US20210284968A1 - Method for inducing alveolar epithelial progenitor cells - Google Patents

Method for inducing alveolar epithelial progenitor cells Download PDF

Info

Publication number
US20210284968A1
US20210284968A1 US17/194,761 US202117194761A US2021284968A1 US 20210284968 A1 US20210284968 A1 US 20210284968A1 US 202117194761 A US202117194761 A US 202117194761A US 2021284968 A1 US2021284968 A1 US 2021284968A1
Authority
US
United States
Prior art keywords
cells
inhibitor
cpm
cell population
medium
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.)
Pending
Application number
US17/194,761
Inventor
Kenji Osafune
Shimpei GOTOH
Isao Ito
Michiaki Mishima
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 US17/194,761 priority Critical patent/US20210284968A1/en
Publication of US20210284968A1 publication Critical patent/US20210284968A1/en
Pending 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
    • C12N5/0688Cells from the lungs or the respiratory tract
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/01Modulators of cAMP or cGMP, e.g. non-hydrolysable analogs, phosphodiesterase inhibitors, cholera toxin
    • 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
    • C12N2501/065Modulators of histone acetylation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/117Keratinocyte growth factors (KGF-1, i.e. FGF-7; KGF-2, i.e. FGF-12)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/16Activin; Inhibin; Mullerian inhibiting substance
    • 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
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/385Hormones with nuclear receptors of the family of the retinoic acid recptor, e.g. RAR, RXR; Peroxisome proliferator-activated receptor [PPAR]
    • 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
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • 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
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • 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
    • C12N2501/70Enzymes
    • C12N2501/72Transferases (EC 2.)
    • C12N2501/727Kinases (EC 2.7.)
    • 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
    • C12N2501/70Enzymes
    • C12N2501/73Hydrolases (EC 3.)
    • 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
    • C12N2501/999Small molecules not provided for elsewhere
    • 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
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem cells

