the like.
  • All these genes are present in common in mammals, including humans. Genes derived from optionally chosen mammals (e.g., humans, mice, rats, bovines, sheep, horses, monkeys) can be used in the present invention. In addition to wild-type gene, mutant genes whose translation products have several (e.g., 1 to 10, preferably 1 to 6, more preferably 1 to 4, more preferably 1 to 3, particularly preferably 1 or 2) amino acids substituted, inserted, and/or deleted, and possess a function similar to that of the wild type gene product, can also be utilized. For example, as c-Myc genes, the wild type, a gene encoding stable type mutant (T58A) and the like may be used. The same applies to other gene products.
  • In addition to the aforementioned genes, a gene that encodes a factor that induces cell immortalization may further be combined. As disclosed in WO2007/69666, for example, the TERT gene, and one or more kind of genes selected from the group consisting of the following genes: SV40 Large T antigen, HPV16 E6, HPV16 E7, and Bmil, can be used singly, or in combination as appropriate.
  • Examples of preferable combinations include:
  • (f) a combination of five kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, and the TERT gene;
    (g) a combination of five kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, and the SV40 Large T antigen gene; (h) a combination of six kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, the TERT gene, and the SV40 Large T antigen gene; and (i) a combination of seven kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, a Myc family gene, a Lin family gene, the Nanog gene, and the TERT gene or the SV40 Large T antigen gene.
  • As required, the Klf family gene may be excluded from the aforementioned combinations.
  • Furthermore, in addition to the aforementioned genes, one or more kind of genes selected from the group consisting of Fbx15, ERas, ECAT15-2, Tcl1, and β-catenin may be combined, and/or one or more kind of genes selected from the group consisting of ECAT1, Esg1, Dnmt3L, ECAT8, Gdf3, Sox15, ECAT15-1, Fth117, Sal14, Rex1, UTF1, Stella, Stat3, and Grb2 may also be combined. These combinations are specifically described in WO2007/69666.
  • If one or more kind of these genes are already expressed in the somatic cell to be reprogrammed, the gene(s) can be excluded from the genes to be introduced. When one or more kind of these genes are introduced into a somatic cell to be reprogrammed using a vector to be integrated into a chromosome, such as a retrovirus, the remaining one or more genes can be introduced using a non-viral expression vector according to the method of the present invention. Alternatively, when one or more kind of the gene products of these genes are introduced into a nucleus by means of fused protein or nuclear microinjection, the remaining one or more genes can be introduced using a non-viral expression vector according to the method of the present invention.
  • Particularly preferable combinations of genes are,
  • (1) a combination of two kinds of genes consisting of Oct3/4 and Sox2;
    (2) a combination of three kinds of genes consisting of Oct3/4, Klf4, and Sox2;
    (3) a combination of four kinds of genes consisting of Oct3/4, Klf4, Sox2, and c-Myc;
    (4) a combination of four kinds of genes consisting of Oct3/4, Sox2, Lin28, and Nanog;
    (5) a combination of five kinds of genes consisting of Oct3/4, Sox2, c-Myc, TERT, and SV40 Large T antigen;
    (6) a combination of six kinds of genes consisting of Oct3/4, Klf4, Sox2, c-Myc, TERT, and SV40 Large T antigen;
    (7) a combination of six kinds of genes consisting of Oct3/4, Klf4, c-Myc, Sox2, Lin28, and Nanog;
    (8) a combination of seven kinds of genes consisting of Oct3/4, Klf4, c-Myc, Sox2, Lin28, Nanog, and TERT or SV40 Large T antigen,
    and the like.
  • In addition to the aforementioned genes, a gene that encodes a factor that induces cell immortalization may further be combined. As disclosed in WO2007/69666, for example, one kind or more of genes selected from the group consisting of the TERT gene, and the following genes: HPV16 E6, HPV16 E7, and Bmil, can be used singly, or in combination as appropriate.
  • When reprogramming is performed using nerve stem cells endogenously expressing Sox2 and c-Myc, or the like as a somatic cell source, a combination of two kinds of genes consisting of Oct3/4 and Klf4, or a combination of two kinds of genes consisting of Oct3/4 and c-Myc (see Nature, Published online, 29 Jun. 2008, p 1-5 (doi:10.1038/nature07061)) can also be mentioned.
  • In the combinations (3), (5), (6), and (7) above, L-Myc can be used in place of c-Myc.
  • It should be noted that combinations of genes are not limited thereto. Additionally, the scope of the present invention includes a method wherein one or more genes selected from among the above-described genes are introduced into a somatic cell using a non-viral expression vector, and the remaining gene or gene product is introduced into the somatic cell by another means. For example, it is also possible to introduce one or more genes selected from among the above-described genes into a somatic cell using a non-viral expression vector, and to introduce the remaining gene into the somatic cell using a viral vector such as retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector.
  • When two or more kinds of genes that encode reprogramming factors are introduced into a somatic cell using non-viral expression vectors, some of the two or more kinds of genes to be introduced can be introduced into a somatic cell at a time different from that for other genes, or all kinds of genes to be introduced can be concurrently introduced into a somatic cell; however, it is preferable that all genes to be introduced be concurrently introduced into a somatic cell. When two or more kinds of different non-viral expression vectors are used to introduce two or more kinds of genes, all kinds of non-viral expression vectors can be concurrently introduced into a somatic cell; this represents a preferred embodiment of the present invention.
  • In the method of the present invention, as genes that encode reprogramming factors, for example, a combination of four kinds of genes consisting of an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene can be used. A combination of three kinds of genes consisting of an Oct family gene, a Klf family gene, and a Sox family gene, or a combination of two kinds of genes selected from among the aforementioned three kinds of genes can also be used.
  • In the method of the present invention, it is preferable that the above-described four kinds, three kinds, or two kinds of genes be concurrently introduced into a somatic cell. To introduce the above-described four kinds, three kinds, or two kinds of genes, one kind of non-viral expression vector incorporating all these genes may be used. Alternatively, several kinds of non-viral expression vectors may be used in combination as appropriate, so as to cover all the combinations of these genes. When several kinds of non-viral expression vectors are used, it is preferable that preferably two or three kinds, more preferably two kinds of non-viral expression vectors be used. It is preferable that these non-viral expression vectors be concurrently introduced into a somatic cell.
  • If the number of genes introduced exceeds four kinds, several kinds of non-viral expression vectors may be combined as appropriate, so as to cover all the combinations of these genes. When several kinds of non-viral expression vectors are used, it is preferable that preferably two to five kinds, more preferably two to four kinds, more preferably three or four of non-viral expression vectors be used. These non-viral expression vectors are preferably concurrently introduced-into a somatic cell.
  • An example of a preferable method is a method wherein one non-viral expression vector harboring an Oct family gene, a Klf family gene, and a Sox family gene, and one non-viral expression vector harboring a Myc family gene are introduced into a somatic cell concurrently or at different times; in this method, it is preferable that the two kinds of non-viral expression vectors be concurrently introduced into the somatic cell. In another preferred embodiment, it is also possible to use a method wherein one non-viral expression vector harboring an Oct family gene, a Klf family gene, a Sox family gene, and a Myc family gene is introduced into a somatic cell.
  • In a preferred embodiment of the present invention, in a combination of four kinds of genes consisting of an Oct3/4, Klf4, Sox2, and c-Myc, or an optionally chosen combination of three kinds or two kinds selected from among these four kinds of genes, preferably the combination or three kinds or two kinds of genes, wherein said combination does not contain c-Myc, can be used. This preferred embodiment is hereinafter described specifically, to which the scope of the present invention is never limited.
  • (a1) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4, Sox2 and c-Myc, is introduced into a somatic cell.
    (b1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4, Sox2 and c-Myc, and a second non-viral expression vector, more preferably a plasmid vector, harboring the remaining two kinds of genes selected from among Oct3/4, Klf4, Sox2 and c-Myc, are introduced into a somatic cell. Preferably, the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
    (c1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring three kinds of genes selected from among Oct3/4, Klf4, Sox2 and c-Myc, and a second non-viral expression vector, more a preferably a plasmid vector, harboring the remaining one kind of gene selected from among Oct3/4, Klf4, Sox2 and c-Myc, are introduced into a somatic cell. Preferably, the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
    (d1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2, and a second non-viral expression vector, more preferably a plasmid vector, harboring the remaining one kind of gene selected from among Oct3/4, Klf4 and Sox2, and c-Myc, are introduced into a somatic cell. Preferably, the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
    (e1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2, and a second non-viral expression vector, more preferably a plasmid vector, harboring c-Myc, are introduced into a somatic cell. Preferably, the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
    (f1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector, more preferably a plasmid vector, harboring c-Myc and any one gene out of Oct3/4, Klf4 and Sox2 not contained in the first non-viral expression vector, are introduced into a somatic cell. More specifically, a first non-viral expression vector, preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, or (iii) Oct3/4 and Sox2 in this order in the orientation from the 5′ to 3′ end can be used; the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
    (g1) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector, more preferably a plasmid vector, harboring c-Myc are introduced into a somatic cell. Preferably, the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • The method of (f1) or (g1) can be preferably used when the somatic cell is derived from mouse.
  • In (b1) to (f2) above, for either one of the first non-viral expression vector and the second non-viral expression vector, a viral vector (e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like) can be used in place of the non-viral expression vector.
  • In another preferred embodiment of the present invention, in (a1) to (f2) above, L-Myc can be used in place of c-Myc.
  • In still another preferred embodiment, a combination of three kinds of genes consisting of Oct3/4, Klf4 and Sox2 can be used. This preferred embodiment is hereinafter described specifically, to which the scope of the present invention is never limited.
  • (a2) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2, is introduced into a somatic cell.
    (b2) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end are introduced into a somatic cell.
    (c2) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2, and a second non-viral expression vector, more preferably a plasmid vector, harboring the remaining one kind of gene selected from among Oct3/4, Klf4 and Sox2, are introduced into a somatic cell. Preferably, the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
    (d2) A method wherein a first non-viral expression vector; more preferably a plasmid Vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2 in this order in the orientation from the 5′ to 3′ end, and a second non-viral expression vector, more preferably a plasmid vector, harboring any one gene out of Oct3/4, Klf4 and Sox2 not contained in the first non-viral expression vector are introduced into a somatic cell. More specifically, a first non-viral expression vector, preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, or (iii) Oct3/4 and Sox2 in this order in the orientation from the 5′ to 3′ end can be used, and the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • The method of (b2) or (d2) can be preferably used when the somatic cell is derived from mouse.
  • In (c2) or (d2) above, for either one of the first non-viral expression vector and the second non-viral expression vector, a viral vector (e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like) can also be used in place of the non-viral vector.
  • In still another preferred embodiment of the present invention, a combination of two kinds of genes selected from among Oct3/4, Klf4 and Sox2 can be used. This preferred embodiment is hereinafter described specifically, to which the scope of the present invention is never limited.
  • (a3) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2, is introduced into a somatic cell.
    (b3) A method wherein one kind of non-viral expression vector, more preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, or (iii) Oct3/4 and Sox2 in this order in the orientation from the 5′ to 3′ end, is introduced into a somatic cell.
    (c3) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring one kind of gene selected from among Oct3/4, Klf4 and Sox2, and a second non-viral expression vector, more preferably a plasmid vector, harboring any one gene out of Oct3/4, Klf4 and Sox2 not contained in the first non-viral expression vector, are introduced into a somatic cell. Preferably, the first non-viral expression vector and the second non-viral expression vector can be concurrently introduced into a somatic cell.
  • The method of (b3) can be preferably used when the somatic cell is derived from mouse.
  • In (c3) above, for either one of the first non-viral expression vector and the second non-viral expression vector, a viral vector (e.g., retroviral vector, lentiviral vector, adenoviral vector, adeno-associated viral vector, Sendai viral vector or the like) can be used in place of the non-viral vector.
  • In still another preferred embodiment of the present invention, a combination of six kinds of gene's selected from among Oct3/4, Klf4, Sox2, c-Myc, Lin28 and Nanog can be used. This preferred embodiment is hereinafter described specifically, to which the scope of the present invention is never limited.
  • (a4) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring two kinds of genes selected from among Oct3/4, Klf4 and Sox2, a second non-viral expression vector, more preferably a plasmid vector, harboring the remaining one kind of gene selected from among Oct3/4, Klf4 and Sox2, and a third non-viral expression vector, more preferably a plasmid vector, harboring c-Myc, Lin28 and Nanog genes are introduced into a somatic cell. Preferably, the first, second and third non-viral expression vectors can be concurrently introduced into a somatic cell.
    (b4) A method wherein a first non-viral expression vector, more preferably a plasmid vector, harboring (i) Oct3/4 and Klf4, (ii) Klf4 and Sox2, (iii) Oct3/4 and Sox2 or (iv) Sox2 and Klf4 in this order in the orientation from the 5′ to 3′ end, a second non-viral expression vector, more preferably a plasmid vector, harboring the remaining one kind of gene selected from among Oct3/4, Klf4 and Sox2, and a third non-viral expression vector, more preferably a plasmid vector, harboring c-Myc, Lin28 and Nanog genes in this order in the orientation from the 5′ to 3′ end are introduced into a somatic cell.
  • When a gene encoding a factor that induces cell immortalization, such as TERT, SV40 large T antigen, HPV16 E6, HPV16 E7 or Bmil, is further combined with the two, three, four or six genes mentioned above, it can be preferably incorporated into another non-viral expression vector.
  • In the context above, when a plurality of genes (e.g., Oct family gene, Klf family gene, and Sox family gene) are incorporated in one kind of non-viral expression vector, these genes can preferably be inserted into the non-viral expression vector with an intervening sequence enabling polycistronic expression. By using an intervening sequence enabling polycistronic expression, it is possible to more efficiently express a plurality of genes incorporated in one kind of non-viral expression vector. Useful sequences enabling polycistronic expression include, for example, the 2A sequence of foot-and-mouth disease virus (SEQ ID NO:61, sometimes referred to as FMDV 2A-self-processing sequence) (PLoS ONE 3, e2532, 2008; Stem Cells 25, 1707, 2007), IRES sequence and the like, preferably the 2A sequence. More specifically, when a non-viral expression vector harboring (i) Oct3/4, Klf4 and Sox2, (ii) Oct3/4 and Klf4, (iii) Klf4 and Sox2, (iv) Oct3/4 and Sox2, (v) Sox2 and Klf4 or (vi) c-Myc, Lin28 and Nanog in this order in the orientation from the 5′ to 3′ end is constructed, it is preferable to insert the 2A sequence between these genes. Accordingly, the present invention also provides a use of the 2A sequence for preparing a non-viral expression vector for iPS cell induction, harboring two or more kinds of reprogramming factors.