Definitions

  • the present invention relates to a method for producing alveolar epithelial progenitor cells from pluripotent stem cells.
  • the present invention also relates to a kit used for producing alveolar epithelial progenitor cells from pluripotent stem cells.
  • the lung is one of the most complicated organs, and it is considered to be composed of approximately 40 different types of cells.
  • the pulmonary alveolus is composed of the alveolar space, which stores gas, and the alveolar epithelium, which surrounds the same.
  • the alveolar epithelium is composed of the type I alveolar epithelial cells and the type II alveolar epithelial cells.
  • the former forms a blood-air barrier with the microvascular endothelium surrounding the pulmonary alveolus with the aid of the basal membrane and exchanges the intra-alveolar gas with the blood gas.
  • the latter comprises many lamellar corpuscles, it undergoes exocytosis of pulmonary surfactants, and it forms the alveolar lining layer.
  • ES cells embryonic stem cells
  • iPS cells induced pluripotent stem cells
  • diseases that destroy the pulmonary alveolus include emphysema, interstitial pneumonia, and lymphangioleiomyomatosis.
  • emphysema is treated via symptomatic treatment or conservative treatment at present, and there is no radical treatment therefor.
  • radical treatment available for other pulmonary alveolar diseases, and the development of cell-transfer treatment has been accordingly awaited.
  • the present inventors have conducted concentrated studies in order to attain the above objects. As a result, they discovered that pluripotent stem cells could be induced to differentiate into alveolar epithelial progenitor cells with the use of various growth factors and compounds. This has led to the completion of the present invention.
  • the present invention includes the following.
  • Step (2) culturing the cells obtained in Step (1) in a medium containing a bone morphogenic protein (BMP) inhibitor and a TGF ⁇ inhibitor; and
  • BMP bone morphogenic protein
  • Step (3) culturing the cells obtained in Step (2) in a medium containing BMP4, retinoic acid, and a GSK3 ⁇ inhibitor.
  • Step (3) (4) culturing the cells obtained in Step (3) in a medium containing FGF10;
  • Step (4) culturing the cells obtained in Step (4) in a medium containing a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF.
  • FIG. 1A and FIG. 1B each show a scheme for producing alveolar epithelial progenitor cells from pluripotent stem cells.
  • “Stage I” is synonymous with “Step 1”
  • “Stage II” is synonymous with “Step 2”
  • “Stage III” is synonymous with “Step 3”
  • “Stage IV” is synonymous with “Step 4”
  • “Stage V” is synonymous with “Step 5.”
  • Stages” and “steps” are collectively referred to as “Steps.”
  • FIG. 2A shows images of immunostained CPM and NKX2-1 in the cells after the completion of Step 3.
  • FIG. 2B shows images of immunostained CPM and NKX2-1 in the cells after the completion of Step 2 (upper images) and Step 3 (lower images).
  • FIG. 3A shows percentages of CPM-positive cells after sorting via CPM-based MACS, after the completion of Step 3.
  • “Isotype control” shows the results for the negative control
  • “Pre-sorting” shows the results for the cells before sorting via MACS
  • “CPM positive selection” shows the results for CPM-positive cells after sorting via MACS
  • “CPM negative selection” shows the results for CPM-negative cells after sorting via MACS.
  • FIG. 3B shows percentages of CPM-positive cells after sorting via CPM-based MACS after the completion of Step 3.
  • Pre-sorting shows the results for the cells before sorting via MACS
  • CPM(+) sorting shows the results for the MACS-sorted CPM-positive cells
  • CPM( ⁇ ) sorting shows the results for the MACS-sorted CPM-negative cells.
  • FIG. 4A shows images of immunostained CPM and NKX2-1 in the MACS-sorted CPM-positive cells (CPM(+) sorting) and in the CPM-negative cells (CPM( ⁇ ) sorting) after the completion of Step 3.
  • FIG. 4B shows images of immunostained NKX2-1 in the MACS-sorted CPM-positive cells (CPM(+) sorting) and in the CPM-negative cells (CPM( ⁇ ) sorting) after the completion of Step 3.
  • FIG. 5A shows percentages of NKX2-1-positive cells among the MACS-sorted cells after the completion of Step 3.
  • Pre-sorting shows the results for the cells before sorting via MACS
  • CPM positive selection shows the results for the MACS-sorted CPM-positive cells
  • CPM negative selection shows the results for the MACS-sorted CPM-negative cells.
  • FIG. 5B shows percentages of NKX2-1-positive cells among the MACS-sorted cells after the completion of Step 3.
  • CPM + sorted cells shows percentages of NKX2-1-positive cells among the MACS-sorted CPM-positive cells (92.3 ⁇ 0.7%)
  • CPM ⁇ sorted cells shows percentages of NKX2-1-positive cells among the MACS-sorted CPM-negative cells (22.2 ⁇ 2.3%).
  • FIG. 6 shows the assay results of CPM mRNA levels (left) and NKX2-1 mRNA levels (right) in the MACS-sorted cells on the basis of CPM markers via quantitative PCR after the completion of Step 3.
  • Step 3 shows the results for the cells before sorting
  • Step 3 (+) shows the results for the CPM-positive cells
  • Step 3 ( ⁇ ) shows the results for the CPM-negative cells
  • Fetal Lung shows the results for the fetal pneumocytes
  • Adult Lung shows the results for the adult pneumocytes.
  • FIG. 7 shows the assay results of CPM mRNA levels (left) and NKX2-1 mRNA levels (right) in the MACS-sorted cells via quantitative PCR after the completion of Step 4.
  • Step 4 shows the results for the cells before sorting
  • Step 4 (+) shows the results for the CPM-positive cells
  • Step 4 ( ⁇ ) shows the results for the CPM-negative cells
  • Fetal Lung shows the results for the fetal pneumocytes
  • Adult Lung shows the results for the adult pneumocytes.
  • FIG. 8A shows images of immunostained iPS cells (201B7) in the MACS-sorted CPM-positive cells after the completion of Step 5.
  • FIG. 8B shows images of immunostained iPS cells (SFTPC reporter cells; SFTPC-reporter 201 B 7 ) in the MACS-sorted CPM-positive cells after the completion of Step 5.
  • FIG. 8C shows percentages of EGFP-positive iPS cells (SFTPC reporter cells; SFTPC-reporter 201B7) among the MACS-sorted CPM-positive cells after the completion of Step 5.
  • FIG. 9A and FIG. 9B each show an image of immunostained cells after the completion of Step 5.
  • FIG. 10 shows the assay results of mRNA levels via quantitative PCR of CPM, NKX2-1, SFTPA2, SFTPB, SFTPC, DCLAMP, CCSP (SCGB1A1), SCGB3A2, and NGFR in the MACS-sorted cells after the completion of Step 5.
  • Step 5 shows the results for the cells before sorting
  • Step 5 (+) shows the results for the CPM-positive cells
  • Step 5 ( ⁇ ) shows the results for the CPM-negative cells
  • “Fetal Lung” shows the results for the fetal pneumocytes
  • “Adult Lung” shows the results for the adult pneumocytes.
  • FIG. 11 shows images of CPM in the human fetal lung tissue subjected to staining together with NKX2-1, SFTPC, or T1 ⁇ serving as a marker of the prenatal period, the adenoid period, or the canalicular period of the lung, respectively.
  • FIG. 12 shows images of CPM in the mouse lung tissue at relevant prenatal periods (E12.5, E15.5, and E17.5) subjected to staining together with NKX2-1 serving as a marker of the prenatal period, the adenoid period, or the canalicular period of the lung, respectively.
  • FIG. 13 shows a summary of a method of sorting CPM-positive cells from among the cells after the completion of Step 3 and culturing the sorted cells together with human fetal pulmonary fibroblasts.
  • FIG. 14A shows transmission electron microscopic images of spheroids following the three-dimensional culture of the CPM-positive cells after the completion of Step 3.
  • the images at the center and on the right are each an enlarged view of a part of the image on the left.
  • FIG. 14B shows transmission electron microscopic images of type II alveolar epithelial cells of mouse lungs. The image on the right shows an enlarged view of a part of the image on the left.
  • FIG. 14C shows transmission electron microscopic images of the mouse fetal lung at E17.5. The image on the right shows an enlarged view of a part of the image on the left.
  • “Lu” indicates a lumen.
  • FIG. 15A shows hematoxylin-eosin-stained images of the three-dimensionally cultured CPM-positive cells (left) and CPM-negative cells (right) after the completion of Step 3.
  • the lower images show enlarged views of the upper images.
  • FIG. 15B shows images of the spheroids immunostained with the CPM, NKX2-1, GFP (SFTPC), SFTPC (endogenous), and AQP5 antibodies following the three-dimensional culture of the CPM-positive cells after the completion of Step 3.
  • FIG. 16 shows images of the spheroids immunostained with the CPM, NKX2-1, SFTPC, SFTPB, SFTPA, SFTPD, ID2, and SOX9 antibodies following the three-dimensional culture of the CPM-positive cells after the completion of Step 3.
  • the images on the right show images of alveolar epithelial cell-associated proteins subjected to dual staining with either CPM or NKX2-1.
  • FIG. 17 shows images of the spheroids immunostained with the T1 ⁇ , SFTPC, AQP5, and CAV1 antibodies following the three-dimensional culture of the CPM-positive cells after the completion of Step 3.
  • the present invention provides a method for producing alveolar epithelial progenitor cells (e.g., human alveolar epithelial progenitor cells) from pluripotent stem cells comprising steps of culturing pluripotent stem cells in (1) a medium containing activin A and a GSK3 ⁇ inhibitor, (2) a medium containing a BMP inhibitor and a TGF ⁇ inhibitor, and (3) a medium containing BMP4, retinoic acid, and a GSK3 ⁇ inhibitor.
  • alveolar epithelial progenitor cells e.g., human alveolar epithelial progenitor cells
  • the method for producing alveolar epithelial progenitor cells according to the present invention may comprise a step of extracting CPM-positive cells as the alveolar epithelial progenitor cells, following Step (3).
  • the method for producing alveolar epithelial progenitor cells according to the present invention may further comprise Step (4) of culture in a medium containing FGF10 and Step (5) of culture in a medium containing a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF, following Step (3).
  • the method may further comprise a step of extracting CPM-positive cells as the alveolar epithelial progenitor cells, following Step (5).
  • alveolar epithelial progenitor cells refers to progenitor cells of type I alveolar epithelial cells or type II alveolar epithelial cells, which express CPM or NKX2-1.
  • CPM indicates a polynucleotide shown in the NCBI Accession Number NM_001005502, NM_001874, or NM_198320 or a protein encoded thereby.
  • NKX2-1 indicates a polynucleotide shown in the NCBI Accession Number NM_001079668 or NM_003317 or a protein encoded thereby.
  • markers for alveolar epithelial progenitor cells include polynucleotides selected from the group consisting of SFTPB (NCBI Accession Numbers NM_000542 and NM_198843), SFTPC (NCBI Accession Numbers NM_001172357, NM_001172410, and NM_003018), and CCSP (NCBI Accession Number NM_003357) and proteins encoded by such polynucleotides.
  • a medium used in the step of culturing pluripotent stem cells according to the present invention can be prepared from a medium used for animal cell culture as a basal medium.
  • basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), ⁇ MEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media.
  • a medium may or may not contain blood serum.
  • a medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol.
  • a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts.
  • RPMI 1640 medium supplemented with B27 and antibiotics is preferable.
  • pluripotent stem cells are cultured in a medium prepared by supplementing the basal medium described above with activin A and a GSK3 ⁇ inhibitor.
  • an HDAC inhibitor may further be added.
  • Activin A is a homodimer with two beta A chains, the amino acid sequence of activin A is 100% homologous to that of a protein of a human, mouse, rat, pig, cow, or cat, and, accordingly, the relevant species are not particularly limited.
  • activin A is preferably of an active form with the N-terminal peptide being cleaved, and it is preferably a homodimer comprising, bound thereto via a disulfide bond, the Gly311-Ser426 fragment with the N-terminal peptide of the inhibin beta A chain (e.g., NCBI Accession Number NP_002183) being cleaved.
  • Such activin A is commercially available from, for example, Wako and R&D Systems.
  • the activin A concentration in the medium is, for example, 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 150 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 mg/ml, although the concentration is not limited thereto.
  • the concentration is preferably 100 ng/ml.
  • GSK3 62 inhibitor used herein is defined as a substance that inhibits kinase activity of the GSK-3 ⁇ protein (e.g., the capacity for phosphorylation of ( ⁇ -catenin), and many such substances are already known.
  • Examples thereof include: an indirubin derivative, such as BIO, which is also known as a GSK-3 ⁇ inhibitor IX (6-bromoindirubin-3′-oxime); a maleimide derivative, such as SB216763 (3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indo 1 -3-yl)-1H-pyrrole-2,5-dione); a phenyl ⁇ -bromomethylketone compound, such as a GSK-3 ⁇ inhibitor VII (4-dibromoacetophenone); a cell-membrane-permeable phosphorylated peptide, such as L803-mts, which is also known as a GSK-3 ⁇ peptide inhibitor (i.e., Myr-N-GKEAPPAPPQSpP-NH 2 ); and CHIR99021, such as 6-[2-[4-(2,4-dichlorophenyl)-5-(4-methyl-1H-imidazol-2-yl
  • a GSK-3 ⁇ inhibitor that can be preferably used in the present invention is CHIR99021.
  • the concentration of CHIR99021 in a medium is, for example, 1 nM, 10 nM, 50 nM, 100 nM, 500 nM, 750 nM, 1 ⁇ M, 1.5 ⁇ M, 2 ⁇ M, 2.5 ⁇ M, 3 ⁇ M, 3.5 ⁇ M, 4 ⁇ M, 4.5 ⁇ M, 5 ⁇ M, 6 ⁇ M, 7 ⁇ M, 8 ⁇ M, 9 ⁇ M, 10 ⁇ M, 15 ⁇ M, 20 ⁇ M, 25 ⁇ M, 30 ⁇ M, 40 ⁇ M, or 50 ⁇ M, although the concentration is not limited thereto.
  • the concentration is preferably 1 ⁇ M.
  • HDAC inhibitor is defined as a substance that inhibits or inactivates enzyme activity of histone deacetylase (HDAC).
  • HDAC histone deacetylase
  • examples thereof include low-molecular-weight inhibitors, such as valproic acid (VPA) (Nat. Biotechnol., 26 (7): 795-797, 2008), trichostatin A, sodium butyrate (NaB), MC 1293, and M344; nucleic acid-based expression inhibitors such as siRNAs and shRNAs against HDAC (e.g., HDAC1 siRNA Smartpool ⁇ (Millipore) and HuSH 29mer shRNA Constructs against HDAC1 (OriGene)); and DNA methyltransferase inhibitors (e.g., 5′-azacytidine) (Nat. Biotechnol., 26 (7): 795-797, 2008).
  • VPA valproic acid
  • NaB sodium butyrate
  • MC 1293 MC 1293,
  • An HDAC inhibitor that can be preferably used in the present invention is sodium butyrate (NaB).
  • the concentration of sodium butyrate (NaB) in a medium is, for example, 1 ⁇ M, 10 ⁇ M, 50 ⁇ M, 100 ⁇ M, 250 ⁇ M, 500 ⁇ M, 750 ⁇ M, 1 mM, 2 mM, 3 mM, 4 mM, or 5 mM, although the concentration is not limited thereto.
  • the concentration is preferably 250 ⁇ M.
  • culture may be conducted in a culture vessel treated with a coating agent.
  • a coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may comprise a process of pluripotent stem cell detachment.
  • methods for cell detachment include a method of mechanical detachment and a method of cell detachment involving the use of a cell detachment solution having protease activity and collagenase activity (e.g., AccutaseTM and AccumaxTM or a cell detachment solution having collagenase activity alone. It is preferable that human pluripotent stem cells be detached with the use of a cell detachment solution having protease activity and collagenase activity (and the use of AccutaseTM is particularly preferable).
  • an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • ROCK inhibitor is not particularly limited, provided that it can inhibit functions of Rho kinase (ROCK).
  • ROCK Rho kinase
  • examples thereof include: Y-27632 ((+)-(R)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide dihydrochloride) (e.g., Ishizaki et al., Mol.
  • ROCK inhibitors RNA interference-inducible nucleic acids
  • dominant-negative variants RNA interference-inducible nucleic acids
  • expression vectors thereof Since other low-molecular-weight compounds are known as ROCK inhibitors, such compounds and derivatives thereof can also be used in the present invention (e.g., U.S. Patent Application Publication Nos.
  • ROCK inhibitors can be used.
  • An ROCK inhibitor that can be preferably used in the present invention is Y-27632.
  • the Y-27632 concentration is, for example, 100 nM, 500 nM, 750 nM, 1 ⁇ M, 2 ⁇ M, 3 ⁇ M, 4 ⁇ M, 5 ⁇ M, 6 ⁇ M, 7 ⁇ M, 8 ⁇ M, 9 ⁇ M, 10 ⁇ M, 15 ⁇ M, 20 ⁇ M, 25 ⁇ M, 30 ⁇ M, 40 ⁇ M, or 50 ⁇ M, although the concentration is not limited thereto.
  • the concentration is preferably 10 ⁇ M.
  • culture is conducted at about 30 ⁇ C to 40 ⁇ C, and preferably at about 37 ⁇ 0 C, although the temperature is not limited thereto.
  • Culture is conducted under an atmosphere of air containing CO 2 , and the CO 2 concentration is preferably about 2% to 5%.
  • the culture period is not particularly limited because long-term culture would not cause any problems.
  • the culture period may be at least 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, or 12 days.
  • the culture period is preferably at least 6 days, and it is particularly preferably 6 days.
  • the duration of addition is 1 day or 2 days, with 1 day being preferable.
  • the HDAC inhibitor is further added, the addition is initiated on the day following the initiation of the step, and culture is conducted for at least 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, or 11 days. Culture is preferably conducted for at least 5 days, and particularly preferably for 5 days, in the presence of the HDAC inhibitor.
  • a medium used in this step can be prepared from a medium used for animal cell culture as a basal medium.
  • basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), ⁇ MEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media.
  • a medium may or may not contain blood serum.
  • a medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol.
  • a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts.
  • a medium mixture of DMEM and Ham's F12 supplemented with Glutamax, B27, N2, 3′-thiol glycerol, and ascorbic acid is preferable.
  • the cells obtained in the previous step i.e., the step of culture of pluripotent stem cells in a medium containing activin A and a GSK3 ⁇ inhibitor
  • a medium prepared by supplementing the basal medium with a BMP inhibitor and a TGF ⁇ inhibitor are cultured in a medium prepared by supplementing the basal medium with a BMP inhibitor and a TGF ⁇ inhibitor.
  • BMP inhibitors include: protein-based inhibitors, such as Chordin, Noggin, and Follistatin; dorsomorphin (i.e., 6-[4-(2-piperidin-1-yl-ethoxy)phenyl]-3-pyridin-4-yl-pyrazolo[1,5-a]pyrimidine) and a derivative thereof (P. B. Yu et al., 2007, Circulation, 116: II_60; P. B. Yu et al., 2008, Nat. Chem. Biol., 4:33-41; J.
  • protein-based inhibitors such as Chordin, Noggin, and Follistatin
  • dorsomorphin i.e., 6-[4-(2-piperidin-1-yl-ethoxy)phenyl]-3-pyridin-4-yl-pyrazolo[1,5-a]pyrimidine
  • LDN-193189 i.e., 4-(6-(4-(piperazin-1-yl)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinoline.
  • Dorsomorphin and LDN-193189 are commercially available from Sigma-Aldrich and Stemgent, respectively.
  • a BMP inhibitor that can be preferably used in the present invention is Noggin.
  • the concentration of Noggin in a medium is not particularly limited, provided that BMP can be inhibited.
  • concentration is 1 ng/ml, 10 ng/ml, 50 ng/ml, 100 ng/ml, 200 ng/ml, 30 ng/ml, 400 ng/ml, 50 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, 1 ⁇ g/ml, or 2 ⁇ g/ml, although the concentration is not limited thereto.
  • the concentration is preferably 200 ng/ml.
  • TGF ⁇ inhibitor refers to a substance that inhibits signal transmission from the binding of TGF ⁇ to a receptor leading to SMAD.
  • a TGF ⁇ inhibitor is not particularly limited, provided that such substance inhibits TGF ⁇ from binding to a receptor; i.e., the ALK family, or such substance inhibits phosphorylation of SMAD caused by the ALK family. Examples thereof include Lefty- 1 (e.g., NCBI Accession Nos.
  • SB431542 (4-(4-(benzo[d][1,3]dioxo 1 -5-yl)-5-(pyridin-2-yl)-1H-imidazol-2-yl)benzamide), SB202190 (R. K. Lindemann, et al., Mol. Cancer, 2003, 2: 20), SB505124 (GlaxoSmithKline), NPC30345, SD093, SD908, SD208 (Scios), LY2109761, LY364947, LY580276 (Lilly Research Laboratories), A-83-01 (WO 2009/146408), and derivatives thereof.
  • a TGF ⁇ inhibitor that can be preferably used in the present invention is SB431542.
  • the SB431542 concentration in a medium is not particularly limited, provided that TGF ⁇ is inhibited.
  • concentration can be 1 ⁇ M, 2 ⁇ M, 3 ⁇ M, 4 ⁇ M, 5 ⁇ M, 6 ⁇ M, 7 ⁇ M, 8 ⁇ M, 9 ⁇ M, 10 ⁇ M, 15 ⁇ M, 20 ⁇ M, 25 ⁇ M, 30 ⁇ M, 35 ⁇ M, 40 ⁇ M, 45 ⁇ M, 50 ⁇ M, 60 ⁇ M, 70 ⁇ M, 80 ⁇ M, 90 ⁇ M, 100 ⁇ M, 200 ⁇ M, 300 ⁇ M, 400 ⁇ M, or 500 ⁇ M, although the concentration is not limited thereto.
  • the concentration is preferably 10 ⁇ M.
  • culture may be conducted in a culture vessel treated with a coating agent.
  • coating agents include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above.
  • cells may be detached and reseeded in a culture vessel.
  • particular cells may be selected, and, for example, SOX17- and/or FOXA2-positive cells may be selected and used in this step.
  • This method is preferably implemented by means of media exchange.
  • an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • culture is conducted at about 30 ⁇ C to 40 ⁇ C, and preferably at about 37 ⁇ C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO 2 , and the CO 2 concentration is preferably about 2% to 5%.
  • the culture period is not particularly limited because long-term culture would not cause any problems.
  • the culture period may be at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 8 days.
  • the culture period is preferably 4 days.
  • a medium used in this step can be prepared from a medium used for animal cell culture as a basal medium.
  • basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), ⁇ MEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media.
  • a medium may or may not contain blood serum.
  • a medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol.
  • a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts.
  • a medium mixture of DMEM and Ham's F12 supplemented with Glutamax, B27, N2, 3′-thiol glycerol, and ascorbic acid is preferable.
  • the cells obtained in the previous step i.e., the step of culture in a medium containing a BMP inhibitor and a TGF ⁇ inhibitor
  • a medium prepared by supplementing the basal medium with BMP4, retinoic acid, and a GSK3 ⁇ inhibitor are cultured in a medium prepared by supplementing the basal medium with BMP4, retinoic acid, and a GSK3 ⁇ inhibitor.
  • BMP4 refers to a protein encoded by the polynucleotide shown in the NCBI Accession Number NM_001202, NM_130850, or NM_130851, and it may be in an active form resulting from cleavage by a protease.
  • the BMP4 concentration in a medium is not particularly limited.
  • such concentration may be 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 ⁇ g/ml, although the concentration is not limited thereto.
  • the concentration is preferably 100 ng/ml.
  • ATRA all-trans retinoic acid
  • AM580 4-[[(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)carbonyl]amino]-benzoic acid (AM580) (Tamura, K. et al., Cell Differ.
  • TNPB 4-[(1E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)-1-propen-1-yl]-benzoic acid (TTNPB) (Strickland, S., et al., Cancer Res., 43: 5268-5272, 1983), retinol palmitate, retinol, retinal, 3-dehydroretinoic acid, 3-dehydroretinol, 3-dehydroretinal, and compounds described in Abe, E., et al., Proc. Natl. Acad.
  • the retinoic acid concentration in a medium is not particularly limited.
  • such concentration can be 1 nM, 5 nM, 10 nM, 15 nM, 20 nM, 25 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, or 1 ⁇ M, although the concentration is not limited thereto.
  • the concentration is preferably 50 nM.
  • the GSK3 ⁇ inhibitor as described above can be used in this step, and the GSK3 ⁇ inhibitor is preferably CHIR99021.
  • the CHIR99021 concentration in a medium is, for example, 1 nM, 10 nM, 50 nM, 100 nM, 500 nM, 750 nM, 1 ⁇ M, 1.5 ⁇ M, 2 ⁇ M, 2.5 ⁇ M, 3 ⁇ M, 3.5 ⁇ M, 4 ⁇ M, 4.5 ⁇ M, 5 ⁇ M, 6 ⁇ M, 7 ⁇ M, 8 ⁇ M, 9 ⁇ M, 10 ⁇ M, 15 ⁇ M, 20 ⁇ M, 25 ⁇ M, 30 ⁇ M, 40 ⁇ M, or 50 ⁇ M, although the concentration is not limited thereto.
  • the concentration is preferably 2.5 ⁇ M.
  • culture may be conducted in a culture vessel treated with a coating agent.
  • a coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above.
  • cells may be detached and reseeded in a culture vessel.
  • particular cells may be selected, and, for example, SOX2-, SOX17-, and/or FOXA2-positive cells may be selected and used in this step.
  • This method is preferably implemented by means of media exchange.
  • an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • culture is conducted at about 30 ⁇ C to 40 ⁇ C, and preferably at about 37 ⁇ C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO 2 , and the CO 2 concentration is preferably about 2% to 5%.
  • the culture period is not particularly limited because long-term culture would not cause any problems.
  • the culture period may be at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 8 days.
  • the culture period is preferably at least 4 days, and more preferably 4 days.
  • a medium used in this step can be prepared from a medium used for animal cell culture as a basal medium.
  • basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), ⁇ MEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media.
  • a medium may or may not contain blood serum.
  • a medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol.
  • a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts.
  • a medium mixture of DMEM and Ham's F12 supplemented with Glutamax, B27, N2, 3′-thiol glycerol, and ascorbic acid is preferable.
  • the cells obtained in the previous step i.e., the step of culture in a medium containing BMP4, retinoic acid, and a GSK3 ⁇ inhibitor
  • a medium prepared by supplementing the basal medium with FGF10 i.e., the step of culture in a medium containing BMP4, retinoic acid, and a GSK3 ⁇ inhibitor
  • FGF10 refers to a protein encoded by the polynucleotide shown in the NCBI Accession Number NM_004465, and it may be in an active form resulting from cleavage by a protease.
  • FGF10 is commercially available from, for example, Life Technologies or Wako.
  • the FGF10 concentration in a medium is not particularly limited.
  • such concentration may be 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 ⁇ g/ml, although the concentration is not limited thereto.
  • the concentration is preferably 100 ng/ml.
  • culture may be conducted in a culture vessel treated with a coating agent.
  • a coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above.
  • cells may be detached and reseeded in a culture vessel.
  • particular cells may be selected, and, for example, NKX2-1- and/or FOXA2-positive cells may be selected and used in this step.
  • This method is preferably implemented by means of media exchange.
  • an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • culture is conducted at about 30 ⁇ C to 40 ⁇ C, and preferably at about 37 ⁇ C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO 2 , and the CO 2 concentration is preferably about 2% to 5%.
  • the culture period is not particularly limited because long-term culture would not cause any problems.
  • the culture period may be at least 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days.
  • the culture period is preferably at least 7 days, and more preferably 7 days.
  • a medium used in this step can be prepared from a medium used for animal cell culture as a basal medium.
  • basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), ⁇ MEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media.
  • a medium may or may not contain blood serum.
  • a medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, ITS Premix, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol.
  • a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts.
  • Ham's F12 medium containing albumin, buffer agents (e.g., HEPES), calcium chloride, ITS Premix, and antibiotics is preferable.
  • the cells obtained in the previous step are cultured in a medium prepared by supplementing the basal medium with a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF.
  • steroid drug refers to a steroidal anti-inflammatory drug, such as glucocorticoid or a synthetic derivative thereof. Specific examples thereof include hydrocortisone, hydrocortisone succinate, prednisolone, methylprednisolone, methylprednisolone succinate, triamcinolone, triamcinolone acetonide, dexamethasone, and betamethasone.
  • a steroid drug that can be preferably used in the present invention is dexamethasone.
  • the dexamethasone concentration in a medium is not particularly limited.
  • such concentration may be 1 nM, 5 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, or 1 ⁇ M, although the concentration is not limited thereto.
  • the concentration is preferably 50 nM.
  • cAMP derivative refers to a compound with a modified cyclic AMP substituent. Examples thereof include cyclic adenosine monophosphate (cAMP), 8-bromo cyclic adenosine monophosphate (8-Br-cAMP or 8Br-cAMP), 8-chloro-cyclic adenosine monophosphate (8-Cl-cAMP), 8-(4-chlorophenylthio)cyclic adenosine monophosphate (8-CPT-cAMP), and dibutyryl cyclic adenosine monophosphate (DB-cAMP).
  • cAMP cyclic adenosine monophosphate
  • 8-Br-cAMP or 8Br-cAMP 8-chloro-cyclic adenosine monophosphate
  • 8-Cl-cAMP 8-(4-chlorophenylthio)cyclic adenosine monophosphate
  • DB-cAMP dibutyryl cyclic a
  • a cAMP derivative that can be preferably used in the present invention is 8-Br-cAMP.
  • the concentration of 8-Br-cAMP in a medium is not particularly limited.
  • concentration can be 1 ⁇ M, 5 ⁇ M, 10 ⁇ M, 20 ⁇ M, 30 ⁇ M, 40 ⁇ M, 50 ⁇ M, 60 ⁇ M, 70 ⁇ M, 80 ⁇ M, 90 ⁇ M, 100 ⁇ M, 200 ⁇ M, 300 ⁇ M, 400 ⁇ M, 500 ⁇ M, 600 ⁇ M, 700 ⁇ M, 800 ⁇ M, 900 ⁇ M, or 1 mM, although the concentration is not limited thereto.
  • the concentration is preferably 100 ⁇ M.
  • phosphodiesterase inhibitor used herein refers to a compound that inhibits phosphodiesterase (PDE), so as to increase the concentration of cAMP or cGMP in the cells.
  • PDE phosphodiesterase
  • examples thereof include 1,3-dimethylxanthine, 6,7-dimethoxy-1-(3,4-dimethoxybenzyl)isoquinoline, 4- ⁇ [3′,4′-(methylenedioxy)benzyl]amino ⁇ -6-methoxyquinazoline, 8-methoxymethyl-3-isobutyl-1-methylxanthine, and 3-isobutyl-1-methylxanthine (IBMX).
  • a phosphodiesterase inhibitor that can be preferably used in the present invention is IBMX.
  • the IBMX concentration in a medium is not particularly limited.
  • such concentration can be 1 ⁇ M, 5 ⁇ M, 10 ⁇ M, 20 ⁇ M, 30 ⁇ M, 40 ⁇ M, 50 ⁇ M, 60 ⁇ M, 70 ⁇ M, 80 ⁇ M, 90 ⁇ M, 100 ⁇ M, 200 ⁇ M, 300 ⁇ M, 400 ⁇ M, 500 ⁇ M, 600 ⁇ M, 700 ⁇ M, 800 ⁇ M, 900 ⁇ M, or 1 mM, although the concentration is not limited thereto.
  • the concentration is preferably 100 ⁇ M.
  • KGF refers to a protein encoded by the polynucleotide shown in the NCBI Accession Number NM_002009, and it may be in an active form resulting from cleavage by a protease.
  • KGF is commercially available from, for example, Wako.
  • the concentration of KGF in a medium is not particularly limited.
  • such concentration can be 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 ⁇ g/ml, although the concentration is not limited thereto.
  • the concentration is preferably 100 ng/ml.
  • culture may be conducted in a culture vessel treated with a coating agent.
  • a coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above.
  • cells may be detached and reseeded in a culture vessel.
  • particular cells may be selected, and, for example, NKX2-1-positive cells may be selected and used in this step.
  • This method is preferably implemented by means of media exchange.
  • an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • culture is conducted at about 30 ⁇ C to 40 ⁇ C, and preferably at about 37 ⁇ C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO 2 , and the CO 2 concentration is preferably about 2% to 5%.
  • the culture period is not particularly limited because long-term culture would not cause any problems.
  • the culture period may be at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 8 days.
  • the culture period is preferably at least 4 days, and it is more preferably 4 days.
  • the present invention provides a method for three-dimensional culture of alveolar epithelial progenitor cells for further maturation of alveolar epithelial progenitor cells.
  • three-dimensional culture is carried out by subjecting cells to float culture in the form of cell masses (i.e., spheroids).
  • spheroids i.e., spheroids
  • three-dimensional culture can be carried out with the use of, for example, Cell Culture Inserts provided by BD.
  • three-dimensional culture may be conducted in the presence of other cell species.
  • other cell species include human pulmonary fibroblasts and human fetal pulmonary fibroblasts.
  • Such cells are commercially available from, for example, American Type Culture Collection (ATCC) and DV Biologics.
  • the medium used for three-dimensional culture according to the present invention may be a medium that is used in the step of culture conducted in a medium containing a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF.
  • a medium supplemented with an extracellular matrix may be preferable.
  • the ratio of the volume of the extracellular matrix to the volume of the medium is not particularly limited. For example, these substances can be mixed at a ratio of 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, or 1:5.
  • an extracellular matrix is a supramolecular structure that exists outside the cell, and it may be a naturally occurring or artificial (recombinant) structure.
  • extracellular matrices examples thereof include substances such as collagen, proteoglycan, fibronectin, hyaluronic acid, tenascin, entactin, elastin, fibrillin, and laminin, and fragments thereof.
  • extracellular matrices may be used in combination.
  • extracellular matrices may be prepared from cells such as BD MatrigelTM.
  • An example of an artificial structure is a laminin fragment.
  • the period for three-dimensional culture is not particularly limited because long-term culture would not cause any problems.
  • the culture period may be at least 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, or 12 days.
  • the culture period is preferably at least 10 days, and particularly preferably 10 days, 11 days, or 12 days.
  • Pluripotent stem cells that can be used in the present invention are stem cells that have the potential to differentiate into any types of cells existing in organisms (i.e., pluripotency) and have the potential to grow. Examples thereof include embryonic stem cells (ES cells), nuclear transfer-derived embryonic stem cells from cloned embryos (ntES cells), germline stem cells (GS cells), embryonic germ cells (EG cells), induced pluripotent stem cells (iPS cells), and pluripotent cells derived from cultured fibroblasts and myeloid stem cells (Muse cells).
  • ES cells embryonic stem cells
  • ntES cells nuclear transfer-derived embryonic stem cells from cloned embryos
  • GS cells germline stem cells
  • EG cells embryonic germ cells
  • iPS cells induced pluripotent stem cells
  • the use of iPS cells or Muse cells is preferable because cells of interest can be obtained without destroying
  • ES cells are pluripotent stem cells having the potential to grow through autoreproduction, and they are established from embryoblasts of early embryos (e.g., blastocysts) of mammalians such as humans or mice.
  • ES cells are embryo-derived stem cells originating from embryoblasts of blastocysts, which are embryos after the 8 -cell stage and the morula stage of fertilized eggs. Such ES cells have the potential to differentiate into any types of cells constituting an adult; that is, so-called pluripotency and potential to grow through autoreproduction. ES cells were discovered in mice in 1981 (M. J. Evans and M. H. Kaufman, 1981, Nature 292: 154-156). Thereafter, ES cells of primates, such as humans and monkeys, were also established (J. A. Thomson, et al., 1998, Science 282: 1145-1147; J. A. Thomson, et al., 1995, Proc. Natl. Acad.
  • ES cells can be established by extracting embryoblasts from blastocysts of fertilized eggs of target animals and culturing the embryoblasts in fibroblast feeders. Cells can be maintained via subculture with the use of a medium supplemented with substances such as leukemia inhibitory factors (LIF) and basic fibroblast growth factors (bFGF).
  • LIF leukemia inhibitory factors
  • bFGF basic fibroblast growth factors
  • Human and monkey ES cells can be established and maintained by the methods described in, for example, U.S. Pat. No. 5,843,780; Thomson J. A., et al., 1995, Proc. Natl. Acad. Sci., U.S.A., 92: 7844-7848; Thomson, J. A., et al., 1998, Science 282: 1145-1147; H.
  • Human ES cells can be maintained with the use of a medium for the production of ES cells, such as a DMEM/F-12 medium supplemented with 0.1 mM 2-mercaptoethanol, 0.1 mM nonessential amino acids, 2 mM L-glutamic acid, 20% KSR, and 4 ng/ml bFGF, at 37 ⁇ C in the presence of 5% CO 2 in a moist atmosphere (H. Suemori, et al., 2006, Biochem. Biophys. Res. Commun., 345: 926-932). It is necessary that ES cells be subjected to subculture every 3 or 4 days. Subculture can be carried out with the use of 0.25% trypsin and 0.1 mg/ml collagenase IV in PBS containing 1 mM CaCl 2 and 20% KSR.
  • a medium for the production of ES cells such as a DMEM/F-12 medium supplemented with 0.1 mM 2-mercaptoethanol, 0.1 mM nonessential
  • ES cells can be selected via real-time PCR using the expression of a gene marker such as alkaline phosphatase, Oct-3/4, or Nanog as an indicator.
  • a gene marker such as alkaline phosphatase, Oct-3/4, or Nanog
  • OCT-3/4, NANOG, or ECAD can be employed as an indicator (E. Kroon et al., 2008, Nat. Biotechnol., 26: 443-452).
  • Human ES cells e.g., WA01 (H1) and WA09 (H9) are available from the WiCell Research Institute, and KhES-1, KhES-2, and KhES-3 are available from the Institute for Frontier Medical Sciences, Kyoto University (Kyoto, Japan).
  • Germline stem cells are spermary-derived pluripotent stem cells that serve as sources for spermatogenesis. As with the case of ES cells, germline stem cells can be differentiated into various types of cells. For example, germline stem cells may be implanted into mouse blastocysts, so that chimeric mice may be produced (M. Kanatsu-Shinohara et al., 2003, Biol. Reprod., 69: 12-616; K. Shinohara et al., 2004, Cell, 119: 1001-1012). Germline stem cells are capable of autoreproduction in a medium containing glial cell line-derived neurotrophic factors (GDNF).
  • GDNF glial cell line-derived neurotrophic factors
  • germline stem cells can be obtained by repeating subculture under the same culture conditions as with those used for ES cells (Masanori Takebayashi et al., 2008, Experimental Medicine, Vol. 26, No. 5 (extra edition), pp. 41-46, Yodosha, Tokyo, Japan).
  • embryonic germ cells are pluripotent cells that are established from primordial germ cells during the prenatal period.
  • Embryonic germ cells can be established by culturing primordial germ cells in the presence of substances such as LIF, bFGF, or stem cell factors (Y. Matsui et al., 1992, Cell, 70: 841-847; J. L. Resnicket al., 1992, Nature, 359: 550-551).
  • iPS cells can be prepared by introducing particular reprogramming factors into somatic cells in the form of DNA or proteins.
  • iPS cells are artificial stem cells derived from somatic cells that have substantially the same properties as ES cells, such as pluripotency and potential to grow through autoreproduction (K. Takahashi and S. Yamanaka, 2006, Cell, 126: 663-676; K. Takahashi et al., 2007, Cell, 131: 861-872; J. Yu et al., 2007, Science, 318: 1917-1920; Nakagawa, M. et al., Nat. Biotechnol., 26: 101-106, 2008; WO 2007/069666).
  • Reprogramming factors may be 4 composed of genes that are expressed specifically in ES cells, gene products or non-cording RNA thereof, or genes that play key roles in maintenance of the undifferentiated state of ES cells, gene products or non-coding RNA thereof, or low-molecular-weight compounds.
  • genes included in reprogramming factors include Oct3/4, Sox2, Sox1, Sox3, Sox15, Sox17, K1f4, K1f2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15-2, Tcll, beta-catenin, Lin28b, Sa111, Sa114, Esrrb, Nr5a2, Tbx3, and Glis1.
  • Such reprogramming factors may be used alone or in combination. Examples of combinations of reprogramming factors are described in WO 2007/069666, WO 2008/118820, WO 2009/007852, WO 2009/032194, WO 2009/058413, WO 2009/057831, WO 2009/075119, WO 2009/079007, WO 2009/091659, WO 2009/101084, WO 2009/101407, WO 2009/102983, WO 2009/114949, WO 2009/117439, WO 2009/126250, WO 2009/126251, WO 2009/126655, WO 2009/157593, WO 2010/009015, WO 2010/033906, WO 2010/033920, WO 2010/042800, WO 2010/050626, WO 2010/056831, WO 2010/068955, WO 2010/098419, WO 2010/102267, WO 2010/111409, WO 2010/111422, WO 2010/115050, WO 2010/124
  • HDAC histone deacetylase
  • VPA valproic acid
  • trichostatin A sodium butyrate
  • MC 1293 trichostatin A
  • M344 nucleic acid-based expression inhibitors
  • siRNAs and shRNAs against HDAC e.g., HDAC1 siRNA Smartpoor (Millipore) and HuSH 29mer shRNA constructs against HDAC1 (OriGene)
  • MEK inhibitors e.g., PD184352, PD98059, U0126, SL327, and PD0325901
  • glycogen synthase kinase-3 inhibitors e.g., Bio and CHIR99021
  • DNA methyltransferase inhibitors e.g., 5-azacytidine
  • histone methyltransferase inhibitors e.g., low-
  • reprogramming factors are in the form of proteins, for example, they may be introduced into somatic cells by a technique such as lipofection, fusion with cell-permeable peptides (e.g., HIV-derived TAT and polyarginine), or microinjection.
  • a technique such as lipofection, fusion with cell-permeable peptides (e.g., HIV-derived TAT and polyarginine), or microinjection.
  • reprogramming factors in the form of DNA can be introduced into somatic cells by a technique involving the use of a vector such as a virus, plasmid, or artificial chromosome vector, lipofection, a technique involving the use of a liposome, or microinjection, for example.
  • virus vectors include retrovirus vectors, lentivirus vectors (Cell, 126, pp. 663-676, 2006; Cell, 131, pp. 861-872, 2007; Science, 318, pp. 1917-1920, 2007), adenovirus vectors (Science, 322, 945-949, 2008), adeno-associated virus vectors, and Sendai virus vectors (WO 2010/008054).
  • artificial chromosome vectors examples include human artificial chromosome (HAC) vectors, yeast artificial chromosome (YAC) vectors, and bacterial artificial chromosome (BAC, PAC) vectors. Plasmids for mammalian animal cells can be used (Science, 322: 949-953, 2008). Vectors can comprise regulatory sequences, such as promoters, enhancers, ribosome-binding sequences, terminators, or polyadenylation sites, so that nuclear reprogramming substances can express.
  • HAC human artificial chromosome
  • YAC yeast artificial chromosome
  • BAC bacterial artificial chromosome
  • Plasmids for mammalian animal cells can be used (Science, 322: 949-953, 2008).
  • Vectors can comprise regulatory sequences, such as promoters, enhancers, ribosome-binding sequences, terminators, or polyadenylation sites, so that nuclear reprogramming substances can express.
  • vectors can comprise selection marker sequences, such as drug tolerance genes (e.g., kanamycin tolerance genes, ampicillin tolerance genes, and puromycin tolerance genes), thymidine kinase genes, or diphtheria toxin genes, and reporter gene sequences, such as green fluorescent proteins (GFP), P-glucuronidase (GUS), or FLAG, according to need.
  • drug tolerance genes e.g., kanamycin tolerance genes, ampicillin tolerance genes, and puromycin tolerance genes
  • thymidine kinase genes e.g., ampicillin tolerance genes, and puromycin tolerance genes
  • reporter gene sequences such as green fluorescent proteins (GFP), P-glucuronidase (GUS), or FLAG
  • reprogramming factors are in the form of RNA, for example, they may be introduced into somatic cells by a technique such as lipofection or microinjection.