  • The number of repeats of the manipulation to introduce a non-viral expression vector into a somatic cell is not particularly limited, as far as the effect of the present invention of reprogramming a somatic cell to produce an induced pluripotent stem cell can be accomplished, the transfection can be performed once or more optionally chosen times (e.g., once to 10 times, once to 5 times or the like). When two or more kinds of non-viral expression vectors are introduced into a somatic cell, it is preferable that these all kinds of non-viral expression vectors be concurrently introduced into a somatic cell; however, even in this case, the transfection can be performed once or more optionally chosen times (e.g., once to 10 times, once to 5 times or the like), preferably the transfection can be repeatedly performed twice or more (e.g., 3 times or 4 times).
  • When the transfection is repeated twice or more, the time interval is exemplified by, but not limited to, 12 hours to 1 week, preferably 12 hours to 4 days, for example, 1 day to 3 days.
  • As used herein, the term “induced pluripotent stem cell (iPS cell)” refers to a cell possessing properties similar to that of ES cells, more specifically including undifferentiated cells reprogrammed from somatic cells possessing pluripotency and proliferating (self-renewal) capability. It should be noted, however, that this term must not be construed as limiting in any sense, and must be construed in the broadest sense. A method of preparing an induced pluripotent stem cell by means of hypothetical nuclear reprogramming factors is described in WO2005/80598 (in this publication, the term ES-like cell is used), and a method of isolating an induced pluripotent stem cell is also described specifically. WO2007/69666 discloses specific examples of reprogramming factors and methods of somatic cell reprogramming using the same. Therefore, it is desirable that in embodying the present invention, those skilled in the art refer to these publications.
  • In addition to the gene that encodes a reprogramming factor, a regulatory sequence required for transcription (e.g., promoter, enhancer, and/or terminator and the like) is preferably operably linked to the gene in the non-viral expression vector.
  • As the promoter, a DNA sequence exhibiting transcription activity in somatic cells can be used, and the promoter can be chosen as appropriate according to animal species and kind of somatic cell. Examples of useful promoters that can be expressed in mammalian cells include a promoter of the IE (immediate early) gene of cytomegalovirus (human CMV), initial promoter of SV40, promoter of retrovirus, metallothionein promoter, heat shock promoter, SRα promoter and the like. An enhancer of the IE gene of human CMV may be used along with a promoter. A useful promoter is the CAG promoter (comprising cytomegalovirus enhancer, chicken β-actin promoter and β-globin gene polyA signal site).
  • The non-viral expression vector may incorporate a DNA sequence that allows the autonomous replication of the expression vector in a mammalian somatic cell. An example of the DNA sequence is the SV40 replication origin.
  • The non-viral expression vector is preferably an expression vector autonomously replicable outside the chromosome, and the non-viral expression vector is preferably one that is not integrated in the chromosome. More preferable examples include plasmid vectors. Examples of the plasmid vector include, but are not limited to, Escherichia coli-derived plasmids (ColE-series plasmids such as pBR322, pUC18, pUC19, pUC118, pUC119, and pBluescript, and the like), Actinomyces-derived plasmids (pIJ486 and the like), Bacillus subtilis-derived plasmids (e.g., pUB110, pSH19 and others), yeast-derived plasmids (YEp13, YEp 24, Ycp50 and the like) and the like, as well as artificial plasmid vectors and the like.
  • Examples of easily available non-viral expression vectors include, but are not limited to, pCMV6-XL3 (OriGene Technologies Inc.), EGFP-C1 (Clontech), pGBT-9 (Clontech), pcDNAI (FUNAKOSHI), pcDMB (FUNAKOSHI), pAGE107 (Cytotechnology, 3,133, 1990), pCDM8 (Nature, 329, 840, 1987), pcDNAI/AmP (Invitrogen), pREP4 (Invitrogen), pAGE103 (J. Blochem., 101, 1307, 1987), pAGE210 and the like.
  • The non-viral expression vector may incorporate a selectable marker as required. Examples of the selectable marker include genes that are deficient in the host cell, such as the dihydrofolate reductase (DHFR) gene or the Schizosaccaromyces pombe TPI gene, and genes for resistance to drugs such as ampicillin, kanamycin, tetracycline, chloramphenicol, neomycin, or hygromycin.
  • While a non-viral expression vector such as plasmid vector introduced into a somatic cell is typically not integrated into the genome of the cell, under selection pressure for iPS cell induction, increased integration efficiency of non-viral expression vector may be observed due to the necessity of stable expression of reprogramming factors. Accordingly, when the iPS cells of interest are intended to use for regenerative medicine and the like, the non-viral expression vector can preferably contain a sequence enabling the excicion of transgenes, such as loxP sequence (Chang et al., STEM CELLS Published Online: 12 Feb. 2009 (doi: 10.1002/stem.39)), piggyback transposon (Kaji et al., Nature advance online publication 1 Mar. 2009 (doi:10.1038/nature07864); Woltjen et al., Nature advance online publication 1 Mar. 2009 (doi:10.1038/nature07863)) and tetracycline responsive element in promoter region (Tet-OnR & Tet-Off R Gene Expression Systems, Clontech).
  • A method of ligating a gene that encodes a reprogramming factor, a promoter, an enhancer, and/or a terminator and the like, used in the present invention, in an appropriate order to construct a non-viral expression vector capable of expressing the reprogramming factor in the somatic cell, is obvious to those skilled in the art.
  • When two or more kinds of genes that encode reprogramming factors are used, the genes may be incorporated in one non-viral expression vector. Alternatively, two or more kinds of non-viral expression vectors incorporating different genes may be used. In the latter case, one non-viral expression vector incorporating two or more kinds of genes and a non-viral expression vector incorporating one or more kind genes different therefrom can be combined as appropriate.
  • Any method of expression vector introduction into an animal cell available to those skilled in the art can be used to introduce a non-viral expression vector into a somatic cell. Examples of useful methods include the use of a transfection reagent such as the FuGENE 6 transfection reagent (Roche), the use of a microporator, the electroporation method, the calcium phosphate method, the lipofection method, the DEAE-dextran-mediated transfection method, the transfection method, the microinjection method, the cationic lipid-mediated transfection method, and the like. Nucleofection can also be used to introduce a gene. These methods may be used in combination.
  • In introducing a non-viral expression vector into a somatic cell, the expression vector may be introduced into the somatic cell being cultured on feeder cells, and may be introduced only into the somatic cell. To increase expression vector introduction efficiency, the latter method is sometimes suitable. The feeder cells used may be those for cultivation of embryonic stem cells; for example, primary culture fibroblasts from a 14- to 15-day mouse embryo, STO (fibroblast-derived cell line) and the like, treated with a chemical agent such as mitomycin C or exposed to radiation, and the like can be used.
  • By culturing a somatic cell incorporating a non-viral expression vector under appropriate conditions, it is possible to allow nuclear reprogramming to progress autonomically, and to produce an induced pluripotent stem cell from the somatic cell. The step of culturing a somatic cell incorporating a non-viral expression vector to obtain an induced pluripotent stem cell can be performed in the same manner as a conventional method using a retrovirus; for example, this can be achieved as described in publications such as Cell, 126, pp. 1-14, 2006; Cell, 131, pp. 1-12, 2007; and Science, 318, pp. 1917-1920, 2007. In producing a human induced pluripotent stem cell, it is sometimes desirable that the cell culture density after expression vector introduction be set at a level lower than that for ordinary animal cell culture. For example, it is preferable to continue the cultivation at a cell density of 10,000 to 100,000 cells, preferably about 50,000 cells per cell culture dish. Any medium can be used for the cultivation, chosen as appropriate by those skilled in the art; for example, in producing a human induced pluripotent stem cell, it is sometimes preferable to use a medium suitable of human ES cell culture. Regarding the choice of medium and culturing conditions, the aforementioned publications serve for references.
  • The resulting induced pluripotent stem cells can be identified using various markers characteristic of undifferentiated cells; means for this identification are also described in the aforementioned publications specifically and in detail. Various media allowing the maintenance of undifferentiated state and pluripotency of ES cells or media not allowing the maintenance of these properties are known in the art; by using appropriate media in combination, an induced pluripotent stem cell can be isolated efficiently. The differentiation potential and proliferation potential of the isolated induced pluripotent stem cells are easily confirmable for those skilled in the art by utilizing a method of identification in common use for ES cells. When the resulting induced pluripotent stem cell is proliferated under appropriate conditions, a colony of induced pluripotent stem cells is obtained; it is possible to identify the presence of an induced pluripotent stem cell on the basis of the shape of the colony. For example, it is known that mouse induced pluripotent stem cells form raised colonies, whereas human induced pluripotent stem cells form flat colonies, and the shapes of these colonies are extremely similar to those of mouse ES cell and human ES cell colonies, respectively; therefore, it is possible for those skilled in the art to identify the resulting induced pluripotent stem cell on the basis of the shape of the colony. When reprogramming is performed using a somatic cell having a gene incorporating a marker gene such as GFP downstream of a promoter of gene specifically expressing in ES cells, it is possible to identify an induced pluripotent stem cell if the cell becomes positive for the marker (GFP).
  • “Somatic cells” to be reprogrammed by the method of the present invention refers to any cells except totipotent and pluripotent cells such as early embryos and ES cells, and the choice thereof is not limited. For example, as well as somatic cells in the fetal stage, neonatal somatic cells and mature somatic cells may be used. Preferably, somatic cells derived from mammals, including humans, are used; more preferably human- or mouse-derived somatic cells are used. Specifically, (1) tissue stem cells (somatic stem cells) such as nerve stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells, (2) tissue progenitor cells, or (3) differentiated cells such as lymphocytes, epithelial cells, muscle cells, fibroblasts (dermal cells and the like), hair cells, liver cells, and gastromucosal cells can be mentioned. When an induced pluripotent stem cell is used to treat a disease, it is desirable to use somatic cells separated from a patient to be treated or from another person sharing the same type of HLA as that of the patient; for example, somatic cells involved in disease and somatic cells involved in disease treatment and the like can be used.
  • In the present invention, to increase the efficiency of induced pluripotent stem cell establishment, addition to the introduction of a non-viral expression vector of the present invention, various establishment efficiency improvers may be introduced or added. Examples of tPS cell establishment efficiency improvers include, but are not limited to, histone deacetylase (HDAC) inhibitors [e.g., valproic acid (VPA) (Nat. Biotechnol., 26(7): 795-797 (2008)), low-molecular inhibitors such as trichostatin A, sodium butyrate, MC 1293, and M344, nucleic acid-based expression inhibitors such as siRNA and shRNA against HDAC (e.g., HDAC1 siRNA Smartpool® (Millipore), HuSH 29mer shRNA Constructs against HDAC1 (OriGene) and the like), and the like], G9a histone methyltransferase inhibitors [e.g., low-molecular inhibitors such as BIX-01294 (Cell Stem Cell, 2: 525-528 (2008)), nucleic acid-based expression inhibitors such as siRNA and shRNA against G9a (e.g., G9a siRNA (human) (Santa Cruz Biotechnology) and the like) and the like], L-channel calcium agonist (e.g., Bayk8644) (Cell Stem Cell, 3, 568-574 (2008)), UTF1 (Cell Stem Cell, 3, 475-479 (2008)), Wnt Signaling (e.g., soluble Wnt3a) (Cell Stem Cell, 3, 132-135 (2008)), 2i/LIF (2i is an inhibitor of mitogen-activated protein kinase signaling and glycogen synthase kinase-3; PloS Biology, 6(10), 2237-2247 (2008)), p53 inhibitors (e.g., siRNA and shRNA against p53 (Cell Stem Cell, 3, 475-479 (2008)) and the like. The nucleic acid-based expression inhibitors may be in the form of expression vectors harboring a DNA that encodes siRNA or shRNA. In this case, the DNA that encodes siRNA or shRNA may be inserted into a non-viral expression vector of the present invention, together with reprogramming factors.
  • The induced pluripotent stem cell produced by the method of the present invention is not subject to limitations concerning the use thereof, and can be used for all types of studies and investigations with the use of ES cells and for the treatment of diseases using ES cells, in place of ES cells. For example, by treating an induced pluripotent, stem cell obtained from a somatic cell collected from a patient by the method of the present invention with retinoic acid, a growth factor such as EGF, or glucocorticoid and the like, desired differentiated cells (e.g., nerve cells, myocardial cells, blood cells and the like) can be induced to form an appropriate tissue. By returning the differentiated cell or tissue thus obtained to the patient, stem cell therapy by autologous cell transplantation can be accomplished. It should be noted that the use of an induced pluripotent stem cell of the present invention is not limited to the above-described particular embodiment.
  • The present invention also provides a non-viral expression vector for use in the above-described method of producing an induced pluripotent stem cell, i.e., a non-viral expression vector (preferably a plasmid vector) incorporating at least one gene that encodes a reprogramming factor. The structure of the vector is as described in detail in the section of a method of producing an induced pluripotent stem cell of the present invention.
  • An example is a non-viral expression vector incorporating an Oct family gene, a Klf family gene, and a Sox family gene, preferably incorporated in this order in the orientation from the 5′ to 3′ end. A more preferable example is a non-viral expression vector incorporating these genes with an intervening sequence enabling polycistronic expression, particularly preferably a non-viral expression vector wherein OCT3/4, Klf4 and Sox 2 are incorporated with an intervening sequence enabling polycistronic expression, preferably FMDV 2A-self-processing sequence, in this order in the orientation from the 5′ to 3′ end.
  • Since a non-viral expression vector such as plasmid vector introduced into a somatic cell is typically not integrated into the genome of the cell, in a preferred embodiment, the present invention provides an induced pluripotent stem cell wherein transgenes are not integrated into the genome. Since such iPS cell reduces a risk causing tumorigenesis in tissues or organs differentiated therefrom and/or disturbance (e.g., disruption or activation) of an endogenous gene, it can preferably be used for regenerative medicine such as cell transplantation therapy.
  • However, under selection pressure for iPS cell induction, increased integration efficiency of non-viral expression vector can be observed due to the necessity of stable expression of reprogramming factors. Therefore, in another preferred embodiment, the present invention provides an induced pluripotent stem cell wherein transgenes are integrated into the genome in the form of plasmid. Such iPS cell can reduce a risk causing tumorigenesis in tissues or organs differentiated therefrom as compared to an iPS cell induced by retroviral infection. In addition, the transgenes can be excised from the genome as necessary using a Cre/loxP system (Chang et al., 2009 (supra)) or a piggyback transposon vector and piggyback transposon (Kaji et al., 2009 (supra); Woltjen et al., 2009 (supra)) or tetracycline dependent gene induction. A Cre recombinase or transposase for the excision can be introduced into and expressed in the iPS cell using a plasmid vector or adenoviral vector. In the case of using tetracycline dependent gene induction, Tet-repressor protein or mutated Tet-repressor protein is concomitantly expressed.
  • EXAMPLES
  • The present invention is hereinafter described in more detail by means of the following Examples, which, however, are not to be construed as limiting the scope of the invention.
  • Example 1
  • Mice having a Nanog reporter were used as an experimental system (Okita et al. Nature, Vol. 448, pp. 313-317, 2007). These mice were prepared by incorporating EGFP and a puromycin resistance gene into the Nanog gene locus of a BAC (bacterial artificial chromosome) purchased from BACPAC Resources. The mouse Nanog gene is expressed specifically in pluripotent cells such as ES cells and early embryos. Mouse iPS cells positive for this reporter have been shown to possess a differentiation potential nearly equivalent to that of ES cells. These Nanog reporter mice were mated with Fbx15 reporter mice (Tokuzawa et al. Mol Cell Biol, Vol. 23, 2699-2708 (2003)), whereby mutant mice having both the Nanog reporter and the Fbx15 reporter were generated.
  • The plasmid used for reprogramming was prepared by treating pCX-EGFP (a plasmid supplied by Dr. Masaru Okabe at Osaka University: FEBS Letters, 407, 313-319, 1997) with EcoRI, and inserting a construct wherein the coding regions of Oct3/4, Sox2, and Klf4 (all mouse-derived genes) are ligated via the 2A sequence of foot-and-mouth disease virus in the order of Oct3/4, Klf4, and Sox2, in place of EGFP (pCX-2A-mOKS; FIG. 2). Likewise, a plasmid with the coding region of c-Myc inserted thereinto was prepared (pCX-c-Myc; FIG. 2).
  • In preparing the construct of the 2A sequence and Oct3/4, Klf4, and Sox2 ligated together, first, sense and antisense oligonucleotides comprising the 2A sequence of foot-and-mouth disease virus (SEQ ID NO:61), upstream restriction endonuclease sites (XbaI and BglII), and downstream restriction endonuclease sites (BspHI, Mfel and PstI), were annealed and inserted into pBluescript II KS (−) vector digested with the XbaI and PstI (pBS-2A). Subsequently, a mouse cDNA that encodes Oct3/4 or Klf4 was amplified by PCR, the translation termination codon was replaced with a BamHI site, and each cDNA was cloned into pCR2.1. Subsequently, the cDNAs of Oct3/4 and Klf4 were ligated with pBS-2A using an appropriate restriction endonuclease to yield pBS-Oct3/4-2A and pBS-Klf4-2A. Subsequently, Klf4-2A was inserted into pBS-Oct3/4-2A in frame using an appropriate restriction endonuclease, whereby pBS-Oct3/4-2A-Klf4-2A was produced. Subsequently, the resulting Oct3/4-2A-Klf4-2A construct was ligated with a cDNA of Sox2 having a translation termination codon in frame, using an appropriate restriction endonuclease. Finally, the resulting Oct3/4-2A-Klf4-2A-Sox2-STOP construct, wherein the 2A sequences and Oct3/4, Klf4, and Sox2 were ligated together, was inserted into the EcoRI site of pCX-EGFP, whereby pCX-2A-mOKS was prepared.
  • Fibroblasts (MEF) were isolated from the aforementioned mutant mouse fetus (13.5 days after fertilization). Not expressing the Nanog gene, MEF does not express EGFP producing green fluorescence. As such, the MEFs were sown to a 6-well culture plate (Falcon), previously coated with 0.1% gelatin (Sigma), at 1.3×105 cells per well. The culture medium used being DMEM/10% FCS (DMEM (Nacalai Tesque) supplemented with 10% fetal calf serum), the MEFs were cultured at 37° C., 5% CO2. The following day, 4.5 μL of the FuGene6 transfection reagent (Roche) was added in 100 μL of Opti-MEM I Reduced-Serum Medium (Invitrogen), and the medium was allowed to stand at room temperature for 5 minutes. Thereafter, 1.5 μg of an expression vector (pCX-2A-mOKS) was added, and the medium was allowed to stand at room temperature for 15 minutes, after which the medium was added to a MEF culture medium. The following day, the medium was removed, and 1.5 μg of another expression vector (pCX-c-Myc) was introduced with the FuGene6 transfection reagent as described above.
  • The following day, the culture medium was replaced with a fresh supply (DMEM/10% FCS) and an expression vector (pCX-2A-mOKS) was introduced as described above; the day after, the culture medium was replaced with an ES cell culture medium (DMEM (Nacalai Tesque) supplemented with 15% fetal calf serum, 2 mM L-glutamine (Invitrogen), 100 μM non-essential amino acids (Invitrogen), 100 μM 2-mercaptoethanol (Invitrogen), 50 U/mL penicillin (Invitrogen) and 50 mg/mL streptomycin (Invitrogen)), and an expression vector (pCX-c-Myc) was introduced using the FuGene6 transfection reagent as described above.
  • The following day, the medium was replaced with an ES cell culture medium. On day 9 after sowing, the MEF culture medium was removed, and the cells were washed by the addition of PBS 2 mL. After the PBS was removed, 0.25% Trypsin/1 mM EDTA (Invitrogen) was added, and the reaction was carried out at 37° C. for about 5 minutes. After cells rose, an ES cell culture medium was added, the cells were suspended, and 1×105(Exp432A) or 2×105(Exp432B) cells were sown onto a 100 mm dish with feeder cells sown thereto previously. The feeder cells used were SNL cells that had been treated with mitomycin C to terminate their cell division.
  • Subsequently, the ES cell culture medium was replaced with a fresh supply every two days until a visible colony emerged; colonization began around day 17, and complete colonization was observed around day 24 (FIG. 1). The time schedule above is summarized in Exp432 in FIGS. 1 and 3.
  • The cells obtained became GFP-positive gradually, exhibited a morphology indistinguishable from that of mouse ES cells (432A-1 in FIG. 4), tested positive for various ES cell markers at similar levels as with ES cells (iPS-432A-1 in FIG. 5), and produced adult chimeric mice. Based on the colony shape characteristic of mouse iPS cells and GFP-positive results and results positive for other non-differentiation markers, it was concluded that by introducing the above-described expression vector into MEF cells, nuclear reprogramming was completely advanced to produce an iPS cell, and the iPS cell proliferated and formed the visible colony. Hence, these results showed that an iPS cell could be prepared without using a retrovirus or a lentivirus. PCR analysis detected the integration of the above-described expression vector into the host genome (iPS-432A-1 in FIG. 6).
  • Example 2
  • To avoid the integration of pCX-2A-mOKS and pCX-c-Myc into the host genome, the transfection protocol was modified.
  • On days 1, 3, 5, and 7 after the start of the experiment, pCX-2A-mOKS and pCX-c-Myc were transfected together (Exp440 in FIG. 3). As a result, many GFP-positive colonies were obtained, and cells morphologically indistinguishable from ES cells were produced (440A-3 in FIG. 4). The cells obtained expressed the ES cell markers at the same level as with ES cells (iPS-440A in FIG. 5). To examine for the integration of the plasmid DNA into the genome, 16 sets of PCR primers capable of amplifying each portion of the plasmid were designed (FIGS. 2, 13 and 14). In 9 of the 11 GFP-positive clones obtained by the modified protocol, no amplification of an exogenous DNA was observed (FIG. 6). Furthermore, in Southern blot analysis, no integration of an exogenous gene was detected in these clones (FIG. 11). Although the possible presence of a small plasmid fragment cannot be ruled out definitely, the above results showed that these iPS cells did not have the pCX-2A-mOKS and pCX-c-Myc plasmids integrated into the host genome.
  • To rule out the possibility that the iPS cells without integration are derived from possibly contaminating Nanog-GFP ES cells, SSLP analysis was performed. In Exp440 in FIG. 3, MEF cells from five fetuses were used. In the SSLP analysis, these five fetuses were distinguishable, and the derivations of the iPS cells without integration were identified (FIG. 12). This analysis also showed that the iPS cells without integration differed from the ES cells derived from the 12954 strain (FIG. 12).
  • Example 3
  • To confirm the pluripotency of iPS cells without integration, iPS cells obtained as described in Example 2 were subcutaneously transplanted to nude mice. All clones tested (440A-3, -4, -8 and -10) produced tumors, which included a broad range of cell types, including cells derived from all the three germ layers (FIG. 7). Furthermore, iPS cells without integration were injected into ICR mouse blastocysts. Judging from the coat colors, adult chimeras were obtained from all clones injected (440A-3, -4, -6, -8, -9 and -10) (FIG. 8). In these chimeric mice, PCR analysis did not detect the integration of any of the transgenes (FIG. 9). The PCR analysis detected both the Nanog and Fbx15 reporters in the chimeras (FIG. 9). Combined with the fact that iPS cells without integration emerged from the double reporter mice, and that the inventor's laboratory does not keep double reporter ES cells, these results showed that the chimeras were derived from iPS cells without integration, rather than from contaminating ES cells. Hence, these results confirmed that the iPS cells without integration possessed pluripotency.
  • Long-term examination of 71 chimeric mice obtained and offspring thereof showed that in the chimeric mice derived from an iPS cell prepared by introducing 4 genes (Oct3/4, Klf4, Sox2, c-Myc) using a retrovirus, and offspring thereof, compared with normal mice, the mortality rate began to rise earlier, whereas the chimeric mice derived from an iPS cell without integration of the 4 genes and offspring thereof exhibited a survival curve similar to that of normal mice.
  • When chimeric mice obtained and wild mice were mated, F1 mice were obtained; therefore, it was confirmed that iPS cells without integration contributed to the germline (germline-transmission).
  • Example 4
  • Human dental pulp stem cells (clone name; DP31, PCT/JP2008/068320, J. Dent. Res., 87(7):676-681 (2008)) were used as an experimental system. The DP31 was allowed to express the mouse ecotropic virus receptor Slc7a1 gene using a lentivirus as described in Cell, 131, 861-872 (2007). These cells were cultured using the MSCGM bullet kit (Lonza).
  • The plasmids used for reprogramming were prepared from pCX-EGFP (supplied by Dr. Masaru Okabe at Osaka University, FEBS Letters, 407, 313-319, 1997) in the same manner as Example 1. Specifically, the pCX-EGFP was treated with EcoRI, and a construct with the coding regions of SOX2 and KLF4 ligated via the 2A sequence of foot-and-mouth disease virus therein was inserted in place of EGFP, whereby the plasmid pCX-hSK was prepared. Likewise, a plasmid with c-Myc, Lin28, and Nanog ligated via the 2A sequence (pCX-hMLN) therein, a plasmid with the OCT3/4 coding region inserted therein (pCX-hOCT3/4), and a plasmid with the SV40 Large T antigen inserted therein (pCX-SV40LT) were prepared.
  • The DP31 cultured in a 100 mm dish was washed with PBS, 0.25% Trypsin/1 mM EDTA (Invitrogen) was added, and the reaction was carried out at 37° C. for about 5 minutes. After cells rose, MSCGM was added, the cells were suspended, and 6×105 cells were recovered in a 15 mL tube. The cells were centrifuged at 800 rpm for 5 minutes; after the supernatant was removed, and the expression plasmids were introduced using the Human Dermal Fibroblast Nucleofector Kit (Amaxa). The amounts of plasmids used were 0.5 μg for pCX-hOCT3/4, 1.0 μg for pCX-hSK, 1.5 μg for pCX-hMLN, and 0.5 μg for pCX-SV40LT. After the treatment, the cells were sown to a 6-well plate. After being cultured with MSCGM for 10 days, the cells were again washed with PBS, 0.25% Trypsin/1 mM EDTA (Invitrogen) was added, and the reaction was carried out at 37° C. for about 5 minutes. After cells rose, MSCGM was added, the cells were suspended, and 1×105 cells were sown onto a 100 mm dish with feeder cells sown thereto previously. The feeder cells used were SNL cells that had been treated with mitomycin C to terminate their cell division. Thereafter, until a colony began to be observed, the medium was replaced with a fresh supply every two days. The medium used was prepared by mixing equal volumes of a primate ES cell culture medium (ReproCELL) supplemented with MSCGM and bFGF (4 ng/mL), respectively. Colonization began around day 19, confirming the establishment of human iPS cell (FIG. 15).
  • Next, fetal human HDF (Cell applications, INC) was transfected with the same seven kinds of genes as described above. After the transfection, the cells were cultured using a primate ES cell culture medium (ReproCELL) supplemented with 4 ng/ml recombinant human bFGF (WAKO). MSTO cells served as feeder cells. Photographs of cells on day 31 after transfection (5 clones: 203A-1 to 203A-5, of which 203A-4 was picked up as a negative control) are shown in FIG. 16, and photographs of cells in the 2nd subculture are shown in FIG. 17. The 203A-1 to 203A-3 and 203A-5 clones exhibited a typical ES cell-like morphology, confirming the establishment of human iPS cells.
  • These cells were subjected to genomic-PCR analysis, and examined for the integration of the transgenes into the genome. The results are shown in FIG. 18. In all clones, the integration of Oct3/4 (pCX-hOCT3/4) and c-Myc (pCX-hMLN) was detected. The integration of Klf4 (pCX-hSK) was detected in the clones other than 203A-4. The integration of SV40LT (pCX-SV40LT) was not detected in any of the clones.
  • Example 5
  • Dental pulp stem cells DP31, used in Example 4, were transfected with six kinds of genes, excluding the SV40 Large T antigen (pCX-hSK, pCX-hMLN, pCX-hOCT3/4), in the same manner as Example 4. Photographs of cells on day 35 after the transfection (5 clones: 217A-1 to -4 and -6) are shown in FIG. 19. Photographs of cells in the 2nd subculture are shown in FIG. 20. All clones exhibited a typical ES cell-like morphology, confirming the establishment of human iPS cells.
  • These human iPS cell clones established (217A-1 to 217A-4, 217A-6) were subjected to genomic-PCR analysis. The results are shown in FIG. 21. In all these clones, the integration of the transgenes was demonstrated.
  • Example 6
  • An HDF cell line derived from a 6-year-old Japanese female (HDF-120; JCRB) was allowed to express the Slc7a1 gene. The resulting cells (HDF-120-Slc) were transfected with the aforementioned six kinds of genes and an shRNA against p53 (shRNA2: SEQ ID NO:62) (vectors introduced: pCX-hOCT3/4, pCX-hSK, pCX-hMLN-shp53).
  • Each of pCX-hOCT3/4 (0.5 μg), pCX-hSK (1.0 μg), and pCX-hMLN-shp53 (1.5 μg) was electrically introduced into 6.0×105 cells of HDF-120-Slc using Microporator (100 μL tip, 1600 V, 10 ms, 3 times). Ten days later, each vector was once again electrically introduced under the same conditions, and the cells were sown onto MSTO (100 mm dish). These cells were cultured using DMEM/10% FCS until day 10, thereafter using a primate ES cell culture medium (ReproCELL) supplemented with 4 ng/ml recombinant human bFGF (WAKO). Photographs of cells on day 35 after the first electroporation are shown in FIG. 22. Photographs of cells after passage culture are shown in FIG. 23. A typical ES cell-like morphology was exhibited, confirming the establishment of human iPS cells. Genomic-PCR analysis demonstrated the integration of the transgenes (lane 279A-2 in FIG. 24).
  • Example 7
  • Expression vectors separately incorporating the four kinds of genes Oct3/4, Klf4, Sox2 and c-Myc (pCX-Oct4, pCX-Sox2, pCX-Klf4, pCX-c-Myc) were introduced into MEF cells derived from a Nanog reporter mouse (Okita et al. Nature, Vol. 448, pp. 313-317, 2007) per the protocol in Example 2.
  • First, the Nanog reporter MEF cells were sown onto a gelatin-coated 6-well plate (1.3×105 cells/well), and transfected with each of pCX-Oct4 (0.37 μg), pCX-Sox2 (0.36 μg), pCX-Klf4 (0.39 μg), and pCX-c-Myc (0.38 μg) using FuGene6 on days 1, 3, 5, and 7. On day 9, 1×106 cells (1.0) or 0.2×106 cells (0.2) were sown onto MSTO-PH or gelatin (100-mm dish), and colonies were selected on day 25. Photographs of cells after the selection are shown in FIG. 25. A colony shape characteristic of mouse iPS cells and GFP-positive results were obtained, confirming the establishment of mouse iPS cells. The mouse iPS cell clones established (497A-1 to A-5) were subjected to genomic-PCR analysis. The results are shown in FIG. 26. Both 497A-2 and 497A-5 were shown to be iPS cells without integration of any of the exogenous genes.
  • INDUSTRIAL APPLICABILITY
  • According to the method of the present invention, it is possible to prepare a highly safe induced pluripotent stem cell from, for example, a patient's somatic cell. The cells obtained by differentiating the induced pluripotent stem cell (e.g., myocardial cells, insulin-producing cells, nerve cells and the like) can be safely used for stem cell transplantation therapy for a broad range of diseases, including heart failure, insulin-dependent diabetes, Parkinson's disease and spinal injury.
  • While the present invention has been described with emphasis on preferred embodiments, it is obvious to those skilled in the art that the preferred embodiments can be modified. The present invention intends that the present invention can be embodied by methods other than those described in detail in the present specification. Accordingly, the present invention encompasses all modifications encompassed in the gist and scope of the appended “Claims”.
  • The contents disclosed in any publication cited here, including patents and patent applications, are hereby incorporated in their entireties by reference, to the extent that they have been disclosed herein.
  • This application is based on U.S. provisional patent application Nos. 61/071,508, 61/136,246, 61/136,615 and 61/193,363, the contents of which are hereby incorporated by reference.