  • RNA comprising 5-methylcytidine and pseudouridine (TriLink Biotechnologies) incorporated therein may be used, so as to suppress degradation (Warren L, 2010, Cell Stem Cell 7: 618-630).
  • culture media used for iPS cell induction include DMEM containing 10% to 15% FBS, a DMEM/F12 or DME medium (such medium may adequately contain, for example, LIF, penicillin/streptomycin, puromycin, L-glutamine, nonessential amino acids, and (3-mercaptoethanol), commercially available culture media (e.g., a medium for mouse ES cell culture; TX-WES medium, Thrombo X), a medium for primate ES cell culture (a medium for primate ES/iPS cell culture, ReproCELL Incorporated), and a serum-free medium (mTeSR, Stemcell Technology).
  • DMEM containing 10% to 15% FBS
  • a DMEM/F12 or DME medium such medium may adequately contain, for example, LIF, penicillin/streptomycin, puromycin, L-glutamine, nonessential amino acids, and (3-mercaptoethanol
  • commercially available culture media e.g., a medium for mouse
  • somatic cells are brought into contact with reprogramming factors in a 10% FBS-containing DMEM or DMEM/F12 medium, culture is conducted at 37 ⁇ C in the presence of 5% CO 2 for about 4 to 7 days, and the cells are reseeded in feeder cells (e.g., mitomycin C-treated STO cells or SNL cells).
  • feeder cells e.g., mitomycin C-treated STO cells or SNL cells.
  • Culture is reinitiated in a medium for bFGF-containing primate ES cell culture about 10 days after the somatic cells are first brought into contact with the reprogramming factors, and iPS-like colonies can then be formed at least about 30 to 45 days after such contact.
  • culture may be conducted in a 10% FBS-containing DMEM medium (this medium can further contain LIF, penicillin/streptomycin, puromycin, L-glutamine, nonessential amino acids, (3-mercaptoethanol, or the like, according to need) in feeder cells (e.g., mitomycin C-treated STO cells or SNL cells) at 37 ⁇ C in the presence of 5% CO 2 , and ES-like colonies can then be formed at least about 25 to 30 days later.
  • feeder cells e.g., mitomycin C-treated STO cells or SNL cells
  • somatic cells to be reprogrammed instead of feeder cells is preferable (Takahashi K, et al., 2009, PLoS One, 4: e8067 or WO 2010/137746), or use of an extracellular matrix (e.g., laminin-5 (WO 2009/123349) and Matrigel (BD)) is preferable.
  • extracellular matrix e.g., laminin-5 (WO 2009/123349) and Matrigel (BD)
  • culture may be conducted with the use of a serum-free medium (Sun N, et al., 2009, Proc. Natl. Acad. Sci., U.S.A. 106: 15720-15725).
  • iPS cells may be established under low-oxygen conditions (oxygen concentration of 0.1 % to 15%) (Yoshida Y, et al., 2009, Cell Stem Cell, 5: 237-241 or WO 2010/013845).
  • somatic cells used for nuclear reprogramming is not limited, and it is about 5 ⁇ 10 3 to about 5 ⁇ 10 6 cells per 100 cm 2 of a culture dish.
  • iPS cells can be selected in accordance with the configuration of the formed colonies.
  • drug tolerance genes that express in association with genes that express upon reprogramming of somatic cells e.g., Oct3/4 and Nanog
  • somatic cells e.g., Oct3/4 and Nanog
  • culture can be conducted in a medium containing corresponding drugs (i.e., a selection medium).
  • marker genes are fluorescent protein genes
  • luminescent enzyme genes luminescent substrates may be added.
  • marker genes are chromogenic enzyme genes, chromogenic substrates may be added.
  • iPS cells can be selected.
  • somatic cells refers to any animal cells except for germline cells or pluripotent cells such as egg cells, oocytes, and ES cells (preferably mammalian animal cells, including those of humans).
  • somatic cells include, but are not limited to, embryonic (fetal) somatic cells, neonatal (fetal) somatic cells, and mature healthy or affected somatic cells. Somatic cells may be primary-cultured cells, subcultured cells, or established cells.
  • somatic cells include: (1) tissue stem cells, such as neural stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells (i.e., somatic stem cells); (2) tissue progenitor cells; (3) differentiated cells, such as lymphocytes, epidermic cells, endothelial cells, muscle cells, fibroblasts (e.g., skin cells), hair cells, hepatic cells, gastric mucosal cells, intestinal cells, splenic cells, pancreatic cells (e.g., pancreatic exocrine cells), brain cells, pneumocytes, nephrocytes, and adipocytes.
  • tissue stem cells such as neural stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells (i.e., somatic stem cells)
  • tissue progenitor cells such as lymphocytes, epidermic cells, endothelial cells, muscle cells, fibroblasts (e.g., skin cells), hair cells
  • somatic cells When iPS cells are used as materials for transplantation, use of somatic cells having the same or substantially the same HLA genotype as that of a recipient is preferable, so that rejection would not occur.
  • HLA genotypes are “substantially the same,” such HLA genotypes are concordant with each other to the extent that an immunosuppressive agent is able to suppress immune responses to the transplanted cells.
  • somatic cells have HLA genotypes exhibiting concordance in 3 loci; i.e., HLA-A, HLA-B, and HLA-DR, or in 4 loci; i.e., HLA-A, HLA-B, HLA-DR, and HLA-C.
  • nt ES cells are nuclear transfer-derived ES cells produced from cloned embryos, and such ES cells have substantially the same properties as fertilized egg-derived ES cells (T. Wakayama et al., 2001, Science, 292: 740-743; S. Wakayama et al., 2005, Biol. Reprod., 72: 932-936; J. Byrne et al., 2007, Nature, 450: 497-502).
  • nuclear transfer ES cells i.e., nt ES cells
  • nt ES cells are ES cells that are established from embryoblasts of blastocysts derived from cloned embryos resulting from substitution of an unfertilized egg nucleus with a somatic cell nucleus.
  • nt ES cells are produced by the technique of nuclear transfer (J. B. Cibelli et al., 1998, Nature Biotechnol., 16: 642-646) in combination with the technique of ES cell production (Kiyoka Wakayama et al., 2008, Experimental Medicine, Vol. 25, No. 5 (extra edition), pp. 47-52).
  • somatic cell nuclei are injected into enucleated unfertilized eggs of mammalian animals, and culture is conducted for several hours. Thus, such cells can be reprogrammed.
  • Muse cells are pluripotent stem cells produced by the method described in WO 2011/007900. More specifically, Muse cells are pluripotent cells that are obtained by treating fibroblasts or myeloid interstitial cells with tryp sin for a long period of time (preferably for 8 hours or 16 hours) and conducting float culture. Such cells are positive for SSEA-3 and CD105.
  • the present invention provides a kit used for inducing pluripotent stem cells to differentiate into alveolar epithelial progenitor cells or producing alveolar epithelial progenitor cells from pluripotent stem cells.
  • the kit may comprise growth factors, compounds, a medium, a cell detachment solution, and an agent for coating the culture vessel used for the induction of differentiation.
  • the kit may further comprise documents and/or instructions describing the procedure for the induction of differentiation.
  • alveolar epithelial progenitor cells may constitute a population of cells including alveolar epithelial progenitor cells.
  • a population of cells including alveolar epithelial progenitor cells preferably includes 50%, 60%, 70%, 80%, or 90% or more alveolar epithelial progenitor cells.
  • the present invention provides a method for extracting alveolar epithelial progenitor cells.
  • the cells to be extracted may be alveolar epithelial progenitor cells obtained by the method described above or cells obtained in the process for producing the same, which takes place after the completion of Step (3) of culture in a medium containing BMP4, retinoic acid, and a GSK3 ⁇ inhibitor or Step (4) of culture in a medium containing FGF10.
  • Alveolar epithelial progenitor cells can be extracted with the use of a reagent having CPM-specific affinity. While CPM had been known as a marker of adult type I alveolar epithelial cells, it was not previously known that CPM was expressed in progenitor cells during development. That is, use of CPM as a surface marker in the same manner as with NKX2-1 as a marker of alveolar epithelial progenitor cells has been found for the first time through the present invention.
  • reagents having specific affinity examples include antibodies, aptamers, peptides, and compounds that specifically recognize the substances of interest, with antibodies or fragments thereof being preferable.
  • antibodies may be polyclonal or monoclonal antibodies. Such antibodies can be prepared in accordance with techniques well known in the art (Current protocols in Molecular Biology, Ausubel et al. (editors), 1987, John Wiley and Sons (publisher), Section 11.12-11.13).
  • the antibodies of the present invention are polyclonal antibodies, specifically, proteins encoded by CPM expressed in E. coli or mammalian cells and purified, oligopeptides having partial amino acid sequences, or glycolipids may be purified in accordance with conventional techniques, nonhuman animals such as rabbits may be immunized therewith, and antibodies of interest can be obtained from sera of the immunized animals in accordance with a conventional technique.
  • antibodies of interest can be obtained from hybridoma cells prepared via fusion of spleen cells obtained from immunized nonhuman animals to myeloma cells (Current protocols in Molecular Biology, Ausubel et al. (editors), 1987, John Wiley and Sons (publisher), Section 11.4-11.11).
  • antibody fragments include a part of an antibody (e.g., an Fab fragment) and a synthetic antibody fragment (e.g., a single-stranded Fv fragment, ScFv).
  • Antibody fragments, such as Fab and F(ab)2 fragments can be prepared by a genetic engineering technique well known in the art. For example, antibodies reacting with CPM can be obtained from Leica Microsystems.
  • reagents having relevant affinity may be bound or conjugated to substances that enable detection, such as a fluorescent label, a radioactive label, a chemoluminescent label, an enzyme, biotin, or streptoavidin, or substances that enable isolation and extraction, such as Protein A, Protein G, beads, or magnetic beads.
  • substances that enable detection such as a fluorescent label, a radioactive label, a chemoluminescent label, an enzyme, biotin, or streptoavidin, or substances that enable isolation and extraction, such as Protein A, Protein G, beads, or magnetic beads.
  • reagents having relevant affinity may be indirectly labeled. Labeling may be carried out in accordance with various techniques known in the art. For example, pre-labeled antibodies (secondary antibodies) that specifically bind to the antibodies described above may be used.
  • Alveolar epithelial progenitor cells can be extracted by, for example, a method comprising conjugating particles to a reagent having relevant affinity in order to precipitate the cells, a method involving the use of magnetic beads to select the cells with the aid of magnetism (e.g., MACS), a method involving the use of a cell sorter with the aid of a fluorescent label, or a method involving the use of a support upon which antibodies or the like are immobilized (e.g., a cell enrichment column).
  • the alveolar epithelial progenitor cells obtained in the present invention can be administered to patients afflicted with diseases that destroy the pulmonary alveolus in the form of pharmaceutical preparations.
  • the alveolar epithelial progenitor cells are prepared into the form of a sheet, and the sheet may be applied to the alveolar epithelium of a patient.
  • the alveolar epithelial progenitor cells may be suspended in physiological saline or the like, and the suspension may then be directly implanted in the pulmonary alveolus of the patient.
  • the present invention provides an agent for treatment of pulmonary alveolar diseases comprising alveolar epithelial progenitor cells obtained from pluripotent stem cells in the manner described above.
  • the number of alveolar epithelial progenitor cells contained in the agent for treatment of pulmonary alveolar diseases is not particularly limited, provided that the transplanted grafts are able to survive.
  • the number of the cells may be adequately adjusted in accordance with lesion size or body size.
  • Human iPS cells (201B7) were provided by Professor Yamanaka at Kyoto University and cultured in accordance with a conventional technique (Takahashi K, et al. Cell, 131: 861-872, 2007).
  • SFTPC-reporter 201B7 was produced by introducing an EGFP sequence into a site downstream of the SFTPC initiation codon of the human iPS cells (201B7).
  • FIG. 1 shows a scheme for producing alveolar epithelial progenitor cells from pluripotent stem cells such as iPS cells.
  • the alveolar epithelial progenitor cells were induced by detaching human iPS cells with the use of Accutase, seeding the cells in a 24-well plate coated with Matrigel at 2.0 ⁇ 10 5 cells/well or in a 6-well plate coated with Matrigel at 9.6 ⁇ 10 5 cells/well, and conducting culture under the conditions described below ( FIGS. 1A and 1B ).
  • the seeded cells (Day 0) were cultured in basal medium 1 (RPMI1640 (Nacalai Tesque) containing 2% B27 (Life Technologies) and a 0.5% penicillin/streptomycin stock solution (Life Technologies)) supplemented with 100 ng/ml Activin A (R&D Systems), 1 ⁇ M CHIR99021 and 10 ⁇ M Y-27632.
  • basal medium 1 RPMI1640 (Nacalai Tesque) containing 2% B27 (Life Technologies) and a 0.5% penicillin/streptomycin stock solution (Life Technologies)
  • 100 ng/ml Activin A R&D Systems
  • the medium was exchanged with basal medium 1 containing 100 ng/ml Activin A, 1 ⁇ M CHIR99021, and 0.25 mM NaB
  • the medium was exchanged with another medium under the same conditions on the following day (Day 2) and 3 days later (Day 4), and culture was conducted for 5 days.
  • the seeded cells (Day 0) were cultured in basal medium 1 supplemented with 100 ng/ml Activin A, 1 ⁇ M CHIR99021, and 10 ⁇ M Y-27632.
  • basal medium 1 supplemented with 100 ng/ml Activin A, 1 ⁇ M CHIR99021, and 10 ⁇ M Y-27632.
  • the medium was exchanged with basal medium 1 containing 100 ng/ml Activin A, 1 ⁇ M CHIR99021, 10 ⁇ M Y-27632, and 0.125 mM or 0.25 mM NaB
  • basal medium 1 containing 100 ng/ml Activin A, 1 ⁇ M CHIR99021, and 0.125 mM or 0.25 mM NaB on the following day (Day 2), and the medium was exchanged with another medium under the same conditions 3 days later (Day 4).
  • the cells obtained in Step 1 were cultured in basal medium 2 (DMEM/F12 medium (Life Technologies) containing 1% Glutamax supplement (Life Technologies), 2% B27 supplement, 1% N2 supplement (Life Technologies), 0.8% StemSureTM 50 mmol/l monothioglycerol solution (Wako), 50 ⁇ L-ascorbic acid (Sigma Aldrich), and 0.5% penicillin/streptomycin stock solution) supplemented with 200 ng/ml or 100 ng/ml hNoggin (R&D Systems) and 10 ⁇ M SB-431542 for 4 days. In this case, the medium was exchanged with another medium under the same conditions every other day.
  • basal medium 2 DMEM/F12 medium (Life Technologies) containing 1% Glutamax supplement (Life Technologies), 2% B27 supplement, 1% N2 supplement (Life Technologies), 0.8% StemSureTM 50 mmol/l monothioglycerol solution (Wako), 50 ⁇ L-ascorbic acid (Sigma
  • the cells obtained in Step 2 were cultured in basal medium 2 containing 100 ng/ml hBMP4 (HumanZyme, Inc.), 0.05 ⁇ M all-trans retinoic acid (ATRA), and 2.5 ⁇ M CHIR99021 for 4 days. In this case, the medium was exchanged with another medium under the same conditions every other day.
  • basal medium 2 containing 100 ng/ml hBMP4 (HumanZyme, Inc.), 0.05 ⁇ M all-trans retinoic acid (ATRA), and 2.5 ⁇ M CHIR99021 for 4 days.
  • the medium was exchanged with another medium under the same conditions every other day.
  • the cells obtained in Step 3 were cultured in basal medium 2 containing 100 ng/ml FGF10 (Wako) for 7 days. In this case, the medium was exchanged with another medium under the same conditions every other day.
  • the cells obtained in Step 4 were cultured in basal medium 3 (Ham's F12 media (Wako) containing 3.33% BSA Fraction V Solution (7.5%) (Life Technologies), 15 mM HEPES (Sigma Aldrich), 0.8 mM CaCl2 (Nacalai Tesque), 1% ITS Premix (BD), and 0.5% penicillin/streptomycin stock solution) containing 50 nM dexamethasone (Sigma Aldrich), 0.1 mM 8-Br-cAMP (Biolog Life Science Institute), 0.1 mM 3-isobutyl-1-methylxanthine (IBMX) (Wako), and 100 ng/ml or 50 ng/ml KGF (Wako). Thereafter, the medium was exchanged with another medium under the same conditions every other day. The alveolar epithelial progenitor cells obtained were analyzed 4 days later (Day 25).
  • basal medium 3 Ham's F12 media (Wako) containing 3.33% B
  • Step 3 the cells were subjected to immunostaining so as to inspect CPM and NKX2-1 expression. As a result, cells positive for both such markers were identified ( FIGS. 2A and 2B ).
  • CPM-positive cells were separated from the cells obtained on Day 14 using MACS (Miltenyi Biotec) ( FIGS. 3A and 3 B), and the obtained cells were allowed to adhere to glass slides via cytospinning, followed by immunostaining. As a result, many thereof were found to also be positive for NKX2-1 ( FIGS. 4A and 4B ). The cells were analyzed using a flow cytometer.
  • CPM-positive cells obtained by MACS were found to be NKX2-1-positive ( FIGS. 5A and 5B ).
  • CPM-positive cells identified via MACS were subjected to quantitative RT-PCR, so as to assay CPM and NKX2-1 mRNA levels.
  • sorting of CPM-positive cells was found to result in a remarkable increase in CPM and NKX2-1 mRNA levels ( FIG. 6 ).
  • Step 4 (Day 21)
  • CPM-positive cells were separated from cells using MACS, and the CPM and NKX2-1 mRNA levels were assayed via quantitative RT-PCR.
  • sorting of CPM-positive cells was found to result in a remarkable increase in CPM and NKX2-1 mRNA levels ( FIG. 7 ).
  • Step 5 CPM-positive cells were separated from cells using MACS, and the obtained cells were allowed to adhere to glass slides via cytospinning, followed by immunostaining. As a result, many thereof were found to also be positive for NKX2-1 ( FIG. 8A ).
  • the cells induced to differentiate in the same manner with the use of SFTPC-reporter 201B7 were subjected to immunostaining. As a result, the presence of SFTPC- or proSPB-positive cells was observed in the CPM-positive cells ( FIG. 8B ).
  • the percentage of the SFTPC-positive cells was found to be about 0.9% as a result of flow cytometric analysis ( FIG. 8C ).
  • Step 5 the cells (201B7) after the completion of Step 5 (Day 25) were subjected to immunostaining, and CPM-, NKX2-1-, SFTPB-, SFTPC-, and CCSP-positive cells were identified ( FIGS. 9A and 9B ).
  • marker genes were found to be also positive for CPM.
  • CPM-positive cells obtained using MACS were subjected to quantitative RT-PCR, so as to assay the CPM, NKX2-1, SFTPA2, SFTPB, DCLAMP, SFTPC, CCSP, and NGFR mRNA levels.
  • sorting of CPM-positive cells was found to result in a remarkable increase in the CPM and NKX2-1 mRNA levels ( FIG. 10 ).
  • alveolar epithelial cells or progenitor cells thereof could be induced from iPS cells by the method of the present invention.
  • CPM was found to be capable of recognizing alveolar epithelial progenitor cells at an early stage, such as during the prenatal period (humans: 3 to 7 weeks in the prenatal period; mice: 9 to 14 days in the prenatal period), in addition to the canalicular period of lung development (humans: 16 to 24 weeks in the prenatal period; mice: 16.5 to 17.5 days in the prenatal period) and the adenoid period (humans: 7 to 16 weeks in the prenatal period; mice: 14.0 to 16.5 days in the prenatal period).
  • alveolar epithelial progenitor cells could be recognized and extracted with the use of CPM as an indicator.
  • the CPM-positive cells (2 ⁇ 10 4 cells) extracted via MACS obtained after the completion of Step 3 with the use of SFTPC-reporter 201B7 were transferred to 12-well Cell Culture Inserts (BD Biosciences) supplemented with 400 ⁇ 1 of medium containing Matrigel in combination with basal medium 3 containing 50 nM dexamethasone, 0.1 mM 8-Br-cAMP, 0.1 mM IBMX, and 10 ng/ml KGF at a ratio of 1:1 together with 2 ⁇ 10 6 human fetus-derived pulmonary fibroblasts (PP002-F-1349, DV Biologics).
  • basal medium 3 containing 10 ⁇ M Y-27632, 50 nM dexamethasone, 0.1 mM 8-Br-cAMP, 0.1 mM IBMX, and 10 ng/ml KGF was added to the lower layer of the Cell Culture Inserts so as to form spheroids (cell masses), and culture was conducted for 10 to 12 days ( FIG. 13 ).
  • the resulting spheroids were inspected using a transmission electron microscope and found to be cells having lamella-like structures ( FIG. 14A ).
  • CPM(+)-cell-derived spheroids were found to be in the form of cystic pseudo-lamellar, cylindrical, or cubic cells having cytoplasm that would be stained dark pink, unlike CPM( ⁇ )-cell-derived spheroids having cytoplasm of pale color ( FIG. 15A ). These cells were double positive for both NKX2-1 and CPM. In addition, these cells included SFTPC-positive cells ( FIG. 15B ). In this case, AQPS-positive cells as markers of type I alveolar epithelial cells were found to be present adjacent to SFTPC-positive cells.
  • the alveolar epithelial cell marker expression was inspected and SFTPA, SFTPB, SFTPC, and SFTPD were found to be expressed in the CPM- and NKX2-1-positive cells ( FIG. 16 ). As a result of quantitative PCR, the expression levels of these genes were found to have been elevated via three-dimensional culture. In addition, expression of SOX9 and ID2, which are indicators for induction of the peripheral airway, was observed in CPM- and NKX2-1-positive cells in several spheroids ( FIG. 16 ).
  • PDPN and CAV1 were expressed in fibroblast-like cells in the vicinity of the spheroids (indicated with arrows), they were also expressed in the spheroids (indicated with arrowheads) ( FIG. 17 ).
  • alveolar epithelial cells were induced from CPM-positive cells, and CPM was found to be a useful marker of progenitor cells of alveolar epithelial cells.
  • the CPM-positive cells obtained were found to be induced into mature alveolar epithelial cells via three-dimensional co-culture thereof with human fetus-derived pulmonary fibroblasts.
  • the method of the present invention enables production of alveolar epithelial progenitor cells from pluripotent stem cells.

Abstract

This invention provides a method for stably producing alveolar epithelial progenitor cells from pluripotent stem cells, including steps of culturing pluripotent stem cells in (1) a medium containing activin A and a GSK3β inhibitor, (2) a medium containing a BMP inhibitor and a TGFβ inhibitor, and (3) a medium containing BMP4, retinoic acid, and a GSK3β inhibitor.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a Continuation of U.S. patent application Ser. No. 14/783,382, filed Oct. 8, 2015, which is a 371 of PCT/JP2014/061106, filed Apr. 14, 2014, which in turn claims the benefit of Japanese Patent Application No. 2013-084034, filed Apr. 12, 2013, the contents of each of which are hereby incorporated by reference.
  • TECHNICAL FIELD
  • The present invention relates to a method for producing alveolar epithelial progenitor cells from pluripotent stem cells. The present invention also relates to a kit used for producing alveolar epithelial progenitor cells from pluripotent stem cells.
  • BACKGROUND ART
  • The lung is one of the most complicated organs, and it is considered to be composed of approximately 40 different types of cells. Among them, the pulmonary alveolus is composed of the alveolar space, which stores gas, and the alveolar epithelium, which surrounds the same. In addition, the alveolar epithelium is composed of the type I alveolar epithelial cells and the type II alveolar epithelial cells. The former forms a blood-air barrier with the microvascular endothelium surrounding the pulmonary alveolus with the aid of the basal membrane and exchanges the intra-alveolar gas with the blood gas. The latter comprises many lamellar corpuscles, it undergoes exocytosis of pulmonary surfactants, and it forms the alveolar lining layer.
  • In recent years, cells having pluripotency, such as embryonic stem cells (ES cells) or induced pluripotent stem cells (iPS cells) obtained by introducing undifferentiated-cell-specific genes into somatic cells, have been reported (U.S. Pat. No. 5,843,780 and WO 2007/069666), methods for inducing alveolar epithelial cells from such cells have been reported (Rippon H. J. et al, Cloning Stem Cells 6: 49-56, 2004; Coraux C. et al, Am. J. Respir. Cell Mol. Biol., 32:87-92, 2005; and Morrisey E. E and Hogan B. L., Dev. Cell., 18: 8-23, 2010), and growth factors and the like that are necessary for the induction of such cells have also been reported. However, there are no examples demonstrating the efficient induction of human pulmonary alveolar cells.
  • Examples of diseases that destroy the pulmonary alveolus include emphysema, interstitial pneumonia, and lymphangioleiomyomatosis. In particular, emphysema is treated via symptomatic treatment or conservative treatment at present, and there is no radical treatment therefor. In addition, there is no radical treatment available for other pulmonary alveolar diseases, and the development of cell-transfer treatment has been accordingly awaited.
  • SUMMARY OF THE INVENTION
  • It is an object of the present invention to provide a method for producing alveolar epithelial progenitor cells from pluripotent stem cells. It is another object of the present invention to provide a kit used for producing alveolar epithelial progenitor cells from pluripotent stem cells.
  • The present inventors have conducted concentrated studies in order to attain the above objects. As a result, they discovered that pluripotent stem cells could be induced to differentiate into alveolar epithelial progenitor cells with the use of various growth factors and compounds. This has led to the completion of the present invention.
  • Specifically, the present invention includes the following.
    • [1] A method for producing alveolar epithelial progenitor cells from pluripotent stem cells comprising Steps (1) to (3) below:
  • (1) culturing pluripotent stem cells in a medium containing activin A and a GSK3β inhibitor;
  • (2) culturing the cells obtained in Step (1) in a medium containing a bone morphogenic protein (BMP) inhibitor and a TGFβ inhibitor; and
  • (3) culturing the cells obtained in Step (2) in a medium containing BMP4, retinoic acid, and a GSK3β inhibitor.
    • [2] The method according to [1], which further comprises a step of extracting CPM-positive cells as alveolar epithelial progenitor cells, following Step (3).
    • [3] The method according to [1] or [2], wherein Step (1) further comprises culturing pluripotent stem cells with the addition of an ROCK inhibitor and/or HDAC inhibitor to a medium.
    • [4] The method according to any one of [1] to [3], wherein Step (1) comprises culturing for 6 days or longer.
    • [5] The method according to any one of [1] to [4], wherein Step (2) comprises culturing for 4 days or longer.
    • [6] The method according to any one of [1] to [5], wherein Step (3) comprises culturing for 4 days or longer.
    • [7] The method according to any one of [1] to [6], wherein the GSK3β inhibitor is CHIR99021, the BMP inhibitor is Noggin, and the TGFβ inhibitor is SB431542.
    • [8] The method according to any one of [3] to [7], wherein the ROCK inhibitor is Y-27632 and/or the HDAC inhibitor is sodium butyrate.
    • [9] The method according to any one of [1] to [8], which further comprises Steps (4) and (5), following Step (3) below:
  • (4) culturing the cells obtained in Step (3) in a medium containing FGF10; and
  • (5) culturing the cells obtained in Step (4) in a medium containing a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF.
    • [10] The method according to [9], which further comprises a step of extracting CPM-positive cells as alveolar epithelial progenitor cells, following Step (5).
    • [11] The method according to [9] or [10], wherein Step (4) comprises culturing for 7 days or longer.
    • [12] The method according to any one of [9] to [11], wherein Step (5) comprises culturing for 4 days or longer.
    • [13] The method according to any one of [9] to [12], wherein the steroid drug is dexamethasone, the cAMP derivative is 8Br-cAMP, and the phosphodiesterase inhibitor is 3-isobutyl-1-methylxanthine (IBMX).
    • [14] The method according to any one of [1] to [13], wherein the alveolar epithelial progenitor cells are human alveolar epithelial progenitor cells.
    • [15] A method for producing alveolar epithelial progenitor cells, which further comprises a step of three-dimensional culture of the alveolar epithelial progenitor cells produced by the method according to [1] or [2].
    • [16] Alveolar epithelial progenitor cells produced by the method according to any one of [1] to [15].
    • [17] A kit used for producing alveolar epithelial progenitor cells from pluripotent stem cells, which comprises activin A, a GSK3β inhibitor, a BMP inhibitor, a TGFβ inhibitor, BMP4, retinoic acid, a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, FGF10, and KGF.
    • [18] The kit according to [17], which further comprises an ROCK inhibitor and/or HDAC inhibitor.
    • [19] The kit according to [17] or [18], wherein the GSK3β inhibitor is CHIR99021, the BMP inhibitor is Noggin, the TGFβ inhibitor is SB431542, the steroid drug is dexamethasone, the cAMP derivative is 8Br-cAMP, and the phosphodiesterase inhibitor is IBMX.
    • [20] The kit according to [18] or [19], wherein the ROCK inhibitor is Y-27632 and/or the HDAC inhibitor is sodium butyrate.
    • [21] A method for extracting alveolar epithelial progenitor cells, which comprises a step of extracting CPM-positive cells as alveolar epithelial progenitor cells from a cell population including alveolar epithelial progenitor cells.
  • This description includes part or all of the content as disclosed in the description and/or drawings of Japanese Patent Application No. 2013-084034, which is a priority document of the present application.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A and FIG. 1B each show a scheme for producing alveolar epithelial progenitor cells from pluripotent stem cells. In the figures, “Stage I” is synonymous with “Step 1,” “Stage II” is synonymous with “Step 2,” “Stage III” is synonymous with “Step 3,” “Stage IV” is synonymous with “Step 4,” and “Stage V” is synonymous with “Step 5.” Hereafter, such “stages” and “steps” are collectively referred to as “Steps.”
  • FIG. 2A shows images of immunostained CPM and NKX2-1 in the cells after the completion of Step 3. FIG. 2B shows images of immunostained CPM and NKX2-1 in the cells after the completion of Step 2 (upper images) and Step 3 (lower images).
  • FIG. 3A shows percentages of CPM-positive cells after sorting via CPM-based MACS, after the completion of Step 3. In the figure, “Isotype control” shows the results for the negative control, “Pre-sorting” shows the results for the cells before sorting via MACS, “CPM positive selection” shows the results for CPM-positive cells after sorting via MACS, and “CPM negative selection” shows the results for CPM-negative cells after sorting via MACS. FIG. 3B shows percentages of CPM-positive cells after sorting via CPM-based MACS after the completion of Step 3. “Pre-sorting” shows the results for the cells before sorting via MACS, “CPM(+) sorting” shows the results for the MACS-sorted CPM-positive cells, and “CPM(−) sorting” shows the results for the MACS-sorted CPM-negative cells.
  • FIG. 4A shows images of immunostained CPM and NKX2-1 in the MACS-sorted CPM-positive cells (CPM(+) sorting) and in the CPM-negative cells (CPM(−) sorting) after the completion of Step 3. FIG. 4B shows images of immunostained NKX2-1 in the MACS-sorted CPM-positive cells (CPM(+) sorting) and in the CPM-negative cells (CPM(−) sorting) after the completion of Step 3.
  • FIG. 5A shows percentages of NKX2-1-positive cells among the MACS-sorted cells after the completion of Step 3. In the figure, “Pre-sorting” shows the results for the cells before sorting via MACS, “CPM positive selection” shows the results for the MACS-sorted CPM-positive cells, and “CPM negative selection” shows the results for the MACS-sorted CPM-negative cells. FIG. 5B shows percentages of NKX2-1-positive cells among the MACS-sorted cells after the completion of Step 3. In the figure, “CPM+ sorted cells” shows percentages of NKX2-1-positive cells among the MACS-sorted CPM-positive cells (92.3±0.7%), and “CPM sorted cells” shows percentages of NKX2-1-positive cells among the MACS-sorted CPM-negative cells (22.2±2.3%).
  • FIG. 6 shows the assay results of CPM mRNA levels (left) and NKX2-1 mRNA levels (right) in the MACS-sorted cells on the basis of CPM markers via quantitative PCR after the completion of Step 3. “Step 3 ” shows the results for the cells before sorting, “Step 3 (+)” shows the results for the CPM-positive cells, “Step 3 (−)” shows the results for the CPM-negative cells, “Fetal Lung” shows the results for the fetal pneumocytes, and “Adult Lung” shows the results for the adult pneumocytes.
  • FIG. 7 shows the assay results of CPM mRNA levels (left) and NKX2-1 mRNA levels (right) in the MACS-sorted cells via quantitative PCR after the completion of Step 4. “Step 4” shows the results for the cells before sorting, “Step 4 (+)” shows the results for the CPM-positive cells, “Step 4 (−)” shows the results for the CPM-negative cells, “Fetal Lung” shows the results for the fetal pneumocytes, and “Adult Lung” shows the results for the adult pneumocytes.
  • FIG. 8A shows images of immunostained iPS cells (201B7) in the MACS-sorted CPM-positive cells after the completion of Step 5. FIG. 8B shows images of immunostained iPS cells (SFTPC reporter cells; SFTPC-reporter 201B7) in the MACS-sorted CPM-positive cells after the completion of Step 5. FIG. 8C shows percentages of EGFP-positive iPS cells (SFTPC reporter cells; SFTPC-reporter 201B7) among the MACS-sorted CPM-positive cells after the completion of Step 5.
  • FIG. 9A and FIG. 9B each show an image of immunostained cells after the completion of Step 5.
  • FIG. 10 shows the assay results of mRNA levels via quantitative PCR of CPM, NKX2-1, SFTPA2, SFTPB, SFTPC, DCLAMP, CCSP (SCGB1A1), SCGB3A2, and NGFR in the MACS-sorted cells after the completion of Step 5. “Step 5” shows the results for the cells before sorting, “Step 5 (+)” shows the results for the CPM-positive cells, “Step 5 (−)” shows the results for the CPM-negative cells, “Fetal Lung” shows the results for the fetal pneumocytes, and “Adult Lung” shows the results for the adult pneumocytes.
  • FIG. 11 shows images of CPM in the human fetal lung tissue subjected to staining together with NKX2-1, SFTPC, or T1α serving as a marker of the prenatal period, the adenoid period, or the canalicular period of the lung, respectively.
  • FIG. 12 shows images of CPM in the mouse lung tissue at relevant prenatal periods (E12.5, E15.5, and E17.5) subjected to staining together with NKX2-1 serving as a marker of the prenatal period, the adenoid period, or the canalicular period of the lung, respectively.
  • FIG. 13 shows a summary of a method of sorting CPM-positive cells from among the cells after the completion of Step 3 and culturing the sorted cells together with human fetal pulmonary fibroblasts.
  • FIG. 14A shows transmission electron microscopic images of spheroids following the three-dimensional culture of the CPM-positive cells after the completion of Step 3. In FIG. 14A, the images at the center and on the right are each an enlarged view of a part of the image on the left. FIG. 14B shows transmission electron microscopic images of type II alveolar epithelial cells of mouse lungs. The image on the right shows an enlarged view of a part of the image on the left. FIG. 14C shows transmission electron microscopic images of the mouse fetal lung at E17.5. The image on the right shows an enlarged view of a part of the image on the left. In the figures, “Lu” indicates a lumen.
  • FIG. 15A shows hematoxylin-eosin-stained images of the three-dimensionally cultured CPM-positive cells (left) and CPM-negative cells (right) after the completion of Step 3. The lower images show enlarged views of the upper images. FIG. 15B shows images of the spheroids immunostained with the CPM, NKX2-1, GFP (SFTPC), SFTPC (endogenous), and AQP5 antibodies following the three-dimensional culture of the CPM-positive cells after the completion of Step 3.
  • FIG. 16 shows images of the spheroids immunostained with the CPM, NKX2-1, SFTPC, SFTPB, SFTPA, SFTPD, ID2, and SOX9 antibodies following the three-dimensional culture of the CPM-positive cells after the completion of Step 3. The images on the right show images of alveolar epithelial cell-associated proteins subjected to dual staining with either CPM or NKX2-1.
  • FIG. 17 shows images of the spheroids immunostained with the T1α, SFTPC, AQP5, and CAV1 antibodies following the three-dimensional culture of the CPM-positive cells after the completion of Step 3.
  • EMBODIMENTS FOR CARRYING OUT THE INVENTION
  • Hereafter, the present invention is described in detail.
  • The present invention provides a method for producing alveolar epithelial progenitor cells (e.g., human alveolar epithelial progenitor cells) from pluripotent stem cells comprising steps of culturing pluripotent stem cells in (1) a medium containing activin A and a GSK3β inhibitor, (2) a medium containing a BMP inhibitor and a TGFβ inhibitor, and (3) a medium containing BMP4, retinoic acid, and a GSK3β inhibitor.
  • The method for producing alveolar epithelial progenitor cells according to the present invention may comprise a step of extracting CPM-positive cells as the alveolar epithelial progenitor cells, following Step (3).
  • The method for producing alveolar epithelial progenitor cells according to the present invention may further comprise Step (4) of culture in a medium containing FGF10 and Step (5) of culture in a medium containing a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF, following Step (3). In addition, the method may further comprise a step of extracting CPM-positive cells as the alveolar epithelial progenitor cells, following Step (5).
  • In the present invention, the term “alveolar epithelial progenitor cells” refers to progenitor cells of type I alveolar epithelial cells or type II alveolar epithelial cells, which express CPM or NKX2-1. In this description, the term “alveolar epithelial cells” is not distinguished from the term “alveolar epithelial progenitor cells,” unless otherwise specified. In the present invention, “CPM” indicates a polynucleotide shown in the NCBI Accession Number NM_001005502, NM_001874, or NM_198320 or a protein encoded thereby. In the present invention, “NKX2-1 ” indicates a polynucleotide shown in the NCBI Accession Number NM_001079668 or NM_003317 or a protein encoded thereby.
  • In the present invention, examples of markers for alveolar epithelial progenitor cells include polynucleotides selected from the group consisting of SFTPB (NCBI Accession Numbers NM_000542 and NM_198843), SFTPC (NCBI Accession Numbers NM_001172357, NM_001172410, and NM_003018), and CCSP (NCBI Accession Number NM_003357) and proteins encoded by such polynucleotides.
  • [Step of Culture in a Medium Containing Activin A and a GSK3β Onhibitor]
  • A medium used in the step of culturing pluripotent stem cells according to the present invention can be prepared from a medium used for animal cell culture as a basal medium. Examples of basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), αMEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media. A medium may or may not contain blood serum. A medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol. In addition, a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts. RPMI 1640 medium supplemented with B27 and antibiotics is preferable.
  • In this step, pluripotent stem cells are cultured in a medium prepared by supplementing the basal medium described above with activin A and a GSK3β inhibitor. In this step, an HDAC inhibitor may further be added.
  • Activin A is a homodimer with two beta A chains, the amino acid sequence of activin A is 100% homologous to that of a protein of a human, mouse, rat, pig, cow, or cat, and, accordingly, the relevant species are not particularly limited. In the present invention, activin A is preferably of an active form with the N-terminal peptide being cleaved, and it is preferably a homodimer comprising, bound thereto via a disulfide bond, the Gly311-Ser426 fragment with the N-terminal peptide of the inhibin beta A chain (e.g., NCBI Accession Number NP_002183) being cleaved. Such activin A is commercially available from, for example, Wako and R&D Systems.
  • The activin A concentration in the medium is, for example, 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 150 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 mg/ml, although the concentration is not limited thereto. The concentration is preferably 100 ng/ml.
  • The term “GSK362 inhibitor” used herein is defined as a substance that inhibits kinase activity of the GSK-3β protein (e.g., the capacity for phosphorylation of (β-catenin), and many such substances are already known. Examples thereof include: an indirubin derivative, such as BIO, which is also known as a GSK-3β inhibitor IX (6-bromoindirubin-3′-oxime); a maleimide derivative, such as SB216763 (3-(2,4-dichlorophenyl)-4-(1-methyl-1H-indo1-3-yl)-1H-pyrrole-2,5-dione); a phenyl α-bromomethylketone compound, such as a GSK-3β inhibitor VII (4-dibromoacetophenone); a cell-membrane-permeable phosphorylated peptide, such as L803-mts, which is also known as a GSK-3β peptide inhibitor (i.e., Myr-N-GKEAPPAPPQSpP-NH2); and CHIR99021, such as 6-[2-[4-(2,4-dichlorophenyl)-5-(4-methyl-1H-imidazol-2-yl)pyrimidin-2-ylamino]ethylami no]pyridine-3-carbonitrile, with high selectivity. While such compounds are commercially available from, for example, Calbiochem or Biomol, and easily used, such compounds may be obtained from other companies, or persons may prepare such compounds by themselves.
  • A GSK-3β inhibitor that can be preferably used in the present invention is CHIR99021. In this step, the concentration of CHIR99021 in a medium is, for example, 1 nM, 10 nM, 50 nM, 100 nM, 500 nM, 750 nM, 1 μM, 1.5 μM, 2 μM, 2.5 μM, 3 μM, 3.5 μM, 4 μM, 4.5 μM, 5 μM, 6 μM, 7 μM, 8 μM, 9 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM, 40 μM, or 50 μM, although the concentration is not limited thereto. In this step, the concentration is preferably 1 μM.
  • The term “HDAC inhibitor” is defined as a substance that inhibits or inactivates enzyme activity of histone deacetylase (HDAC). Examples thereof include low-molecular-weight inhibitors, such as valproic acid (VPA) (Nat. Biotechnol., 26 (7): 795-797, 2008), trichostatin A, sodium butyrate (NaB), MC 1293, and M344; nucleic acid-based expression inhibitors such as siRNAs and shRNAs against HDAC (e.g., HDAC1 siRNA Smartpool □ (Millipore) and HuSH 29mer shRNA Constructs against HDAC1 (OriGene)); and DNA methyltransferase inhibitors (e.g., 5′-azacytidine) (Nat. Biotechnol., 26 (7): 795-797, 2008).
  • An HDAC inhibitor that can be preferably used in the present invention is sodium butyrate (NaB). The concentration of sodium butyrate (NaB) in a medium is, for example, 1 μM, 10 μM, 50 μM, 100 μM, 250 μM, 500 μM, 750 μM, 1 mM, 2 mM, 3 mM, 4 mM, or 5 mM, although the concentration is not limited thereto. The concentration is preferably 250 μM.
  • In this step, culture may be conducted in a culture vessel treated with a coating agent. A coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may comprise a process of pluripotent stem cell detachment. Examples of methods for cell detachment include a method of mechanical detachment and a method of cell detachment involving the use of a cell detachment solution having protease activity and collagenase activity (e.g., Accutase™ and Accumax™ or a cell detachment solution having collagenase activity alone. It is preferable that human pluripotent stem cells be detached with the use of a cell detachment solution having protease activity and collagenase activity (and the use of Accutase™ is particularly preferable).
  • When the step comprises a process of cell detachment, an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • An ROCK inhibitor is not particularly limited, provided that it can inhibit functions of Rho kinase (ROCK). Examples thereof include: Y-27632 ((+)-(R)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide dihydrochloride) (e.g., Ishizaki et al., Mol. Pharmacol., 57, 976-983, 2000; Narumiya et al., Methods Enzymol., 325, 273-284, 2000); Fasudil/HA1077 (e.g., Uenata et al., Nature 389: 990-994, 1997); H-1152 (e.g., Sasaki et al., Pharmacol. Ther., 93: 225-232, 2002); Wf-536 (e.g., Nakajima et al., Cancer Chemother. Pharmacol., 52 (4): 319-324, 2003) and derivatives thereof; antisense nucleic acids against ROCK; RNA interference-inducible nucleic acids (e.g., siRNA); dominant-negative variants; and expression vectors thereof. Since other low-molecular-weight compounds are known as ROCK inhibitors, such compounds and derivatives thereof can also be used in the present invention (e.g., U.S. Patent Application Publication Nos. 2005/0209261, 2005/0192304, 2004/0014755, 2004/0002508, 2004/0002507, 2003/0125344, and 2003/0087919, WO 2003/062227, WO 2003/059913, WO 2003/062225, WO 2002/076976, and WO 2004/039796). In the present invention, one or more types of ROCK inhibitors can be used.
  • An ROCK inhibitor that can be preferably used in the present invention is Y-27632. The Y-27632 concentration is, for example, 100 nM, 500 nM, 750 nM, 1 μM, 2 μM, 3 μM, 4 μM, 5 μM, 6 μM, 7 μM, 8 μM, 9 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM, 40 μM, or 50 μM, although the concentration is not limited thereto. The concentration is preferably 10 μM.
  • Concerning culture conditions, culture is conducted at about 30□C to 40□C, and preferably at about 37□0C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO2, and the CO2 concentration is preferably about 2% to 5%.
  • The culture period is not particularly limited because long-term culture would not cause any problems. For example, the culture period may be at least 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, or 12 days. The culture period is preferably at least 6 days, and it is particularly preferably 6 days. When the ROCK inhibitor is added, the duration of addition is 1 day or 2 days, with 1 day being preferable. When the HDAC inhibitor is further added, the addition is initiated on the day following the initiation of the step, and culture is conducted for at least 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, or 11 days. Culture is preferably conducted for at least 5 days, and particularly preferably for 5 days, in the presence of the HDAC inhibitor.
  • [Step of Culture in a Medium Containing BMP Inhibitor and TGFβ Inhibitor]
  • A medium used in this step can be prepared from a medium used for animal cell culture as a basal medium. Examples of basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), αMEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media. A medium may or may not contain blood serum. A medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol. In addition, a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts. A medium mixture of DMEM and Ham's F12 supplemented with Glutamax, B27, N2, 3′-thiol glycerol, and ascorbic acid is preferable.
  • In this step, the cells obtained in the previous step (i.e., the step of culture of pluripotent stem cells in a medium containing activin A and a GSK3β inhibitor) are cultured in a medium prepared by supplementing the basal medium with a BMP inhibitor and a TGFβ inhibitor.
  • Examples of BMP inhibitors include: protein-based inhibitors, such as Chordin, Noggin, and Follistatin; dorsomorphin (i.e., 6-[4-(2-piperidin-1-yl-ethoxy)phenyl]-3-pyridin-4-yl-pyrazolo[1,5-a]pyrimidine) and a derivative thereof (P. B. Yu et al., 2007, Circulation, 116: II_60; P. B. Yu et al., 2008, Nat. Chem. Biol., 4:33-41; J. Hao et al., 2008, PLoS ONE, 3 (8): e2904); and LDN-193189 (i.e., 4-(6-(4-(piperazin-1-yl)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)quinoline). Dorsomorphin and LDN-193189 are commercially available from Sigma-Aldrich and Stemgent, respectively.
  • A BMP inhibitor that can be preferably used in the present invention is Noggin. The concentration of Noggin in a medium is not particularly limited, provided that BMP can be inhibited. For example, such concentration is 1 ng/ml, 10 ng/ml, 50 ng/ml, 100 ng/ml, 200 ng/ml, 30 ng/ml, 400 ng/ml, 50 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, 1 μg/ml, or 2 μg/ml, although the concentration is not limited thereto. The concentration is preferably 200 ng/ml.
  • The term “TGFβ inhibitor” used herein refers to a substance that inhibits signal transmission from the binding of TGFβ to a receptor leading to SMAD. A TGFβ inhibitor is not particularly limited, provided that such substance inhibits TGFβ from binding to a receptor; i.e., the ALK family, or such substance inhibits phosphorylation of SMAD caused by the ALK family. Examples thereof include Lefty-1 (e.g., NCBI Accession Nos. mouse NM_010094 and human NM_020997), SB431542 (4-(4-(benzo[d][1,3]dioxo1-5-yl)-5-(pyridin-2-yl)-1H-imidazol-2-yl)benzamide), SB202190 (R. K. Lindemann, et al., Mol. Cancer, 2003, 2: 20), SB505124 (GlaxoSmithKline), NPC30345, SD093, SD908, SD208 (Scios), LY2109761, LY364947, LY580276 (Lilly Research Laboratories), A-83-01 (WO 2009/146408), and derivatives thereof.
  • A TGFβ inhibitor that can be preferably used in the present invention is SB431542. The SB431542 concentration in a medium is not particularly limited, provided that TGFβ is inhibited. For example, such concentration can be 1 μM, 2 μM, 3 μM, 4 μM, 5 μM, 6 μM, 7 μM, 8 μM, 9 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM, 35 μM, 40 μM, 45 μM, 50 μM, 60 μM, 70 μM, 80 μM, 90 μM, 100 μM, 200 μM, 300 μM, 400 μM, or 500 μM, although the concentration is not limited thereto. The concentration is preferably 10 μM.
  • In this step, culture may be conducted in a culture vessel treated with a coating agent. Examples of coating agents include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above. Alternatively, cells may be detached and reseeded in a culture vessel. When cells are to be detached, particular cells may be selected, and, for example, SOX17- and/or FOXA2-positive cells may be selected and used in this step. This method is preferably implemented by means of media exchange.
  • When the step comprises a process of cell detachment, an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • Concerning culture conditions, culture is conducted at about 30□C to 40□C, and preferably at about 37□C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO2, and the CO2 concentration is preferably about 2% to 5%.
  • The culture period is not particularly limited because long-term culture would not cause any problems. For example, the culture period may be at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 8 days. The culture period is preferably 4 days.
  • [Step of Culture in a Medium Containing BMP4, Retinoic Acid, and GSK3β Inhibitor]
  • A medium used in this step can be prepared from a medium used for animal cell culture as a basal medium. Examples of basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), αMEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media. A medium may or may not contain blood serum. A medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol. In addition, a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts. A medium mixture of DMEM and Ham's F12 supplemented with Glutamax, B27, N2, 3′-thiol glycerol, and ascorbic acid is preferable.
  • In this step, the cells obtained in the previous step (i.e., the step of culture in a medium containing a BMP inhibitor and a TGFβ inhibitor) are cultured in a medium prepared by supplementing the basal medium with BMP4, retinoic acid, and a GSK3β inhibitor.
  • The term “BMP4” used herein refers to a protein encoded by the polynucleotide shown in the NCBI Accession Number NM_001202, NM_130850, or NM_130851, and it may be in an active form resulting from cleavage by a protease.
  • The BMP4 concentration in a medium is not particularly limited. For example, such concentration may be 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 μg/ml, although the concentration is not limited thereto. The concentration is preferably 100 ng/ml.
  • While all-trans retinoic acid (ATRA) is exemplified as retinoic acid, artificially modified retinoic acid that retains functions of naturally occurring retinoic acid may be used. Examples thereof include 4-[[(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)carbonyl]amino]-benzoic acid (AM580) (Tamura, K. et al., Cell Differ. Dev., 32: 17-26, 1990), 4-[(1E)-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)-1-propen-1-yl]-benzoic acid (TTNPB) (Strickland, S., et al., Cancer Res., 43: 5268-5272, 1983), retinol palmitate, retinol, retinal, 3-dehydroretinoic acid, 3-dehydroretinol, 3-dehydroretinal, and compounds described in Abe, E., et al., Proc. Natl. Acad. Sci., (U.S.A.) 78: 4990-4994, 1981; Schwartz, E. L.et al., Proc. Am. Assoc. Cancer Res., 24: 18, 1983; Tanenaga, K. et al., Cancer Res., 40: 914-919, 1980.
  • The retinoic acid concentration in a medium is not particularly limited. For example, such concentration can be 1 nM, 5 nM, 10 nM, 15 nM, 20 nM, 25 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, or 1 μM, although the concentration is not limited thereto. The concentration is preferably 50 nM.
  • The GSK3β inhibitor as described above can be used in this step, and the GSK3β inhibitor is preferably CHIR99021. In this step, the CHIR99021 concentration in a medium is, for example, 1 nM, 10 nM, 50 nM, 100 nM, 500 nM, 750 nM, 1 μM, 1.5 μM, 2 μM, 2.5 μM, 3 μM, 3.5 μM, 4 μM, 4.5 μM, 5 μM, 6 μM, 7 μM, 8 μM, 9 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM, 40 μM, or 50 μM, although the concentration is not limited thereto. In this step, the concentration is preferably 2.5 μM.
  • In this step, culture may be conducted in a culture vessel treated with a coating agent. A coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above. Alternatively, cells may be detached and reseeded in a culture vessel. When cells are to be detached, particular cells may be selected, and, for example, SOX2-, SOX17-, and/or FOXA2-positive cells may be selected and used in this step. This method is preferably implemented by means of media exchange.
  • When the step comprises a process of cell detachment, an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • Concerning culture conditions, culture is conducted at about 30□C to 40□C, and preferably at about 37□C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO2, and the CO2 concentration is preferably about 2% to 5%.
  • The culture period is not particularly limited because long-term culture would not cause any problems. For example, the culture period may be at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 8 days. The culture period is preferably at least 4 days, and more preferably 4 days.
    • [Step of Culture in a Medium Containing FGF10]
  • A medium used in this step can be prepared from a medium used for animal cell culture as a basal medium. Examples of basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), αMEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media. A medium may or may not contain blood serum. A medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol. In addition, a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts. A medium mixture of DMEM and Ham's F12 supplemented with Glutamax, B27, N2, 3′-thiol glycerol, and ascorbic acid is preferable.
  • In this step, the cells obtained in the previous step (i.e., the step of culture in a medium containing BMP4, retinoic acid, and a GSK3β inhibitor) are cultured in a medium prepared by supplementing the basal medium with FGF10.
  • The term “FGF10 ” used herein refers to a protein encoded by the polynucleotide shown in the NCBI Accession Number NM_004465, and it may be in an active form resulting from cleavage by a protease. Such FGF10 is commercially available from, for example, Life Technologies or Wako.
  • The FGF10 concentration in a medium is not particularly limited. For example, such concentration may be 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 μg/ml, although the concentration is not limited thereto. The concentration is preferably 100 ng/ml.
  • In this step, culture may be conducted in a culture vessel treated with a coating agent. A coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above. Alternatively, cells may be detached and reseeded in a culture vessel. When cells are to be detached, particular cells may be selected, and, for example, NKX2-1- and/or FOXA2-positive cells may be selected and used in this step. This method is preferably implemented by means of media exchange.
  • When the step comprises a process of cell detachment, an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • Concerning culture conditions, culture is conducted at about 30□C to 40□C, and preferably at about 37□C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO2, and the CO2 concentration is preferably about 2% to 5%.
  • The culture period is not particularly limited because long-term culture would not cause any problems. For example, the culture period may be at least 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days. The culture period is preferably at least 7 days, and more preferably 7 days.
    • [Step of Culture in a Medium Containing a Steroid Drug, a cAMP Derivative, a Phosphodiesterase Inhibitor, and KGF]
  • A medium used in this step can be prepared from a medium used for animal cell culture as a basal medium. Examples of basal media include IMDM medium, Medium 199, Eagle's Minimum Essential Medium (EMEM), αMEM medium, Dulbecco's modified Eagle's Medium (DMEM), Ham's F12 medium, RPMI 1640 medium, Fischer's medium, Neurobasal Medium (Life Technologies), and a mixture of any such media. A medium may or may not contain blood serum. A medium may optionally contain one or more serum substitutes selected from among, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (an FBS serum substitute used for ES cell culture), N2 supplements (Invitrogen), B27 supplements (Invitrogen), fatty acid, insulin, ITS Premix, collagen precursors, trace elements, 2-mercaptoethanol, and 3′-thiol glycerol. In addition, a medium can contain one or more substances selected from among, for example, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), nonessential amino acids, vitamins, growth factors, low-molecular-weight compounds, antibiotics, antioxidants, pyruvic acids, buffer agents, and inorganic salts. Ham's F12 medium containing albumin, buffer agents (e.g., HEPES), calcium chloride, ITS Premix, and antibiotics is preferable.
  • In this step, the cells obtained in the previous step (i.e., the step of culture in a medium containing FGF10) are cultured in a medium prepared by supplementing the basal medium with a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF.
  • The term “steroid drug” used herein refers to a steroidal anti-inflammatory drug, such as glucocorticoid or a synthetic derivative thereof. Specific examples thereof include hydrocortisone, hydrocortisone succinate, prednisolone, methylprednisolone, methylprednisolone succinate, triamcinolone, triamcinolone acetonide, dexamethasone, and betamethasone.
  • A steroid drug that can be preferably used in the present invention is dexamethasone. The dexamethasone concentration in a medium is not particularly limited. For example, such concentration may be 1 nM, 5 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, or 1 μM, although the concentration is not limited thereto. The concentration is preferably 50 nM.
  • The term “cAMP derivative” used herein refers to a compound with a modified cyclic AMP substituent. Examples thereof include cyclic adenosine monophosphate (cAMP), 8-bromo cyclic adenosine monophosphate (8-Br-cAMP or 8Br-cAMP), 8-chloro-cyclic adenosine monophosphate (8-Cl-cAMP), 8-(4-chlorophenylthio)cyclic adenosine monophosphate (8-CPT-cAMP), and dibutyryl cyclic adenosine monophosphate (DB-cAMP).
  • A cAMP derivative that can be preferably used in the present invention is 8-Br-cAMP. The concentration of 8-Br-cAMP in a medium is not particularly limited. For example, such concentration can be 1 μM, 5 μM, 10 μM, 20 μM, 30 μM, 40 μM, 50 μM, 60 μM, 70 μM, 80 μM, 90 μM, 100 μM, 200 μM, 300 μM, 400 μM, 500 μM, 600 μM, 700 μM, 800 μM, 900 μM, or 1 mM, although the concentration is not limited thereto. The concentration is preferably 100 μM.
  • The term “phosphodiesterase inhibitor” used herein refers to a compound that inhibits phosphodiesterase (PDE), so as to increase the concentration of cAMP or cGMP in the cells. Examples thereof include 1,3-dimethylxanthine, 6,7-dimethoxy-1-(3,4-dimethoxybenzyl)isoquinoline, 4-{[3′,4′-(methylenedioxy)benzyl]amino}-6-methoxyquinazoline, 8-methoxymethyl-3-isobutyl-1-methylxanthine, and 3-isobutyl-1-methylxanthine (IBMX).
  • A phosphodiesterase inhibitor that can be preferably used in the present invention is IBMX. The IBMX concentration in a medium is not particularly limited. For example, such concentration can be 1 μM, 5 μM, 10 μM, 20 μM, 30 μM, 40 μM, 50 μM, 60 μM, 70 μM, 80 μM, 90 μM, 100 μM, 200 μM, 300 μM, 400 μM, 500 μM, 600 μM, 700 μM, 800 μM, 900 μM, or 1 mM, although the concentration is not limited thereto. The concentration is preferably 100 μM.
  • The term “KGF” used herein refers to a protein encoded by the polynucleotide shown in the NCBI Accession Number NM_002009, and it may be in an active form resulting from cleavage by a protease. Such KGF is commercially available from, for example, Wako.
  • The concentration of KGF in a medium is not particularly limited. For example, such concentration can be 10 ng/ml, 20 ng/ml, 30 ng/ml, 40 ng/ml, 50 ng/ml, 60 ng/ml, 70 ng/ml, 80 ng/ml, 90 ng/ml, 100 ng/ml, 200 ng/ml, 300 ng/ml, 400 ng/ml, 500 ng/ml, 600 ng/ml, 700 ng/ml, 800 ng/ml, 900 ng/ml, or 1 μg/ml, although the concentration is not limited thereto. The concentration is preferably 100 ng/ml.
  • In this step, culture may be conducted in a culture vessel treated with a coating agent. A coating agent may be a naturally occurring or artificially synthesized extracellular matrix. Examples thereof include BD Matrigel, collagen, gelatin, laminin, heparan sulfate proteoglycan, entactin, and a combination of any thereof, with Matrigel being preferable.
  • This step may be implemented by exchanging the cell culture medium obtained in the previous step with the medium described above. Alternatively, cells may be detached and reseeded in a culture vessel. When cells are to be detached, particular cells may be selected, and, for example, NKX2-1-positive cells may be selected and used in this step. This method is preferably implemented by means of media exchange.
  • When the step comprises a process of cell detachment, an ROCK inhibitor may be added to a medium, so as to inhibit pluripotent stem cell death caused by detachment.
  • Concerning culture conditions, culture is conducted at about 30□C to 40□C, and preferably at about 37□C, although the temperature is not limited thereto. Culture is conducted under an atmosphere of air containing CO2, and the CO2 concentration is preferably about 2% to 5%.
  • The culture period is not particularly limited because long-term culture would not cause any problems. For example, the culture period may be at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 8 days. The culture period is preferably at least 4 days, and it is more preferably 4 days.
  • [Three-Dimensional Culture]
  • The present invention provides a method for three-dimensional culture of alveolar epithelial progenitor cells for further maturation of alveolar epithelial progenitor cells. According to the present invention, three-dimensional culture is carried out by subjecting cells to float culture in the form of cell masses (i.e., spheroids). According to the present invention, three-dimensional culture can be carried out with the use of, for example, Cell Culture Inserts provided by BD.
  • According to the present invention, three-dimensional culture may be conducted in the presence of other cell species. Examples of other cell species that may be used include human pulmonary fibroblasts and human fetal pulmonary fibroblasts. Such cells are commercially available from, for example, American Type Culture Collection (ATCC) and DV Biologics.
  • The medium used for three-dimensional culture according to the present invention may be a medium that is used in the step of culture conducted in a medium containing a steroid drug, a cAMP derivative, a phosphodiesterase inhibitor, and KGF. Use of a medium supplemented with an extracellular matrix may be preferable. The ratio of the volume of the extracellular matrix to the volume of the medium is not particularly limited. For example, these substances can be mixed at a ratio of 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, or 1:5. In the present invention, an extracellular matrix is a supramolecular structure that exists outside the cell, and it may be a naturally occurring or artificial (recombinant) structure. Examples thereof include substances such as collagen, proteoglycan, fibronectin, hyaluronic acid, tenascin, entactin, elastin, fibrillin, and laminin, and fragments thereof. These extracellular matrices may be used in combination. For example, extracellular matrices may be prepared from cells such as BD Matrigel™. An example of an artificial structure is a laminin fragment.
  • The period for three-dimensional culture is not particularly limited because long-term culture would not cause any problems. For example, the culture period may be at least 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, or 12 days. The culture period is preferably at least 10 days, and particularly preferably 10 days, 11 days, or 12 days.
    • [Pluripotent Stem Cells]
  • Pluripotent stem cells that can be used in the present invention are stem cells that have the potential to differentiate into any types of cells existing in organisms (i.e., pluripotency) and have the potential to grow. Examples thereof include embryonic stem cells (ES cells), nuclear transfer-derived embryonic stem cells from cloned embryos (ntES cells), germline stem cells (GS cells), embryonic germ cells (EG cells), induced pluripotent stem cells (iPS cells), and pluripotent cells derived from cultured fibroblasts and myeloid stem cells (Muse cells). In the present invention, the use of iPS cells or Muse cells is preferable because cells of interest can be obtained without destroying embryos.
  • (A) Embryonic Stem Cells
  • ES cells are pluripotent stem cells having the potential to grow through autoreproduction, and they are established from embryoblasts of early embryos (e.g., blastocysts) of mammalians such as humans or mice.
  • ES cells are embryo-derived stem cells originating from embryoblasts of blastocysts, which are embryos after the 8-cell stage and the morula stage of fertilized eggs. Such ES cells have the potential to differentiate into any types of cells constituting an adult; that is, so-called pluripotency and potential to grow through autoreproduction. ES cells were discovered in mice in 1981 (M. J. Evans and M. H. Kaufman, 1981, Nature 292: 154-156). Thereafter, ES cells of primates, such as humans and monkeys, were also established (J. A. Thomson, et al., 1998, Science 282: 1145-1147; J. A. Thomson, et al., 1995, Proc. Natl. Acad. Sci., U.S.A., 92: 7844-7848; J. A. Thomson, et al., 1996, Biol. Reprod., 55: 254-259; J. A. Thomson and V. S. Marshall, 1998, Curr. Top. Dev. Biol., 38: 133-165).
  • ES cells can be established by extracting embryoblasts from blastocysts of fertilized eggs of target animals and culturing the embryoblasts in fibroblast feeders. Cells can be maintained via subculture with the use of a medium supplemented with substances such as leukemia inhibitory factors (LIF) and basic fibroblast growth factors (bFGF). Human and monkey ES cells can be established and maintained by the methods described in, for example, U.S. Pat. No. 5,843,780; Thomson J. A., et al., 1995, Proc. Natl. Acad. Sci., U.S.A., 92: 7844-7848; Thomson, J. A., et al., 1998, Science 282: 1145-1147; H. Suemori et al., 2006, Biochem. Biophys. Res. Commun., 345: 926-932; M. Ueno et al., 2006, Proc. Natl. Acad. Sci. U.S.A., 103:9554-9559; H. Suemori et al., 2001, Dev. Dyn., 222:273-279; H. Kawasaki et al., 2002, Proc. Natl. Acad. Sci. U.S.A., 99:1580-1585; and Klimanskaya I, et al., 2006, Nature 444: 481-485.
  • Human ES cells can be maintained with the use of a medium for the production of ES cells, such as a DMEM/F-12 medium supplemented with 0.1 mM 2-mercaptoethanol, 0.1 mM nonessential amino acids, 2 mM L-glutamic acid, 20% KSR, and 4 ng/ml bFGF, at 37□C in the presence of 5% CO2 in a moist atmosphere (H. Suemori, et al., 2006, Biochem. Biophys. Res. Commun., 345: 926-932). It is necessary that ES cells be subjected to subculture every 3 or 4 days. Subculture can be carried out with the use of 0.25% trypsin and 0.1 mg/ml collagenase IV in PBS containing 1 mM CaCl2 and 20% KSR.
  • In general, ES cells can be selected via real-time PCR using the expression of a gene marker such as alkaline phosphatase, Oct-3/4, or Nanog as an indicator. When human ES cells are to be selected, in particular, the expression of a gene marker such as OCT-3/4, NANOG, or ECAD can be employed as an indicator (E. Kroon et al., 2008, Nat. Biotechnol., 26: 443-452).
  • Human ES cells (e.g., WA01 (H1) and WA09 (H9)) are available from the WiCell Research Institute, and KhES-1, KhES-2, and KhES-3 are available from the Institute for Frontier Medical Sciences, Kyoto University (Kyoto, Japan).
  • (B) Germline Stem Cells
  • Germline stem cells are spermary-derived pluripotent stem cells that serve as sources for spermatogenesis. As with the case of ES cells, germline stem cells can be differentiated into various types of cells. For example, germline stem cells may be implanted into mouse blastocysts, so that chimeric mice may be produced (M. Kanatsu-Shinohara et al., 2003, Biol. Reprod., 69: 12-616; K. Shinohara et al., 2004, Cell, 119: 1001-1012). Germline stem cells are capable of autoreproduction in a medium containing glial cell line-derived neurotrophic factors (GDNF). In addition, germline stem cells can be obtained by repeating subculture under the same culture conditions as with those used for ES cells (Masanori Takebayashi et al., 2008, Experimental Medicine, Vol. 26, No. 5 (extra edition), pp. 41-46, Yodosha, Tokyo, Japan).
  • (C) Embryonic Germ Cells
  • As with ES cells, embryonic germ cells are pluripotent cells that are established from primordial germ cells during the prenatal period. Embryonic germ cells can be established by culturing primordial germ cells in the presence of substances such as LIF, bFGF, or stem cell factors (Y. Matsui et al., 1992, Cell, 70: 841-847; J. L. Resnicket al., 1992, Nature, 359: 550-551).
  • (D) Induced Pluripotent Stem Cells
  • Induced pluripotent stem (iPS) cells can be prepared by introducing particular reprogramming factors into somatic cells in the form of DNA or proteins. iPS cells are artificial stem cells derived from somatic cells that have substantially the same properties as ES cells, such as pluripotency and potential to grow through autoreproduction (K. Takahashi and S. Yamanaka, 2006, Cell, 126: 663-676; K. Takahashi et al., 2007, Cell, 131: 861-872; J. Yu et al., 2007, Science, 318: 1917-1920; Nakagawa, M. et al., Nat. Biotechnol., 26: 101-106, 2008; WO 2007/069666). Reprogramming factors may be 4composed of genes that are expressed specifically in ES cells, gene products or non-cording RNA thereof, or genes that play key roles in maintenance of the undifferentiated state of ES cells, gene products or non-coding RNA thereof, or low-molecular-weight compounds. Examples of genes included in reprogramming factors include Oct3/4, Sox2, Sox1, Sox3, Sox15, Sox17, K1f4, K1f2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15-2, Tcll, beta-catenin, Lin28b, Sa111, Sa114, Esrrb, Nr5a2, Tbx3, and Glis1. Such reprogramming factors may be used alone or in combination. Examples of combinations of reprogramming factors are described in WO 2007/069666, WO 2008/118820, WO 2009/007852, WO 2009/032194, WO 2009/058413, WO 2009/057831, WO 2009/075119, WO 2009/079007, WO 2009/091659, WO 2009/101084, WO 2009/101407, WO 2009/102983, WO 2009/114949, WO 2009/117439, WO 2009/126250, WO 2009/126251, WO 2009/126655, WO 2009/157593, WO 2010/009015, WO 2010/033906, WO 2010/033920, WO 2010/042800, WO 2010/050626, WO 2010/056831, WO 2010/068955, WO 2010/098419, WO 2010/102267, WO 2010/111409, WO 2010/111422, WO 2010/115050, WO 2010/124290, WO 2010/147395, WO 2010/147612, Huangfu D, et al., 2008, Nat. Biotechnol., 26: 795-797, Shi Y, et al., 2008, Cell Stem Cell, 2: 525-528, Eminli S, et al., 2008, Stem Cells, 26: 2467-2474, Huangfu D, et al., 2008, Nat. Biotechnol., 26: 1269-1275, Shi Y, et al., 2008, Cell Stem Cell, 3, 568-574, Zhao Y, et al., 2008, Cell Stem Cell, 3: 475-479, Marson A, 2008, Cell Stem Cell, 3, 132-135, Feng B., et al., 2009, Nat Cell Biol., 11: 197-203, R. L. Judson et al., 2009, Nat. Biotech., 27: 459-461, Lyssiotis C A, et al., 2009, Proc. Natl. Acad. Sci., U.S.A. 106: 8912-8917, Kim J B, et al., 2009, Nature, 461: 649-643, Ichida, J. K., et al., 2009, Cell Stem Cell, 5: 491-503, Heng J. C. et al., 2010, Cell Stem Cell, 6: 167-74, Han J, et al., 2010, Nature, 463: 1096-100, Mali P, et al., 2010, and Stem Cells, 28: 713-720, Maekawa M, et al., 2011, Nature, 474: 225-9.