Claims (5)

What is claimed is:
1. A method of producing human induced pluripotent stem (iPS) cells population and having cells where the vector is not integrated into the genome, the method comprising the steps of
a) obtaining a starting population of human somatic cells;
b) obtaining one or more iPS reprogramming vectors comprising a DNA sequence that allows the autonomous replication of the expression vector outside a chromosome in a somatic cell and one or more expression cassettes encoding iPS reprogramming factors;
c) introducing the iPS reprogramming vector(s) into the human somatic cells;
d) culturing the human somatic cells that comprise the iPS reprogramming vector(s) to effect expression of said reprogramming factors such that traits consistent with embryonic stem cells arise in at least a portion of cells in the cultured cells; and
e) further culturing cells having the embryonic stem cell traits for a sufficient number of generations in the absence of positive selection for the presence of the iPS reprogramming vector(s) to provide human iPS cells that are essentially free of iPS reprogramming vector genetic elements.
2. The method of claim 1, wherein cells of the starting cell population are fibroblasts, keratinocytes, hematopoietic cells, mesenchymal cells, liver cells, stomach cells, or β cells.
3. The method of claim 1, wherein the iPS reprogramming factors comprise Sox-2 and Oct-4.
4. The method of claim 3, wherein the reprogramming factors further comprise Nanog or Lin28.
5. The method of claim 1, further comprising a step of differentiating the human iPS cells provided in step e).
US14/502,374 2008-05-02 2014-09-30 Method of nuclear reprogramming Abandoned US20150072417A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/502,374 US20150072417A1 (en) 2008-05-02 2014-09-30 Method of nuclear reprogramming
US16/702,391 US20200172875A1 (en) 2008-05-02 2019-12-03 Method of nuclear reprogramming
US18/146,644 US20230282445A1 (en) 2008-05-02 2022-12-27 Method of nuclear reprogramming

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US7150808P 2008-05-02 2008-05-02
US13624608P 2008-08-21 2008-08-21
US13661508P 2008-09-19 2008-09-19
US19336308P 2008-11-21 2008-11-21
PCT/JP2009/058873 WO2009133971A1 (en) 2008-05-02 2009-05-01 Method of nuclear reprogramming
US73311810A 2010-05-03 2010-05-03
US13/572,593 US9499797B2 (en) 2008-05-02 2012-08-10 Method of making induced pluripotent stem cells
US14/502,374 US20150072417A1 (en) 2008-05-02 2014-09-30 Method of nuclear reprogramming

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/572,593 Continuation US9499797B2 (en) 2008-05-02 2012-08-10 Method of making induced pluripotent stem cells

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/702,391 Continuation US20200172875A1 (en) 2008-05-02 2019-12-03 Method of nuclear reprogramming

Publications (1)

Publication Number Publication Date
US20150072417A1 true US20150072417A1 (en) 2015-03-12

Family

ID=41255176

Family Applications (5)

Application Number Title Priority Date Filing Date
US12/733,118 Abandoned US20100279404A1 (en) 2008-05-02 2009-05-01 Method of nuclear reprogramming
US13/572,593 Active US9499797B2 (en) 2008-05-02 2012-08-10 Method of making induced pluripotent stem cells
US14/502,374 Abandoned US20150072417A1 (en) 2008-05-02 2014-09-30 Method of nuclear reprogramming
US16/702,391 Abandoned US20200172875A1 (en) 2008-05-02 2019-12-03 Method of nuclear reprogramming
US18/146,644 Pending US20230282445A1 (en) 2008-05-02 2022-12-27 Method of nuclear reprogramming

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US12/733,118 Abandoned US20100279404A1 (en) 2008-05-02 2009-05-01 Method of nuclear reprogramming
US13/572,593 Active US9499797B2 (en) 2008-05-02 2012-08-10 Method of making induced pluripotent stem cells

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/702,391 Abandoned US20200172875A1 (en) 2008-05-02 2019-12-03 Method of nuclear reprogramming
US18/146,644 Pending US20230282445A1 (en) 2008-05-02 2022-12-27 Method of nuclear reprogramming

Country Status (9)