  • Factors that are used to enhance cell establishment efficiency are within the scope of the reprogramming factors described above. Examples thereof include: histone deacetylase (HDAC) inhibitors, such as low-molecular-weight inhibitors, including valproic acid (VPA), trichostatin A, sodium butyrate, MC 1293, and M344, and nucleic acid-based expression inhibitors, including siRNAs and shRNAs against HDAC (e.g., HDAC1 siRNA Smartpoor (Millipore) and HuSH 29mer shRNA constructs against HDAC1 (OriGene)); MEK inhibitors (e.g., PD184352, PD98059, U0126, SL327, and PD0325901); glycogen synthase kinase-3 inhibitors (e.g., Bio and CHIR99021); DNA methyltransferase inhibitors (e.g., 5-azacytidine); histone methyltransferase inhibitors (e.g., low-molecular-weight inhibitors, such as BIX-01294, and nucleic acid-based expression inhibitors against Suv39h1, Suv39h2, SetDB1 and G9a, such as siRNAs and shRNAs); an L-channel calcium agonist (e.g., Bayk8644); butyric acid, TGFβ, and ALKS inhibitors (e.g., LY364947, SB431542, 616453, and A-83-01); p53 inhibitors (e.g., siRNA and shRNA against p53); ARID3A inhibitors (e.g., siRNA and shRNA against ARID3A), miRNA, such as miR-291-3p, miR-294, miR-295, and mir-302, Wnt signaling (e.g., soluble Wnt3a), neuro-peptide Y, prostaglandins (e.g., prostaglandin E2 and prostaglandinJ2), hTERT, SV40LT, UTF1, IRX6, GLIS1, PITX2, and DMRTB1. Factors that are used for the improvement of established efficiency are not particularly distinguished from reprogramming factors.
  • When reprogramming factors are in the form of proteins, for example, they may be introduced into somatic cells by a technique such as lipofection, fusion with cell-permeable peptides (e.g., HIV-derived TAT and polyarginine), or microinjection.
  • In contrast, reprogramming factors in the form of DNA can be introduced into somatic cells by a technique involving the use of a vector such as a virus, plasmid, or artificial chromosome vector, lipofection, a technique involving the use of a liposome, or microinjection, for example. Examples of virus vectors include retrovirus vectors, lentivirus vectors (Cell, 126, pp. 663-676, 2006; Cell, 131, pp. 861-872, 2007; Science, 318, pp. 1917-1920, 2007), adenovirus vectors (Science, 322, 945-949, 2008), adeno-associated virus vectors, and Sendai virus vectors (WO 2010/008054). Examples of artificial chromosome vectors include human artificial chromosome (HAC) vectors, yeast artificial chromosome (YAC) vectors, and bacterial artificial chromosome (BAC, PAC) vectors. Plasmids for mammalian animal cells can be used (Science, 322: 949-953, 2008). Vectors can comprise regulatory sequences, such as promoters, enhancers, ribosome-binding sequences, terminators, or polyadenylation sites, so that nuclear reprogramming substances can express. In addition, vectors can comprise selection marker sequences, such as drug tolerance genes (e.g., kanamycin tolerance genes, ampicillin tolerance genes, and puromycin tolerance genes), thymidine kinase genes, or diphtheria toxin genes, and reporter gene sequences, such as green fluorescent proteins (GFP), P-glucuronidase (GUS), or FLAG, according to need. The vector may comprise LoxP sequences in positions downstream and upstream of a gene encoding an reprogramming factor or a gene encoding a promoter and an reprogramming factor binding thereto, so as to eliminate such gene after the vector is introduced into somatic cells.
  • When reprogramming factors are in the form of RNA, for example, they may be introduced into somatic cells by a technique such as lipofection or microinjection. Alternatively, RNA comprising 5-methylcytidine and pseudouridine (TriLink Biotechnologies) incorporated therein may be used, so as to suppress degradation (Warren L, 2010, Cell Stem Cell 7: 618-630).
  • Examples of culture media used for iPS cell induction include DMEM containing 10% to 15% FBS, a DMEM/F12 or DME medium (such medium may adequately contain, for example, LIF, penicillin/streptomycin, puromycin, L-glutamine, nonessential amino acids, and (3-mercaptoethanol), commercially available culture media (e.g., a medium for mouse ES cell culture; TX-WES medium, Thrombo X), a medium for primate ES cell culture (a medium for primate ES/iPS cell culture, ReproCELL Incorporated), and a serum-free medium (mTeSR, Stemcell Technology).
  • For example, somatic cells are brought into contact with reprogramming factors in a 10% FBS-containing DMEM or DMEM/F12 medium, culture is conducted at 37□C in the presence of 5% CO2 for about 4 to 7 days, and the cells are reseeded in feeder cells (e.g., mitomycin C-treated STO cells or SNL cells). Culture is reinitiated in a medium for bFGF-containing primate ES cell culture about 10 days after the somatic cells are first brought into contact with the reprogramming factors, and iPS-like colonies can then be formed at least about 30 to 45 days after such contact.
  • Alternatively, culture may be conducted in a 10% FBS-containing DMEM medium (this medium can further contain LIF, penicillin/streptomycin, puromycin, L-glutamine, nonessential amino acids, (3-mercaptoethanol, or the like, according to need) in feeder cells (e.g., mitomycin C-treated STO cells or SNL cells) at 37□C in the presence of 5% CO2, and ES-like colonies can then be formed at least about 25 to 30 days later. Alternatively, use of the somatic cells to be reprogrammed instead of feeder cells is preferable (Takahashi K, et al., 2009, PLoS One, 4: e8067 or WO 2010/137746), or use of an extracellular matrix (e.g., laminin-5 (WO 2009/123349) and Matrigel (BD)) is preferable.
  • In addition, culture may be conducted with the use of a serum-free medium (Sun N, et al., 2009, Proc. Natl. Acad. Sci., U.S.A. 106: 15720-15725). In order to enhance cell establishment efficiency, iPS cells may be established under low-oxygen conditions (oxygen concentration of 0.1 % to 15%) (Yoshida Y, et al., 2009, Cell Stem Cell, 5: 237-241 or WO 2010/013845).
  • During the culture, medium exchange is initiated 2 days after the initiation of culture, and the medium is exchanged with fresh medium once a day. The number of somatic cells used for nuclear reprogramming is not limited, and it is about 5×103 to about 5×106 cells per 100 cm2 of a culture dish.
  • iPS cells can be selected in accordance with the configuration of the formed colonies. When drug tolerance genes that express in association with genes that express upon reprogramming of somatic cells (e.g., Oct3/4 and Nanog) are introduced as marker genes, in contrast, culture can be conducted in a medium containing corresponding drugs (i.e., a selection medium). Thus, established iPS cells can be selected. When marker genes are fluorescent protein genes, fluorescent microscopic observation may be carried out. When marker genes are luminescent enzyme genes, luminescent substrates may be added. When marker genes are chromogenic enzyme genes, chromogenic substrates may be added. Thus, iPS cells can be selected.
  • The term “somatic cells” used herein refers to any animal cells except for germline cells or pluripotent cells such as egg cells, oocytes, and ES cells (preferably mammalian animal cells, including those of humans). Examples of somatic cells include, but are not limited to, embryonic (fetal) somatic cells, neonatal (fetal) somatic cells, and mature healthy or affected somatic cells. Somatic cells may be primary-cultured cells, subcultured cells, or established cells. Specific examples of somatic cells include: (1) tissue stem cells, such as neural stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells (i.e., somatic stem cells); (2) tissue progenitor cells; (3) differentiated cells, such as lymphocytes, epidermic cells, endothelial cells, muscle cells, fibroblasts (e.g., skin cells), hair cells, hepatic cells, gastric mucosal cells, intestinal cells, splenic cells, pancreatic cells (e.g., pancreatic exocrine cells), brain cells, pneumocytes, nephrocytes, and adipocytes.
  • When iPS cells are used as materials for transplantation, use of somatic cells having the same or substantially the same HLA genotype as that of a recipient is preferable, so that rejection would not occur. When HLA genotypes are “substantially the same,” such HLA genotypes are concordant with each other to the extent that an immunosuppressive agent is able to suppress immune responses to the transplanted cells. For example, such somatic cells have HLA genotypes exhibiting concordance in 3 loci; i.e., HLA-A, HLA-B, and HLA-DR, or in 4 loci; i.e., HLA-A, HLA-B, HLA-DR, and HLA-C.
  • (E) Nuclear Transfer-Derived ES Cells from Cloned Embryos
  • “nt ES cells” are nuclear transfer-derived ES cells produced from cloned embryos, and such ES cells have substantially the same properties as fertilized egg-derived ES cells (T. Wakayama et al., 2001, Science, 292: 740-743; S. Wakayama et al., 2005, Biol. Reprod., 72: 932-936; J. Byrne et al., 2007, Nature, 450: 497-502). Specifically, nuclear transfer ES cells (i.e., nt ES cells) are ES cells that are established from embryoblasts of blastocysts derived from cloned embryos resulting from substitution of an unfertilized egg nucleus with a somatic cell nucleus. nt ES cells are produced by the technique of nuclear transfer (J. B. Cibelli et al., 1998, Nature Biotechnol., 16: 642-646) in combination with the technique of ES cell production (Kiyoka Wakayama et al., 2008, Experimental Medicine, Vol. 25, No. 5 (extra edition), pp. 47-52). In the case of nuclear transfer, somatic cell nuclei are injected into enucleated unfertilized eggs of mammalian animals, and culture is conducted for several hours. Thus, such cells can be reprogrammed.
  • (F) Multilineage-Differentiating Stress Enduring Cells (Muse Cells)
  • Muse cells are pluripotent stem cells produced by the method described in WO 2011/007900. More specifically, Muse cells are pluripotent cells that are obtained by treating fibroblasts or myeloid interstitial cells with tryp sin for a long period of time (preferably for 8 hours or 16 hours) and conducting float culture. Such cells are positive for SSEA-3 and CD105.
  • [Kit for Inducing Pluripotent Stem Cells to Differentiate into Alveolar Epithelial Progenitor Cells]
  • The present invention provides a kit used for inducing pluripotent stem cells to differentiate into alveolar epithelial progenitor cells or producing alveolar epithelial progenitor cells from pluripotent stem cells. The kit may comprise growth factors, compounds, a medium, a cell detachment solution, and an agent for coating the culture vessel used for the induction of differentiation. The kit may further comprise documents and/or instructions describing the procedure for the induction of differentiation.
  • [Method for Selecting Alveolar Epithelial Progenitor Cells]
  • In the present invention, alveolar epithelial progenitor cells may constitute a population of cells including alveolar epithelial progenitor cells. In the present invention, a population of cells including alveolar epithelial progenitor cells preferably includes 50%, 60%, 70%, 80%, or 90% or more alveolar epithelial progenitor cells.
  • Accordingly, the present invention provides a method for extracting alveolar epithelial progenitor cells. The cells to be extracted may be alveolar epithelial progenitor cells obtained by the method described above or cells obtained in the process for producing the same, which takes place after the completion of Step (3) of culture in a medium containing BMP4, retinoic acid, and a GSK3β inhibitor or Step (4) of culture in a medium containing FGF10. Alveolar epithelial progenitor cells can be extracted with the use of a reagent having CPM-specific affinity. While CPM had been known as a marker of adult type I alveolar epithelial cells, it was not previously known that CPM was expressed in progenitor cells during development. That is, use of CPM as a surface marker in the same manner as with NKX2-1 as a marker of alveolar epithelial progenitor cells has been found for the first time through the present invention.
  • Examples of reagents having specific affinity that can be used in the present invention include antibodies, aptamers, peptides, and compounds that specifically recognize the substances of interest, with antibodies or fragments thereof being preferable.
  • In the present invention, antibodies may be polyclonal or monoclonal antibodies. Such antibodies can be prepared in accordance with techniques well known in the art (Current protocols in Molecular Biology, Ausubel et al. (editors), 1987, John Wiley and Sons (publisher), Section 11.12-11.13). When the antibodies of the present invention are polyclonal antibodies, specifically, proteins encoded by CPM expressed in E. coli or mammalian cells and purified, oligopeptides having partial amino acid sequences, or glycolipids may be purified in accordance with conventional techniques, nonhuman animals such as rabbits may be immunized therewith, and antibodies of interest can be obtained from sera of the immunized animals in accordance with a conventional technique. In the case of monoclonal antibodies, in contrast, antibodies of interest can be obtained from hybridoma cells prepared via fusion of spleen cells obtained from immunized nonhuman animals to myeloma cells (Current protocols in Molecular Biology, Ausubel et al. (editors), 1987, John Wiley and Sons (publisher), Section 11.4-11.11). Examples of antibody fragments include a part of an antibody (e.g., an Fab fragment) and a synthetic antibody fragment (e.g., a single-stranded Fv fragment, ScFv). Antibody fragments, such as Fab and F(ab)2 fragments, can be prepared by a genetic engineering technique well known in the art. For example, antibodies reacting with CPM can be obtained from Leica Microsystems.
  • In order to recognize or separate cells that express CPM, reagents having relevant affinity may be bound or conjugated to substances that enable detection, such as a fluorescent label, a radioactive label, a chemoluminescent label, an enzyme, biotin, or streptoavidin, or substances that enable isolation and extraction, such as Protein A, Protein G, beads, or magnetic beads.
  • Alternatively, reagents having relevant affinity may be indirectly labeled. Labeling may be carried out in accordance with various techniques known in the art. For example, pre-labeled antibodies (secondary antibodies) that specifically bind to the antibodies described above may be used.
  • Alveolar epithelial progenitor cells can be extracted by, for example, a method comprising conjugating particles to a reagent having relevant affinity in order to precipitate the cells, a method involving the use of magnetic beads to select the cells with the aid of magnetism (e.g., MACS), a method involving the use of a cell sorter with the aid of a fluorescent label, or a method involving the use of a support upon which antibodies or the like are immobilized (e.g., a cell enrichment column).
  • [Agents for Treatment of Pulmonary Alveolar Diseases]
  • The alveolar epithelial progenitor cells obtained in the present invention can be administered to patients afflicted with diseases that destroy the pulmonary alveolus in the form of pharmaceutical preparations. The alveolar epithelial progenitor cells are prepared into the form of a sheet, and the sheet may be applied to the alveolar epithelium of a patient. Alternatively, the alveolar epithelial progenitor cells may be suspended in physiological saline or the like, and the suspension may then be directly implanted in the pulmonary alveolus of the patient. Accordingly, the present invention provides an agent for treatment of pulmonary alveolar diseases comprising alveolar epithelial progenitor cells obtained from pluripotent stem cells in the manner described above.
  • In the present invention, the number of alveolar epithelial progenitor cells contained in the agent for treatment of pulmonary alveolar diseases is not particularly limited, provided that the transplanted grafts are able to survive. The number of the cells may be adequately adjusted in accordance with lesion size or body size.
  • EXAMPLES
  • Hereafter, the present invention is described in greater detail with reference to the examples, although the technical scope of the present invention is not limited to these examples.
  • [iPS Cell Culture]
  • Human iPS cells (201B7) were provided by Professor Yamanaka at Kyoto University and cultured in accordance with a conventional technique (Takahashi K, et al. Cell, 131: 861-872, 2007). In accordance with the method described in Mae S., et al, Nat. Commun., 4: 1367, 2013, according to a gene knock-in technique, SFTPC-reporter 201B7 was produced by introducing an EGFP sequence into a site downstream of the SFTPC initiation codon of the human iPS cells (201B7).
    • [Induction of Alveolar Epithelial Progenitor Cells]
  • FIG. 1 shows a scheme for producing alveolar epithelial progenitor cells from pluripotent stem cells such as iPS cells.
  • The alveolar epithelial progenitor cells were induced by detaching human iPS cells with the use of Accutase, seeding the cells in a 24-well plate coated with Matrigel at 2.0×105 cells/well or in a 6-well plate coated with Matrigel at 9.6×105 cells/well, and conducting culture under the conditions described below (FIGS. 1A and 1B).
  • (Step 1)
  • The seeded cells (Day 0) were cultured in basal medium 1 (RPMI1640 (Nacalai Tesque) containing 2% B27 (Life Technologies) and a 0.5% penicillin/streptomycin stock solution (Life Technologies)) supplemented with 100 ng/ml Activin A (R&D Systems), 1 μM CHIR99021 and 10 μM Y-27632. On the following day (Day 1), the medium was exchanged with basal medium 1 containing 100 ng/ml Activin A, 1 μM CHIR99021, and 0.25 mM NaB, the medium was exchanged with another medium under the same conditions on the following day (Day 2) and 3 days later (Day 4), and culture was conducted for 5 days.
  • Alternatively, the seeded cells (Day 0) were cultured in basal medium 1 supplemented with 100 ng/ml Activin A, 1 μM CHIR99021, and 10 μM Y-27632. On the following day (Day 1), the medium was exchanged with basal medium 1 containing 100 ng/ml Activin A, 1 μM CHIR99021, 10 μM Y-27632, and 0.125 mM or 0.25 mM NaB, the medium was exchanged with basal medium 1 containing 100 ng/ml Activin A, 1 μM CHIR99021, and 0.125 mM or 0.25 mM NaB on the following day (Day 2), and the medium was exchanged with another medium under the same conditions 3 days later (Day 4).
  • (Step 2)
  • The cells obtained in Step 1 (Day 6) were cultured in basal medium 2 (DMEM/F12 medium (Life Technologies) containing 1% Glutamax supplement (Life Technologies), 2% B27 supplement, 1% N2 supplement (Life Technologies), 0.8% StemSure™ 50 mmol/l monothioglycerol solution (Wako), 50 μL-ascorbic acid (Sigma Aldrich), and 0.5% penicillin/streptomycin stock solution) supplemented with 200 ng/ml or 100 ng/ml hNoggin (R&D Systems) and 10 μM SB-431542 for 4 days. In this case, the medium was exchanged with another medium under the same conditions every other day.
  • (Step 3)
  • The cells obtained in Step 2 (Day 10) were cultured in basal medium 2 containing 100 ng/ml hBMP4 (HumanZyme, Inc.), 0.05 μM all-trans retinoic acid (ATRA), and 2.5 μM CHIR99021 for 4 days. In this case, the medium was exchanged with another medium under the same conditions every other day.
  • (Step 4)
  • The cells obtained in Step 3 (Day 14) were cultured in basal medium 2 containing 100 ng/ml FGF10 (Wako) for 7 days. In this case, the medium was exchanged with another medium under the same conditions every other day.
  • (Step 5)
  • After the media were exchanged, the cells obtained in Step 4 (Day 21) were cultured in basal medium 3 (Ham's F12 media (Wako) containing 3.33% BSA Fraction V Solution (7.5%) (Life Technologies), 15 mM HEPES (Sigma Aldrich), 0.8 mM CaCl2 (Nacalai Tesque), 1% ITS Premix (BD), and 0.5% penicillin/streptomycin stock solution) containing 50 nM dexamethasone (Sigma Aldrich), 0.1 mM 8-Br-cAMP (Biolog Life Science Institute), 0.1 mM 3-isobutyl-1-methylxanthine (IBMX) (Wako), and 100 ng/ml or 50 ng/ml KGF (Wako). Thereafter, the medium was exchanged with another medium under the same conditions every other day. The alveolar epithelial progenitor cells obtained were analyzed 4 days later (Day 25).
    • [Cell Analysis]
    (1) After the Completion of Step 3
  • After the completion of Step 3 (Day 14), the cells were subjected to immunostaining so as to inspect CPM and NKX2-1 expression. As a result, cells positive for both such markers were identified (FIGS. 2A and 2B). In addition, CPM-positive cells were separated from the cells obtained on Day 14 using MACS (Miltenyi Biotec) (FIGS. 3A and 3B), and the obtained cells were allowed to adhere to glass slides via cytospinning, followed by immunostaining. As a result, many thereof were found to also be positive for NKX2-1 (FIGS. 4A and 4B). The cells were analyzed using a flow cytometer. As a result, 92% of the CPM-positive cells obtained by MACS were found to be NKX2-1-positive (FIGS. 5A and 5B). Also, CPM-positive cells identified via MACS were subjected to quantitative RT-PCR, so as to assay CPM and NKX2-1 mRNA levels. As a result, sorting of CPM-positive cells was found to result in a remarkable increase in CPM and NKX2-1 mRNA levels (FIG. 6).
  • (2) After the Completion of Step 4
  • After the completion of Step 4 (Day 21), CPM-positive cells were separated from cells using MACS, and the CPM and NKX2-1 mRNA levels were assayed via quantitative RT-PCR. As a result, sorting of CPM-positive cells was found to result in a remarkable increase in CPM and NKX2-1 mRNA levels (FIG. 7).
  • (3) After the Completion of Step 5
  • After the completion of Step 5 (Day 25), CPM-positive cells were separated from cells using MACS, and the obtained cells were allowed to adhere to glass slides via cytospinning, followed by immunostaining. As a result, many thereof were found to also be positive for NKX2-1 (FIG. 8A). In addition, the cells induced to differentiate in the same manner with the use of SFTPC-reporter 201B7 were subjected to immunostaining. As a result, the presence of SFTPC- or proSPB-positive cells was observed in the CPM-positive cells (FIG. 8B). The percentage of the SFTPC-positive cells was found to be about 0.9% as a result of flow cytometric analysis (FIG. 8C).
  • Subsequently, the cells (201B7) after the completion of Step 5 (Day 25) were subjected to immunostaining, and CPM-, NKX2-1-, SFTPB-, SFTPC-, and CCSP-positive cells were identified (FIGS. 9A and 9B). In this case, marker genes were found to be also positive for CPM. In addition, CPM-positive cells obtained using MACS were subjected to quantitative RT-PCR, so as to assay the CPM, NKX2-1, SFTPA2, SFTPB, DCLAMP, SFTPC, CCSP, and NGFR mRNA levels. As a result, sorting of CPM-positive cells was found to result in a remarkable increase in the CPM and NKX2-1 mRNA levels (FIG. 10).
  • As described above, it was demonstrated that alveolar epithelial cells or progenitor cells thereof could be induced from iPS cells by the method of the present invention.
  • [Effects of CPM Marker]
  • CPM expression in human fetus-derived lung tissue (FIG. 11) and mouse fetus-derived lung tissue (E12.5, E15.5, and E17.5) (FIG. 12) was inspected. As a result, expression of CPM in combination with NKX2-1, SFTPC, and T1α was observed. Accordingly, CPM was found to be capable of recognizing alveolar epithelial progenitor cells at an early stage, such as during the prenatal period (humans: 3 to 7 weeks in the prenatal period; mice: 9 to 14 days in the prenatal period), in addition to the canalicular period of lung development (humans: 16 to 24 weeks in the prenatal period; mice: 16.5 to 17.5 days in the prenatal period) and the adenoid period (humans: 7 to 16 weeks in the prenatal period; mice: 14.0 to 16.5 days in the prenatal period).
  • As described above, it was found that alveolar epithelial progenitor cells could be recognized and extracted with the use of CPM as an indicator.
  • [Three-Dimensional Culture]
  • As described above, the CPM-positive cells (2×104 cells) extracted via MACS obtained after the completion of Step 3 with the use of SFTPC-reporter 201B7 were transferred to 12-well Cell Culture Inserts (BD Biosciences) supplemented with 400 μ1 of medium containing Matrigel in combination with basal medium 3 containing 50 nM dexamethasone, 0.1 mM 8-Br-cAMP, 0.1 mM IBMX, and 10 ng/ml KGF at a ratio of 1:1 together with 2×106 human fetus-derived pulmonary fibroblasts (PP002-F-1349, DV Biologics). Also, basal medium 3 containing 10 μM Y-27632, 50 nM dexamethasone, 0.1 mM 8-Br-cAMP, 0.1 mM IBMX, and 10 ng/ml KGF was added to the lower layer of the Cell Culture Inserts so as to form spheroids (cell masses), and culture was conducted for 10 to 12 days (FIG. 13). The resulting spheroids were inspected using a transmission electron microscope and found to be cells having lamella-like structures (FIG. 14A).
  • In addition, the spheroids were subjected to hematoxylin-eosin staining, and CPM(+)-cell-derived spheroids were found to be in the form of cystic pseudo-lamellar, cylindrical, or cubic cells having cytoplasm that would be stained dark pink, unlike CPM(−)-cell-derived spheroids having cytoplasm of pale color (FIG. 15A). These cells were double positive for both NKX2-1 and CPM. In addition, these cells included SFTPC-positive cells (FIG. 15B). In this case, AQPS-positive cells as markers of type I alveolar epithelial cells were found to be present adjacent to SFTPC-positive cells.
  • The alveolar epithelial cell marker expression was inspected and SFTPA, SFTPB, SFTPC, and SFTPD were found to be expressed in the CPM- and NKX2-1-positive cells (FIG. 16). As a result of quantitative PCR, the expression levels of these genes were found to have been elevated via three-dimensional culture. In addition, expression of SOX9 and ID2, which are indicators for induction of the peripheral airway, was observed in CPM- and NKX2-1-positive cells in several spheroids (FIG. 16).
  • While PDPN and CAV1 were expressed in fibroblast-like cells in the vicinity of the spheroids (indicated with arrows), they were also expressed in the spheroids (indicated with arrowheads) (FIG. 17).
  • As described above, it was found that alveolar epithelial cells were induced from CPM-positive cells, and CPM was found to be a useful marker of progenitor cells of alveolar epithelial cells. In addition, the CPM-positive cells obtained were found to be induced into mature alveolar epithelial cells via three-dimensional co-culture thereof with human fetus-derived pulmonary fibroblasts.
  • INDUSTRIAL APPLICABILITY
  • The method of the present invention enables production of alveolar epithelial progenitor cells from pluripotent stem cells.
  • All publications, patents, and patent applications cited herein are incorporated herein by reference in their entirety.