Country Link
US (5) US20100279404A1 (en)
EP (1) EP2268809B1 (en)
JP (1) JP5346925B2 (en)
KR (1) KR101661940B1 (en)
CN (1) CN101855350B (en)
CA (1) CA2695522C (en)
ES (1) ES2722198T3 (en)
SG (1) SG10201400329YA (en)
WO (1) WO2009133971A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9499797B2 (en) 2008-05-02 2016-11-22 Kyoto University Method of making induced pluripotent stem cells
US9714433B2 (en) 2007-06-15 2017-07-25 Kyoto University Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7682828B2 (en) 2003-11-26 2010-03-23 Whitehead Institute For Biomedical Research Methods for reprogramming somatic cells
EP2302034B1 (en) * 2005-08-03 2020-10-14 Astellas Institute for Regenerative Medicine Improved methods of reprogramming animal somatic cells
EP4223769A3 (en) 2005-12-13 2023-11-01 Kyoto University Nuclear reprogramming factor
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
US8440461B2 (en) 2007-03-23 2013-05-14 Wisconsin Alumni Research Foundation Reprogramming somatic cells using retroviral vectors comprising Oct-4 and Sox2 genes
WO2008124133A1 (en) 2007-04-07 2008-10-16 Whitehead Institute For Biomedical Research Reprogramming of somatic cells
US9213999B2 (en) 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
AU2009256202B2 (en) 2008-06-04 2014-07-03 FUJIFILM Cellular Dynamics, Inc. Methods for the production of IPS cells using non-viral approach
WO2009152529A2 (en) * 2008-06-13 2009-12-17 Whitehead Institute For Biomedical Research Nine Cambridge Center Programming and reprogramming of cells
AU2015202237B2 (en) * 2008-06-13 2017-09-28 Whitehead Institute For Biomedical Research Programming and reprogramming of cells
CN102159710B (en) * 2008-07-16 2015-09-02 生物载体株式会社 Use the manufacture of karyomit(e) circles virus vector through the method for initialized cell
JP5562231B2 (en) * 2008-07-30 2014-07-30 国立大学法人京都大学 Efficient method for establishing induced pluripotent stem cells
CN107988261A (en) 2008-08-12 2018-05-04 细胞动力国际有限公司 The method for producing IPS cells
WO2010030003A1 (en) * 2008-09-12 2010-03-18 協和発酵キリン株式会社 System for regulating expression of multiple genes for use in reprogramming of cell
WO2010042490A1 (en) * 2008-10-06 2010-04-15 Boston Medical Center Corporation A single lentiviral vector system for induced pluripotent (ips) stem cells derivation
EP3450545B1 (en) * 2008-10-24 2023-08-23 Wisconsin Alumni Research Foundation Pluripotent stem cells obtained by non-viral reprogramming
WO2010105311A1 (en) * 2009-03-20 2010-09-23 Angioblast Systems, Inc. Production of reprogrammed pluripotent cells
EP3150701B1 (en) 2009-06-05 2018-10-03 FUJIFILM Cellular Dynamics, Inc. Reprogramming t cells and hematopoietic cells
WO2011111614A1 (en) * 2010-03-10 2011-09-15 Kyoto University Method of selecting induced pluripotent stem cell
EP3936608A1 (en) * 2010-03-31 2022-01-12 The Scripps Research Institute Reprogramming cells
WO2011130624A2 (en) 2010-04-16 2011-10-20 Immune Disease Institute, Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof
US8048675B1 (en) 2010-05-12 2011-11-01 Ipierian, Inc. Integration-free human induced pluripotent stem cells from blood
EP2612911B1 (en) * 2010-08-30 2018-01-17 ID Pharma Co., Ltd. Composition for inducing pluripotent stem cell, and use thereof
US20130295064A1 (en) * 2010-10-14 2013-11-07 University Of Central Florida Research Foundation, Inc. Cardiac induced pluripotent stem cells and methods of use in repair and regeneration of myocardium
US9506039B2 (en) 2010-12-03 2016-11-29 Kyoto University Efficient method for establishing induced pluripotent stem cells
WO2012098260A1 (en) 2011-01-21 2012-07-26 Axiogenesis Ag A non-viral system for the generation of induced pluripotent stem (ips) cells
US9228204B2 (en) 2011-02-14 2016-01-05 University Of Utah Research Foundation Constructs for making induced pluripotent stem cells
EP2707479B1 (en) 2011-05-13 2018-01-10 The United States of America, as represented by The Secretary, Department of Health and Human Services Use of zscan4 and zscan4-dependent genes for direct reprogramming of somatic cells
WO2013010045A1 (en) 2011-07-12 2013-01-17 Biotime Inc. Novel methods and formulations for orthopedic cell therapy
WO2014186766A1 (en) * 2013-05-17 2014-11-20 The Broad Institute, Inc. Reprogrammed cells and methods of production and use thereof
WO2015006725A2 (en) * 2013-07-12 2015-01-15 Cedars-Sinai Medical Center Generation of induced pluripotent stem cells from normal human mammary epithelial cells
US9890360B2 (en) 2013-08-28 2018-02-13 Gifu University Method for producing induced pluripotent stem cells
WO2015066488A2 (en) 2013-11-01 2015-05-07 New England Biolabs, Inc. Method for producing induced pluripotent stem cells
EP3359169A4 (en) 2015-10-05 2019-03-13 Orig3N, Inc. Diagnosis and treatment of parkinson's disease based on identification and amelioration of liver dysfunction
RU2741786C2 (en) 2016-05-25 2021-01-28 Энсэрм (Энститю Насьональ Де Ла Санте Э Де Ла Решерш Медикаль) Methods and compositions for treating tumors
US10221395B2 (en) 2016-06-16 2019-03-05 Cedars-Sinai Medical Center Efficient method for reprogramming blood to induced pluripotent stem cells
US11572545B2 (en) 2016-06-16 2023-02-07 Cedars-Sinai Medical Center Efficient method for reprogramming blood to induced pluripotent stem cells
WO2019002399A1 (en) * 2017-06-28 2019-01-03 Sci-Group As Freezing of biological material
JP7408036B2 (en) 2017-11-24 2024-01-05 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Methods and compositions for treating cancer
EP3833383A1 (en) 2018-08-06 2021-06-16 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treating cancers
WO2022207889A1 (en) 2021-04-01 2022-10-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Liver organoid manufacturing methods, liver organoids obtained with the same, and uses thereof
CN113462638B (en) * 2021-06-30 2022-10-25 呈诺再生医学科技(珠海横琴新区)有限公司 Efficient genetic-modification-free iPSC induction and industrialization monoclonal picking platform and application
WO2024020587A2 (en) 2022-07-22 2024-01-25 Tome Biosciences, Inc. Pleiopluripotent stem cell programmable gene insertion

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007006966A2 (en) * 2005-07-13 2007-01-18 Airbus France Device for assisting a vertical guidance approach for aircraft
US8058065B2 (en) * 2005-12-13 2011-11-15 Kyoto University Oct3/4, Klf4, c-Myc and Sox2 produce induced pluripotent stem cells
US8129187B2 (en) * 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US8298825B1 (en) * 2008-08-25 2012-10-30 The General Hospital Corporation TGF-beta receptor inhibitors to enhance direct reprogramming