Claims (24)

1. A cell population predominantly comprising carboxypeptidase M (CPM)-positive cells as alveolar epithelial progenitor cells or alveolar epithelial cells.
2. The cell population according to claim 1, wherein the cell population is produced by extracting CPM-positive cells, as alveolar epithelial progenitor cells, from a cell population comprising alveolar epithelial progenitor cells using CPM as a marker, thereby obtaining a cell population predominantly comprising CPM-positive cells.
3. A method for producing a cell population predominantly comprising CPM-positive cells as alveolar epithelial progenitor cells comprising extracting CPM-positive cells as alveolar epithelial progenitor cells using CPM as a marker, from a cell population comprising alveolar epithelial progenitor cells, thereby obtaining a cell population predominantly comprising CPM-positive cells.
4. The cell population according to claim 1, wherein the cell population is produced by a method for producing alveolar epithelial progenitor cells from pluripotent stem cells comprising Steps (1) to (6):
(1) culturing pluripotent stem cells in a medium containing activin A and a glycogen synthase kinase 3β (GSK3β) inhibitor;
(2) culturing the cells obtained in Step (1) in a medium containing a bone morphogenic protein (BMP) inhibitor and a transforming growth factor β (TGFβ) inhibitor;
(3) culturing the cells obtained in Step (2) in a medium containing BMP4, retinoic acid, and a GSK3β inhibitor;
(4) culturing the cells obtained from Step (3) in a medium containing a fibroblast growth factor 10 (FGF10);
(5) culturing the cells obtained from Step (4) in a medium containing a steroid drug, a cyclic adenosine monophosphate (cAMP) derivative, a phosphodiesterase inhibitor, and a keratinocyte growth factor (KGF); and
(6) extracting CPM-positive cells as alveolar epithelial progenitor cells using the CPM as a marker following Step (5);
thereby obtaining a cell population predominantly comprising CPM-positive cells.
5. The cell population according to claim 4, wherein the medium of Step (1) further comprises a Rho kinase (ROCK) inhibitor and/or a histone deacetylase (HDAC) inhibitor.
6. The cell population according to claim 4, wherein Step (1) comprises culturing for 6 days or longer, Step (2) comprises culturing for 4 days or longer, Step (3) comprises culturing for 4 days or longer, Step (4) comprises culturing for 7 days or longer and Step (5) comprises culturing for 4 days or longer.
7. The cell population according to claim 4, wherein the GSK3β inhibitor is CHIR99021, the BMP inhibitor is Noggin, the TGFβ inhibitor is SB431542, the steroid drug is dexamethasone, the cAMP derivative is 8Br-cAMP, and the phosphodiesterase inhibitor is 3-isobutyl-1-methylxanthine (IBMX).
8. The cell population according to claim 1, wherein the cell population is produced by a method for producing alveolar epithelial progenitor cells from pluripotent stem cells comprising Steps (1) to (3b):
(1) culturing pluripotent stem cells in a medium containing activin A and a glycogen synthase kinase 3β (GSK3β) inhibitor;
(2) culturing the cells obtained in Step (1) in a medium containing a bone morphogenic protein (BMP) inhibitor and a transforming growth factor β (TGFβ) inhibitor;
(3a) culturing the cells obtained in Step (2) in a medium containing BMP4, retinoic acid, and a GSK3β inhibitor; and
(3b) extracting CPM-positive cells as alveolar epithelial progenitor cells using the CPM as a marker following Step (3a);
thereby obtaining a cell population predominantly comprising CPM-positive cells.
9. The cell population according to claim 8, wherein the medium of Step (1) in the method further comprises a Rho kinase (ROCK) inhibitor and/or a histone deacetylase (HDAC) inhibitor.
10. The cell population according to claim 8, wherein Step (1) comprises culturing for 6 days or longer, Step (2) comprises culturing for 4 days or longer, and Step (3a) comprises culturing for 4 days or longer.
11. The cell population according to claim 8, wherein the GSK3β inhibitor is CHIR99021, the BMP inhibitor is Noggin, and the TGFβ inhibitor is SB431542.
12. The cell population according to claim 1, wherein the cell population is produced by a method for producing alveolar epithelial cells from pluripotent stem cells comprising Steps (1) to (3c):
(1) culturing pluripotent stem cells in a medium containing activin A and a glycogen synthase kinase 3β (GSK3β) inhibitor;
(2) culturing the cells obtained in Step (1) in a medium containing a bone morphogenic protein (BMP) inhibitor and a transforming growth factor β (TGFβ) inhibitor;
(3a) culturing the cells obtained in Step (2) in a medium containing BMP4, retinoic acid, and a GSK3β inhibitor;
(3b) extracting CPM-positive cells as alveolar epithelial progenitor cells using the CPM as a marker following Step (3a); and
(3c) co-culturing the extracted alveolar epithelial progenitor cells together with human fetal pulmonary fibroblasts, following Step (3b);
thereby obtaining a cell population predominantly comprising CPM-positive cells.
13. The cell population according to claim 12, wherein the medium of Step (1) further comprises a Rho kinase (ROCK) inhibitor and/or a histone deacetylase (HDAC) inhibitor.
14. The cell population according to claim 12, wherein Step (1) comprises culturing for 6 days or longer, Step (2) comprises culturing for 4 days or longer, and Step (3a) comprises culturing for 4 days or longer.
15. The cell population according to claim 12, wherein the GSK3β inhibitor is CHIR99021, the BMP inhibitor is Noggin, and the TGFβ inhibitor is SB431542.
16. The cell population according to claim 12, wherein step (3c) is conducted by co-culturing in three-dimensional cell culture.
17. The cell population according to claim 1, wherein the CPM-positive cells are human CPM-positive cells.
18. The cell population according to claim 1, wherein the proportion of CPM-positive cells relative to total cells in the cell population is at least 50%.
19. The method according to claim 3, wherein the CPM-positive cells are human CPM-positive cells.
20. The method according to claim 3, wherein the proportion of CPM-positive cells relative to total cells in the obtained cell population is at least 50%.
21. An agent for treatment of a pulmonary alveolar disease comprising the cell population according to claim 1.
22. A method for treating a pulmonary alveolar disease comprising a step of administering the cell population according to claim 1 to a patient afflicted with a disease that destroy pulmonary alveolus.
23. The method according to claim 22, wherein a sheet comprising the cell population is applied to alveolar epithelium of the patient.
24. The method according to claim 22, wherein a suspension comprising the cell population is implanted in pulmonary alveolus of the patient.
US17/194,761 2013-04-12 2021-03-08 Method for inducing alveolar epithelial progenitor cells Pending US20210284968A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/194,761 US20210284968A1 (en) 2013-04-12 2021-03-08 Method for inducing alveolar epithelial progenitor cells

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
JP2013-084034 2013-04-12
JP2013084034 2013-04-12
PCT/JP2014/061106 WO2014168264A1 (en) 2013-04-12 2014-04-14 Method for inducing alveolar epithelium progenitor cells
US201514783382A 2015-10-08 2015-10-08
US17/194,761 US20210284968A1 (en) 2013-04-12 2021-03-08 Method for inducing alveolar epithelial progenitor cells

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/JP2014/061106 Continuation WO2014168264A1 (en) 2013-04-12 2014-04-14 Method for inducing alveolar epithelium progenitor cells
US14/783,382 Continuation US10961508B2 (en) 2013-04-12 2014-04-14 Method for inducing alveolar epithelial progenitor cells

Publications (1)

Publication Number Publication Date
US20210284968A1 true US20210284968A1 (en) 2021-09-16

Family

ID=51689663

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/783,382 Active 2034-07-24 US10961508B2 (en) 2013-04-12 2014-04-14 Method for inducing alveolar epithelial progenitor cells
US17/194,761 Pending US20210284968A1 (en) 2013-04-12 2021-03-08 Method for inducing alveolar epithelial progenitor cells

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/783,382 Active 2034-07-24 US10961508B2 (en) 2013-04-12 2014-04-14 Method for inducing alveolar epithelial progenitor cells

Country Status (5)

Country Link
US (2) US10961508B2 (en)
EP (1) EP2985344B1 (en)
JP (1) JP6461787B2 (en)
CA (1) CA2909230C (en)
WO (1) WO2014168264A1 (en)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201212960D0 (en) * 2012-07-20 2012-09-05 Common Services Agency Erythroid production
CA2909230C (en) 2013-04-12 2021-06-15 Kyoto University Method for inducing alveolar epithelial progenitor cells
WO2016143803A1 (en) * 2015-03-06 2016-09-15 国立大学法人京都大学 Method for inducing differentiation of alveolar epithelial cells
WO2016148307A1 (en) * 2015-03-19 2016-09-22 国立大学法人京都大学 Method for inducing differentiation of airway epithelial cells
AU2016321301B2 (en) * 2015-09-11 2022-06-02 The General Hospital Corporation Regeneration of a functional pulmonary vascular bed
US10624992B2 (en) 2016-05-16 2020-04-21 The General Hospital Corporation Human airway stem cells in lung epithelial engineering
US20180164221A1 (en) 2016-12-07 2018-06-14 Progenity Inc. Gastrointestinal tract detection methods, devices and systems
WO2018112240A1 (en) 2016-12-14 2018-06-21 Progenity Inc. Treatment of a disease of the gastrointestinal tract with a tnf inhibitor
CN110832069B (en) * 2017-05-31 2023-06-20 北昊干细胞与再生医学研究院有限公司 Methods for chemically induced lineage reprogramming
US10975357B2 (en) * 2017-06-27 2021-04-13 Trustees Of Boston University Methods and compositions related to differentiated lung cells
CN111566206A (en) * 2017-11-10 2020-08-21 纪念斯隆-凯特琳癌症中心 Derivation of somatotropin cells from stem cells and uses thereof
CN110218696A (en) * 2018-03-01 2019-09-10 中国科学院广州生物医药与健康研究院 A kind of cultivating system generated for chemical induction multipotent stem cells and the chemical reprogramming method using the cultivating system
EP3810095A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a tnf inhibitor
CN116726361A (en) 2018-11-19 2023-09-12 比奥拉治疗股份有限公司 Methods and devices for treating diseases with biologic therapeutic agents
WO2020198137A1 (en) * 2019-03-25 2020-10-01 The Trustees Of The University Of Pennsylvania Regenerative therapy based on mirna-302 mimics for enhancing host recovery from pneumonia caused by streptococcus pneumoniae
WO2021087354A1 (en) * 2019-10-30 2021-05-06 The General Hospital Corporation High-throughput culture of ipsc-derived alveolar cells
CN111424014B (en) * 2019-11-22 2022-03-29 上海交通大学医学院附属第九人民医院 Culture medium of immortalized cell strain of human jugular auxiliary nerve ganglionic tumor
WO2021119482A1 (en) 2019-12-13 2021-06-17 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
WO2023286852A1 (en) 2021-07-15 2023-01-19 株式会社セルファイバ Structure and use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10961508B2 (en) * 2013-04-12 2021-03-30 Kyoto University Method for inducing alveolar epithelial progenitor cells

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
AU2002245709A1 (en) 2001-03-23 2002-10-08 Bayer Corporation Rho-kinase inhibitors
AR035791A1 (en) 2001-03-23 2004-07-14 Bayer Corp COMPOUND N, N-DIHETEROCICLICO OF AMINA, INHIBITOR OF THE RHO-KINASE, ITS USE FOR THE MANUFACTURE OF A MEDICINAL PRODUCT AND PROCESS FOR THE PREPARATION OF THE COMPOUND
AU2003202263A1 (en) 2002-01-10 2003-07-30 Bayer Healthcare Ag Roh-kinase inhibitors
CA2473510A1 (en) 2002-01-23 2003-07-31 Bayer Pharmaceuticals Corporation Pyrimidine derivatives as rho-kinase inhibitors
US6943172B2 (en) 2002-01-23 2005-09-13 Bayer Pharmaceuticals Corporation Rho-kinase inhibitors
EP1562935B1 (en) 2002-10-28 2006-09-06 Bayer HealthCare AG Heteroaryloxy-substituted phenylaminopyrimidines as rho-kinase inhibitors
US9453219B2 (en) 2003-05-15 2016-09-27 Mello Biotech Taiwan Co., Ltd. Cosmetic designs and products using intronic RNA
EP4223769A3 (en) 2005-12-13 2023-11-01 Kyoto University Nuclear reprogramming factor
EP2137296A2 (en) 2007-03-23 2009-12-30 Wisconsin Alumni Research Foundation Somatic cell reprogramming
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
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
JP5558097B2 (en) 2007-12-10 2014-07-23 国立大学法人京都大学 Efficient nuclear initialization method
AU2008338989A1 (en) 2007-12-17 2009-06-25 Gliamed, Inc. Stem-like cells and method for reprogramming adult mammalian somatic cells
EP2229444B1 (en) 2008-01-16 2019-10-30 Shi-Lung Lin Generation of tumor-free embryonic stem-like pluripotent cells using inducible recombinant rna agents
EP2250252A2 (en) 2008-02-11 2010-11-17 Cambridge Enterprise Limited Improved reprogramming of mammalian cells, and the cells obtained
EP2090649A1 (en) 2008-02-13 2009-08-19 Fondazione Telethon Method for reprogramming differentiated cells
US20110014164A1 (en) 2008-02-15 2011-01-20 President And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
CN102027105B (en) 2008-03-17 2015-03-11 斯克里普斯研究所 Combined chemical and genetic approaches for generation of induced pluripotent stem cells
WO2009114949A1 (en) 2008-03-20 2009-09-24 UNIVERSITé LAVAL Methods for deprogramming somatic cells and uses thereof
CN102131919B (en) 2008-03-31 2017-05-03 东方酵母工业株式会社 Method for proliferation of pluripotent stem cell
WO2009126250A2 (en) 2008-04-07 2009-10-15 Nupotential, Inc. Reprogramming a cell by inducing a pluripotent gene through rna interference
JP2011521969A (en) 2008-05-30 2011-07-28 スムマ ヘルス システムズ エルエルシー Methods of using TGF-β receptor inhibitors or activin-like kinase (ALK) 5 inhibitors, A-83-01 and SB-431542 for treating ocular diseases and wound treatment conditions
CA2695590C (en) 2008-06-27 2018-01-09 Kyoto University Method of efficiently establishing induced pluripotent stem cells
AU2009271149A1 (en) 2008-07-14 2010-01-21 Oklahoma Medical Research Foundation Production of pluripotent cells through inhibition of Bright/ARID3a function
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
CN107012123A (en) 2008-07-30 2017-08-04 国立大学法人京都大学 The method for effectively setting up the multipotential stem cell of induction
WO2010147612A1 (en) 2009-06-18 2010-12-23 Lixte Biotechnology, Inc. Methods of modulating cell regulation by inhibiting p53
US20120034192A1 (en) 2008-09-19 2012-02-09 Young Richard A Compositions and methods for enhancing cell reprogramming
US20120021519A1 (en) 2008-09-19 2012-01-26 Presidents And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
WO2010042800A1 (en) 2008-10-10 2010-04-15 Nevada Cancer Institute Methods of reprogramming somatic cells and methods of use for such cells
US20110250692A1 (en) 2008-10-30 2011-10-13 Kyoto University Method for producing induced pluripotent stem cells
WO2010056831A2 (en) 2008-11-12 2010-05-20 Nupotential, Inc. Reprogramming a cell by inducing a pluripotent gene through use of an hdac modulator
US9045737B2 (en) 2008-12-13 2015-06-02 Dnamicroarray, Inc. Artificial three-dimensional microenvironment niche culture
US8951801B2 (en) 2009-02-27 2015-02-10 Kyoto University Method for making IPS cells
US20120122212A1 (en) 2009-03-06 2012-05-17 Marica Grskovic Tgf-beta pathway inhibitors for enhancement of cellular reprogramming of human cells
WO2010111422A2 (en) 2009-03-25 2010-09-30 The Salk Institute For Biological Studies Induced pluripotent stem cell generation using two factors and p53 inactivation
WO2010111409A2 (en) 2009-03-25 2010-09-30 The Salk Institute For Biological Studies Pluripotent stem cells
US8852940B2 (en) 2009-04-01 2014-10-07 The Regents Of The University Of California Embryonic stem cell specific microRNAs promote induced pluripotency
WO2010124290A2 (en) 2009-04-24 2010-10-28 Whitehead Institute For Biomedical Research Compositions and methods for deriving or culturing pluripotent cells
WO2010137746A1 (en) 2009-05-29 2010-12-02 Kyoto University Method for producing induced pluripotent stem cells and method for culturing the same
WO2010147395A2 (en) 2009-06-16 2010-12-23 Korea Research Institute Of Bioscience And Biotechnology Medium composition comprising neuropeptide y for the generation, maintenance, prologned undifferentiated growth of pluripotent stem cells and method of culturing pluripotent stem cell using the same
US9550975B2 (en) 2009-07-15 2017-01-24 Mari Dezawa SSEA-3 pluripotent stem cell isolated from body tissue
US9234170B2 (en) 2010-04-25 2016-01-12 Mount Sinai School Of Medicine Generation of anterior foregut endoderm from pluripotent cells
EP2596096B1 (en) * 2010-07-21 2018-01-31 Kyoto University Method for inducing differentiation of human pluripotent stem cell into intermediate mesoderm cell
US20130224116A1 (en) * 2010-11-05 2013-08-29 TransBio Ltd. Markers of Endothelial Progenitor Cells and Uses Thereof
EP2484754A1 (en) 2011-02-07 2012-08-08 Medizinische Hochschule Hannover Novel method for the production of differentiated respiratory epithelial cells
JP5710439B2 (en) 2011-10-06 2015-04-30 株式会社日立製作所 Template delivery type cancelable biometric authentication system and method
WO2014018691A1 (en) 2012-07-24 2014-01-30 The Trustees Of Columbia University In The City Of New York Generation of airway and lung progenitors and epithelial cells and three-dimensional anterior foregut spheres
JP2014023519A (en) 2012-07-24 2014-02-06 Trustees Of Columbia Univ In The City Of New York Generation of lung and airway epithelium from human pluripotent stem cells and use thereof
CA2886027C (en) 2012-09-25 2023-03-07 Yale University Differentiation of human ips cells to human alveolar type ii via definitive endoderm

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10961508B2 (en) * 2013-04-12 2021-03-30 Kyoto University Method for inducing alveolar epithelial progenitor cells

Also Published As

Publication number Publication date
EP2985344A4 (en) 2016-09-14
JP6461787B2 (en) 2019-01-30
US20160068816A1 (en) 2016-03-10
CA2909230C (en) 2021-06-15
US10961508B2 (en) 2021-03-30
JPWO2014168264A1 (en) 2017-02-16
WO2014168264A1 (en) 2014-10-16
CA2909230A1 (en) 2014-10-16
EP2985344B1 (en) 2018-07-18
EP2985344A1 (en) 2016-02-17

Similar Documents

Publication Publication Date Title
US20210284968A1 (en) Method for inducing alveolar epithelial progenitor cells
US20220186189A1 (en) Method for inducing differentiation of alveolar epithelial cells
KR102320537B1 (en) New method for inducing dopamine-producing neural precursor cells
AU2017254268B2 (en) Method for producing dopamine-producing neural precursor cells
EP2998391B1 (en) Efficient myocardial cell induction method
EP2737057B1 (en) Novel markers for dopaminergic neuron progenitor cells
WO2015020113A1 (en) Method for producing pancreatic hormone-producing cell
US10377991B2 (en) Method of producing airway epithelial cells
US20210332329A1 (en) Novel renal progenitor cell marker and method for concentrating renal progenitor cells using same
EPITHELS et al. LLLLL GGGG GGGGGGGG LLLLL GGGGGGGG

Legal Events

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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