Family Cites Families (156)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4650764A (en) * 1983-04-12 1987-03-17 Wisconsin Alumni Research Foundation Helper cell
US4861719A (en) * 1986-04-25 1989-08-29 Fred Hutchinson Cancer Research Center DNA constructs for retrovirus packaging cell lines
US4937190A (en) * 1987-10-15 1990-06-26 Wisconsin Alumni Research Foundation Translation enhancer
US5192553A (en) * 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US6140111A (en) * 1987-12-11 2000-10-31 Whitehead Institute For Biomedical Research Retroviral gene therapy vectors and therapeutic methods based thereon
US7070994B2 (en) * 1988-03-21 2006-07-04 Oxford Biomedica (Uk) Ltd. Packaging cells
US5591624A (en) * 1988-03-21 1997-01-07 Chiron Viagene, Inc. Retroviral packaging cell lines
JP2886547B2 (en) * 1988-07-26 1999-04-26 協和醗酵工業株式会社 Method for producing neuraminidase
CA1339354C (en) * 1988-09-01 1997-08-26 The Whitehead Institute For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
JP2928287B2 (en) 1988-09-29 1999-08-03 協和醗酵工業株式会社 Novel polypeptide
JP3051411B2 (en) * 1989-03-14 2000-06-12 持田製薬株式会社 Novel DNA and expression plasmid containing it
US5266491A (en) * 1989-03-14 1993-11-30 Mochida Pharmaceutical Co., Ltd. DNA fragment and expression plasmid containing the DNA fragment
JP2897295B2 (en) * 1989-12-14 1999-05-31 味の素株式会社 DNA construct for high production of retrovirus and cell line for high production of retrovirus
US5652122A (en) * 1989-12-21 1997-07-29 Frankel; Alan Nucleic acids encoding and methods of making tat-derived transport polypeptides
US5817491A (en) * 1990-09-21 1998-10-06 The Regents Of The University Of California VSV G pseusdotyped retroviral vectors
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5834256A (en) * 1993-06-11 1998-11-10 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
FR2707091B1 (en) * 1993-06-30 1997-04-04 Cohen Haguenauer Odile Retroviral vector for gene transfer and expression in eukaryotic cells.
US5534423A (en) 1993-10-08 1996-07-09 Regents Of The University Of Michigan Methods of increasing rates of infection by directing motion of vectors
US5519134A (en) * 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US6013517A (en) * 1994-05-09 2000-01-11 Chiron Corporation Crossless retroviral vectors
US5525735A (en) * 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) * 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
JP4216350B2 (en) * 1994-09-19 2009-01-28 大日本住友製薬株式会社 Recombinant DNA viral vector for animal cell infection
DK0787200T3 (en) * 1994-10-28 2005-08-15 Univ Pennsylvania Improved adenovirus and methods for its use
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5637456A (en) * 1995-02-17 1997-06-10 The University Of Texas, Board Of Regents Rapid test for determining the amount of functionally inactive gene in a gene therapy vector preparation
US5707618A (en) * 1995-03-24 1998-01-13 Genzyme Corporation Adenovirus vectors for gene therapy
US5830725A (en) * 1995-04-28 1998-11-03 The Board Of Trustees For The Leland Stanford Junior University Rapid, stable high-titre production of recombing retrovirus
US5744320A (en) * 1995-06-07 1998-04-28 Promega Corporation Quenching reagents and assays for enzyme-mediated luminescence
BR9610058A (en) * 1995-07-28 1999-07-27 Marie Curie Cancer Care Use of vp22 or an active fragment or homologous portion of the same nucleic acid transport protein mammalian host cell or microbial expression vector and processes to transport a desired protein or peptide to a cell labeling population and to transport a desired molecule in a cell population
JP4053595B2 (en) * 1995-09-22 2008-02-27 メディカル・リサーチ・カウンシル Improvements in or related to mutagenesis of nucleic acids
US5910434A (en) * 1995-12-15 1999-06-08 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
FR2751345B1 (en) 1996-07-16 1998-09-18 Univ Paris Curie HIGHLY PRODUCING PACKAGING LINES
US6255071B1 (en) * 1996-09-20 2001-07-03 Cold Spring Harbor Laboratory Mammalian viral vectors and their uses
US6025192A (en) * 1996-09-20 2000-02-15 Cold Spring Harbor Laboratory Modified retroviral vectors
US6017735A (en) * 1997-01-23 2000-01-25 Marie Curie Cancer Care Materials and methods for intracellular transport and their uses
US6416959B1 (en) * 1997-02-27 2002-07-09 Kenneth Giuliano System for cell-based screening
TR199902512T2 (en) * 1997-04-16 2000-06-21 Amgen Inc. Osteoprotegerin binding proteins and receptors.
AU9200398A (en) * 1997-08-22 1999-03-16 Yale University A process to study changes in gene expression in granulocytic cells
WO2000018885A1 (en) 1998-09-29 2000-04-06 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
US6835567B1 (en) 1998-04-14 2004-12-28 Signal Pharmaceuticals, Inc. PNS cell lines and methods of use therefor
US20020174013A1 (en) * 1998-04-17 2002-11-21 Viztec Inc., A Florida Corporation Chip card advertising method and system
GB9809178D0 (en) 1998-04-29 1998-07-01 Univ Edinburgh Nuclear reprogramming of somatic cells
EP1080218A1 (en) 1998-05-27 2001-03-07 University of Florida Method of preparing recombinant adeno-associated virus compositions by using an iodixanol gradient
FR2779445B1 (en) 1998-06-08 2000-08-11 Univ Nantes ENCAPSIDATION KIT
KR20000006334A (en) * 1998-06-26 2000-01-25 이선경 High efficiency retroviral vectors that contain none of viral coding sequences
US6485959B1 (en) * 1998-10-07 2002-11-26 Cedars Sinai Medical Center Cell preconditioning and cryopresevation medium
AU6339399A (en) 1998-10-16 2000-05-08 Novartis Ag Promotion of self-renewal and improved gene transduction of hematopoietic stem cells by histone deacetylase inhibitors
US6667176B1 (en) 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
IL142748A0 (en) * 1998-11-09 2002-03-10 Univ Monash Embryonic stem cells
US6376246B1 (en) * 1999-02-05 2002-04-23 Maxygen, Inc. Oligonucleotide mediated nucleic acid recombination
US6153432A (en) * 1999-01-29 2000-11-28 Zen-Bio, Inc Methods for the differentiation of human preadipocytes into adipocytes
US6312949B1 (en) * 1999-03-26 2001-11-06 The Salk Institute For Biological Studies Regulation of tyrosine hydroxylase expression
US6773920B1 (en) * 1999-03-31 2004-08-10 Invitrogen Corporation Delivery of functional protein sequences by translocating polypeptides
US6995009B1 (en) * 1999-06-01 2006-02-07 Chugai Seiyaku Kabushiki Kaisha Packaging cell
US7015037B1 (en) * 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
AU7473500A (en) * 1999-09-01 2001-03-26 University Of Pittsburgh Identification of peptides that facilitate uptake and cytoplasmic and/or nucleartransport of proteins, dna and viruses
EP1218489B1 (en) 1999-09-24 2009-03-18 Cybios LLC Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US20030161817A1 (en) * 2001-03-28 2003-08-28 Young Henry E. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US6280718B1 (en) 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
US7544509B2 (en) * 2000-01-24 2009-06-09 Mcgill University Method for preparing stem cell preparations
US6395546B1 (en) * 2000-02-01 2002-05-28 Neurogeneration, Inc. Generation of dopaminergic neurons from human nervous system stem cells
US7439064B2 (en) * 2000-03-09 2008-10-21 Wicell Research Institute, Inc. Cultivation of human embryonic stem cells in the absence of feeder cells or without conditioned medium
US6458589B1 (en) 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
AU6319901A (en) 2000-05-17 2001-11-26 Geron Corp Neural progenitor cell populations
WO2001096532A2 (en) * 2000-06-15 2001-12-20 Tanja Dominko Method of generating pluripotent mammalian cells by fusion of a cytoplast fragment with a karyoplast
DE10031179A1 (en) 2000-06-27 2002-01-31 Amaxa Gmbh Introducing active molecules into the nucleus of eukaryotic cells, useful for transfection and gene therapy, by applying controlled electrical pulse
AU2001280767A1 (en) * 2000-07-31 2002-02-13 Active Motif Peptide-mediated delivery of molecules into cells
JP2002065261A (en) 2000-08-30 2002-03-05 Mitsubishi Kasei Institute Of Life Sciences Method for obtaining reproductive cell
US6910434B2 (en) * 2000-08-31 2005-06-28 Edwin Lundgren Control device for steering kite on a boat
US20020127715A1 (en) 2000-11-27 2002-09-12 Nissim Benvenisty Transfection of human embryonic stem cells
US20080268054A1 (en) 2000-12-04 2008-10-30 Eugene Bell Dermal derived human stem cells and compositions and methods thereof
CA2433419A1 (en) * 2001-01-02 2002-07-25 Stemron, Inc. A method for producing a population of homozygous stem cells having a pre-selected immunotype and/or genotype, cells suitable for transplant derived therefrom, and materials and methods using same
JP2003009854A (en) 2001-04-09 2003-01-14 Kyowa Hakko Kogyo Co Ltd Method for embryoid body formation and use thereof
EP1390518B1 (en) 2001-04-23 2004-10-06 Amaxa GmbH Buffer solution for electroporation and a method comprising the use of the same
DE10119901A1 (en) 2001-04-23 2002-10-24 Amaxa Gmbh Apparatus for electrophoretic transfer of biologically active molecules into cells comprises capacitors connected to high voltage sources, which discharge via power transistor into cuvette holding sample
US7250255B2 (en) * 2001-05-31 2007-07-31 Shinya Yamanaka Genes with ES cell-specific expression
WO2003018780A1 (en) 2001-08-27 2003-03-06 Advanced Cell Technology, Inc. De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
JPWO2003027281A1 (en) * 2001-09-20 2005-01-06 協和醗酵工業株式会社 Skeletal muscle stroma-derived multipotent stem cells
AU2002330465B2 (en) 2001-09-21 2006-07-27 Kyoto University Screening method for a reprogramming agent, reprogramming agent screened by the method, method for using the reprogramming agent, method for differentiating an undifferentiated fusion cell, and production method of cells, tissues and organs.
JP2004248505A (en) 2001-09-21 2004-09-09 Norio Nakatsuji Undifferentiated fusion cell of somatic cell derived from es cell deficient in part or all of transplantation antigen and method for producing the same
US7588937B2 (en) * 2001-10-03 2009-09-15 Wisconsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
DE10162080A1 (en) * 2001-12-10 2003-06-26 Albrecht Mueller Process for the production of stem cells with increased development potential
AU2003238620B2 (en) * 2002-01-31 2008-11-06 Asahi Techno Glass Corporation Liquid for frozen storage of primate embryo stem cells and frozen storage method
WO2003081536A1 (en) 2002-03-26 2003-10-02 So-Woon Kim System and method for 3-dimension simulation of glasses
US7422736B2 (en) 2002-07-26 2008-09-09 Food Industry Research And Development Institute Somatic pluripotent cells
US20040048297A1 (en) * 2002-07-30 2004-03-11 Gene Logic, Inc. Nucleic acid detection assay control genes
JP3736517B2 (en) 2002-11-13 2006-01-18 学校法人近畿大学 Somatic cell nuclear reprogramming factor
AU2003901099A0 (en) 2003-03-11 2003-03-27 Es Cell International Pte Ltd. Methods of inducing differentiation of stem cells
WO2004099372A2 (en) * 2003-05-01 2004-11-18 University Of Florida Anti-scarring ribozymes and methods
US9567591B2 (en) 2003-05-15 2017-02-14 Mello Biotechnology, Inc. Generation of human embryonic stem-like cells using intronic RNA
JPWO2004101775A1 (en) * 2003-05-16 2006-07-13 協和醗酵工業株式会社 Novel adult tissue-derived stem cells and uses thereof
CA2526120A1 (en) 2003-06-03 2005-02-24 Cell Genesys, Inc. Compositions and methods for enhanced expression of recombinant polypeptides from a single vector using a peptide cleavage site
US20050019801A1 (en) * 2003-06-04 2005-01-27 Curis, Inc. Stem cell-based methods for identifying and characterizing agents
WO2005033297A1 (en) 2003-09-19 2005-04-14 The Rockefeller University Compositions, methods and kits relating to reprogramming adult differentiated cells and production of embryonic stem cell-like cells
JP2005095027A (en) 2003-09-22 2005-04-14 Reprocell Inc Undifferentiated state marker promoter of cell and its utilization
CA2542130A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Cell genomically modified to produce polypeptides with an altered glycosylation pattern
EP1682150B1 (en) * 2003-11-10 2012-12-26 The Scripps Research Institute Compositions and methods for inducing cell dedifferentiation
US7682828B2 (en) 2003-11-26 2010-03-23 Whitehead Institute For Biomedical Research Methods for reprogramming somatic cells
CA2549158A1 (en) * 2003-12-01 2005-06-16 Technion Research And Development Foundation Ltd. Methods of generating stem cells and embryonic bodies carrying disease-causing mutations and methods of using same for studying genetic disorders
JP4340736B2 (en) * 2004-01-15 2009-10-07 国立大学法人福井大学 Vector incorporating a reporter gene
WO2005080598A1 (en) * 2004-02-19 2005-09-01 Dainippon Sumitomo Pharma Co., Ltd. Method of screening somatic cell nucleus initializer
AU2005224569B2 (en) * 2004-03-23 2011-07-14 Toshihiro Akaike Method of proliferating pluripotent stem cell
CA2568772A1 (en) * 2004-06-01 2005-12-15 San Diego State University Foundation Expression system based on recombinant vesicular stomatitis virus synthesizing t7 rna polymerase
US20070202592A1 (en) * 2004-07-08 2007-08-30 Yasuo Kitagawa Pluripotent Cells Distributed Ubiquitously In Animal Tissue, Which Proliferate Selectively In Lower-Serum Culture
WO2006084229A2 (en) 2004-07-15 2006-08-10 Primegen Biotech, Llc Use of nuclear material to therapeutically reprogram differentiated cells
WO2006017476A2 (en) * 2004-08-02 2006-02-16 The Research Foundation Of State University Of New York Amino functionalized ormosil nanoparticles as delivery vehicles
JPWO2006035741A1 (en) 2004-09-29 2008-05-15 伸弥 山中 ES cell specific expression gene and use thereof
JP5053850B2 (en) * 2004-10-08 2012-10-24 バークシス コーポレーション Use of RNA trans-splicing for antibody gene transfer and antibody polypeptide production
US20060095319A1 (en) * 2004-10-29 2006-05-04 Cardwell Carlzo B Marketing and compensation method
WO2006088867A2 (en) 2005-02-15 2006-08-24 Medistem Laboratories, Incorporated Method for expansion of stem cells
WO2006093172A1 (en) * 2005-02-28 2006-09-08 Foundation For Biomedical Research And Innovation Method for in vitro amplification of adult stem cells
US20070033061A1 (en) * 2005-04-05 2007-02-08 Achaogen, Inc. Business methods for commercializing antimicrobial and cytotoxic compounds
WO2007026255A2 (en) 2005-06-22 2007-03-08 Universitetet I Oslo Dedifferentiated cells and methods of making and using dedifferentiated cells
EP1910550A4 (en) * 2005-07-21 2009-11-04 Abbott Lab Multiple gene expression including sorf constructs and methods with polyproteins, pro-proteins, and proteolysis
EP2302034B1 (en) 2005-08-03 2020-10-14 Astellas Institute for Regenerative Medicine Improved methods of reprogramming animal somatic cells
EP1957643A2 (en) 2005-11-11 2008-08-20 The University Court Of The University of Edinburgh Reprogramming and genetic modification of cells
EP4223769A3 (en) 2005-12-13 2023-11-01 Kyoto University Nuclear reprogramming factor
US20090227032A1 (en) 2005-12-13 2009-09-10 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
WO2007097494A1 (en) 2006-02-27 2007-08-30 Imgen Co., Ltd. De-differentiation of astrocytes into neural stem cell using bmi-1
US20090252711A1 (en) 2006-05-11 2009-10-08 Andrew Craig Boquest Stem Cells And Methods Of Making And Using Stem Cells
WO2008030610A2 (en) 2006-09-08 2008-03-13 Michigan State University Human transcriptome corresponding to human oocytes and use of said genes or the corresponding polypeptides to trans-differentiate somatic cells
JP2008099662A (en) 2006-09-22 2008-05-01 Institute Of Physical & Chemical Research Method for culturing stem cell
US20080132803A1 (en) * 2006-11-30 2008-06-05 Hyman Friedlander Method and system for doing business by mining the placental-chord complex
US7892830B2 (en) * 2007-01-17 2011-02-22 Wisconsin Alumni Research Foundation Clonal culture of human pluripotent stem cells
WO2008105566A1 (en) 2007-02-27 2008-09-04 Korea Stem Cell Bank System for providing stem cell services using internet and method thereof
US8158415B2 (en) 2007-02-27 2012-04-17 Procell Therapeutics Inc. Combined use of cell permeable Nanog and Oct4 for increasing self-renewal and suppressing differentiation of stem cells
US8440461B2 (en) 2007-03-23 2013-05-14 Wisconsin Alumni Research Foundation Reprogramming somatic cells using retroviral vectors comprising Oct-4 and Sox2 genes
WO2008124133A1 (en) 2007-04-07 2008-10-16 Whitehead Institute For Biomedical Research Reprogramming of somatic cells
AU2008260187A1 (en) 2007-05-29 2008-12-11 Christopher B. Reid Methods for production and uses of multipotent cell populations
US20100184051A1 (en) * 2007-05-30 2010-07-22 The General Hospital Corporation Methods of generating pluripotent cells from somatic cells
US9213999B2 (en) 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
JP2008307007A (en) * 2007-06-15 2008-12-25 Bayer Schering Pharma Ag Human pluripotent stem cell induced from human tissue-originated undifferentiated stem cell after birth
US20120282229A1 (en) 2007-08-01 2012-11-08 Christian Kannemeier Non-viral delivery of transcription factors that reprogram human somatic cells into a stem cell-like state
US20090191160A1 (en) 2007-08-10 2009-07-30 University Of Dayton Methods of producing pluripotent stem-like cells
MX2010002242A (en) 2007-08-31 2010-06-01 Whitehead Biomedical Inst Wnt pathway stimulation in reprogramming somatic cells.
WO2009057831A1 (en) 2007-10-31 2009-05-07 Kyoto University Nuclear reprogramming method
WO2009061442A1 (en) 2007-11-06 2009-05-14 Children's Medical Center Corporation Method to produce induced pluripotent stem (ips) cells form non-embryonic human cells
US9005966B2 (en) 2007-11-19 2015-04-14 The Regents Of The University Of California Generation of pluripotent cells from fibroblasts
US9683232B2 (en) 2007-12-10 2017-06-20 Kyoto University Efficient method for nuclear reprogramming
AU2008286249B2 (en) * 2007-12-10 2013-10-10 Kyoto University Efficient method for nuclear reprogramming
US20090191171A1 (en) * 2008-01-18 2009-07-30 Yupo Ma Reprogramming of Differentiated Progenitor or Somatic Cells Using Homologous Recombination
KR101481164B1 (en) 2008-01-30 2015-01-09 주식회사 미래셀바이오 Method of manufacturing induced pluripotent stem cell originated from somatic cell
US20110014164A1 (en) 2008-02-15 2011-01-20 President And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
JP2011516042A (en) 2008-03-17 2011-05-26 へルムホルツ・ツェントルム・ミュンヘン−ドイチェス・フォルシュングスツェントルム・ヒューア・ゲズントハイト・ウント・ウムヴェルト(ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング) Vector and method for producing vector-free induced pluripotent stem (iPS) cells using site-specific recombination
SG10202103401QA (en) * 2008-03-17 2021-05-28 Scripps Research Inst Combined chemical and genetic approaches for generation of induced pluripotent stem cells
CN101250502A (en) 2008-04-01 2008-08-27 中国科学院上海生命科学研究院 Method for preparing evoked pluripotent stem cell
CN101550406B (en) * 2008-04-03 2016-02-10 北京大学 Prepare the method for pluripotent stem cell, test kit and purposes
US20100021437A1 (en) * 2008-04-07 2010-01-28 The McLean Hospital Corporation Whitehead Institute for Biomedical Research Neural stem cells derived from induced pluripotent stem cells
WO2009133971A1 (en) 2008-05-02 2009-11-05 Kyoto University Method of nuclear reprogramming
EP2128245A1 (en) 2008-05-27 2009-12-02 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Generation of induced pluripotent stem (iPS) cells
AU2009256202B2 (en) 2008-06-04 2014-07-03 FUJIFILM Cellular Dynamics, Inc. Methods for the production of IPS cells using non-viral approach
US20110201110A1 (en) 2008-07-31 2011-08-18 Gifu University Efficient method for establishing induced pluripotent stem cells
US20100062534A1 (en) * 2008-09-09 2010-03-11 The General Hospital Corporation Inducible lentiviral vectors for reprogramming somatic cells
EP3450545B1 (en) 2008-10-24 2023-08-23 Wisconsin Alumni Research Foundation Pluripotent stem cells obtained by non-viral reprogramming

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007006966A2 (en) * 2005-07-13 2007-01-18 Airbus France Device for assisting a vertical guidance approach for aircraft
US8058065B2 (en) * 2005-12-13 2011-11-15 Kyoto University Oct3/4, Klf4, c-Myc and Sox2 produce induced pluripotent stem cells
US8129187B2 (en) * 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US8278104B2 (en) * 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
US8298825B1 (en) * 2008-08-25 2012-10-30 The General Hospital Corporation TGF-beta receptor inhibitors to enhance direct reprogramming

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
Blelloch (Cell Stem Cell, Sept. 2007, Vol. 1, pg 245-247) *
Duinsbergen (Experimental Cell Res. July 9, 2008, Vol. 314, pg 3255-3263) *
Eminli (Stem Cells, July 17, 2008, Vol. 26, pg 2467-2474) *
Feng (Cell Stem Cell, April 3, 2009, Vol. 4, pg 301-312) *
Kanai-Azuma (Development, 2002, Vol. 129, pg 2367-2379 *
Maherali (Cell Stem Cell, July 2007, Vol. 1, pg 55-70) *
Nakagawa (Nat Biotechnol, Jan. 2008 (published online Nov. 30, 2007), Vol. 26: 101-106) *
Okabe (FEBS letters, 1997, Vol. 407, pg 313-319) *
Okita (Nature, 2007, Vol. 448, pg 313-317) *
Okita (Science, Nov. 7, 2008, Vol. 322, pg 949-953) *
Takahashi (Cell, Aug. 25, 2006, Vol. 126, pg 663-676) *
Yan (Stem Cells and Development, 2010, Vol. 19, No. 4, pg 469-480) *
Yu (Science, Dec. 2007, Vol. 318, pg 1917-1920) *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9714433B2 (en) 2007-06-15 2017-07-25 Kyoto University Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
US9499797B2 (en) 2008-05-02 2016-11-22 Kyoto University Method of making induced pluripotent stem cells

Also Published As

Publication number Publication date
CA2695522A1 (en) 2009-11-05
SG10201400329YA (en) 2014-05-29
JP5346925B2 (en) 2013-11-20
US20200172875A1 (en) 2020-06-04
US20230282445A1 (en) 2023-09-07
WO2009133971A1 (en) 2009-11-05
CN101855350B (en) 2014-12-31
ES2722198T3 (en) 2019-08-08
CN101855350A (en) 2010-10-06
JP2011519546A (en) 2011-07-14
KR20110015500A (en) 2011-02-16
KR101661940B1 (en) 2016-10-04
US20130065311A1 (en) 2013-03-14
EP2268809A1 (en) 2011-01-05
US20100279404A1 (en) 2010-11-04
US9499797B2 (en) 2016-11-22
CA2695522C (en) 2019-01-15
EP2268809B1 (en) 2019-02-06
EP2268809A4 (en) 2013-02-27

Similar Documents

Publication Publication Date Title
US20230282445A1 (en) Method of nuclear reprogramming
EP2307539B1 (en) Method of efficiently establishing induced pluripotent stem cells
JP5827220B2 (en) Method for improving the efficiency of establishment of induced pluripotent stem cells
JP5098028B2 (en) Nuclear reprogramming factor
EP2536828B1 (en) Method of efficiently establishing induced pluripotent stem cells
CA2874259C (en) Highly efficient method for establishing induced pluripotent stem cell
CN104630136B (en) Composition and its application used in a kind of method and this method preparing inducing pluripotent stem cells
JP5626619B2 (en) Efficient nuclear initialization method
US9637732B2 (en) Method of efficiently establishing induced pluripotent stem cells
JP2013505701A (en) Efficient method for establishing induced pluripotent stem cells

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION