[go: up one dir, main page]

WO2011016485A1 - METHOD FOR INDUCING DIFFERENTIATION OF iPS CELLS INTO HEPATIC PARENCHYMAL CELLS - Google Patents

METHOD FOR INDUCING DIFFERENTIATION OF iPS CELLS INTO HEPATIC PARENCHYMAL CELLS Download PDF

Info

Publication number
WO2011016485A1
WO2011016485A1 PCT/JP2010/063185 JP2010063185W WO2011016485A1 WO 2011016485 A1 WO2011016485 A1 WO 2011016485A1 JP 2010063185 W JP2010063185 W JP 2010063185W WO 2011016485 A1 WO2011016485 A1 WO 2011016485A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
ips
differentiation
hepatocytes
cell
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.)
Ceased
Application number
PCT/JP2010/063185
Other languages
French (fr)
Japanese (ja)
Inventor
直哉 小林
和秀 山本
雅也 岩室
克身 持立
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.)
Okayama University NUC
Original Assignee
Okayama University NUC
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 Okayama University NUC filed Critical Okayama University NUC
Priority to JP2011525912A priority Critical patent/JPWO2011016485A1/en
Publication of WO2011016485A1 publication Critical patent/WO2011016485A1/en
Anticipated expiration legal-status Critical
Ceased 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
    • C12N5/067Hepatocytes
    • 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
    • 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
    • C12N2500/60Buffer, e.g. pH regulation, osmotic pressure
    • C12N2500/62DMSO
    • 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
    • C12N2501/12Hepatocyte growth factor [HGF]
    • 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
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • the present invention relates to a method for inducing differentiation from iPS cells (induced pluripotent stem cells) to functional hepatocytes and hepatocytes differentiated thereby.
  • Non-Patent Documents 1 to 3 For example, in the reports on the induction of hepatocyte differentiation in ES cells, the mainstream is that the embryoid body formation period is 2 to 2.5 days (Non-Patent Documents 1 to 3).
  • ES cells have pluripotency and can differentiate into any of the three germ layer cells. Moreover, ES cells have infinite proliferation ability in an experimental system. Therefore, it is possible to treat patients with liver diseases by inducing differentiation of many stem cells from ES cells and using them as a material for bioartificial liver.
  • liver diseases by inducing differentiation of many stem cells from ES cells and using them as a material for bioartificial liver.
  • immunosuppressive agents is indispensable, and complications such as infections may occur.
  • ethical problems are involved in the establishment and operation of ES cells.
  • iPS cells do not require embryos of fertilized eggs, and because it is possible to use the patient's own somatic cells, it is expected to be a promising cell source for both ethical issues and rejection. Yes.
  • iPS cell differentiation induction is not yet sufficient.
  • definitive endoderm which is a precursor tissue of hepatic parenchymal cells compared to ES cells.
  • differentiation efficiency is low, and further, the function of the obtained hepatocytes is not sufficient.
  • the basement membrane is composed of proteins such as laminin, entactin, type IV collagen, and extracellular matrix (ECM) such as proteoglycan, which are directly below the basal basal surface, the inner lower surface of epithelial tissue, and contain cells. It is a structure of 50 to 100 nm, and is considered essential for immature epithelial cells to proliferate and differentiate into mature cells to express their original form and function.
  • epithelial cells have been cultured on a collagen matrix in which fibroblasts are embedded, or on a collagen fiber substrate to which Matrigel (Matrigel (registered trademark), Becton Dickinson) has been added.
  • Matrigel Matrigel (registered trademark), Becton Dickinson) has been added.
  • In vitro production methods such as a culture method have been developed, and examples of use as a culture substrate and production of artificial tissue have been reported (Patent Documents 1 to 3).
  • An object of the present invention is to provide a method for efficiently inducing differentiation of functionally superior liver parenchymal cells from iPS cells, and functional liver as a bioartificial liver material without causing ethical problems. Providing parenchymal cells.
  • the present invention is a method for inducing differentiation from iPS cells to hepatocytes, (A) a step of suspension culture of iPS cells to form an embryoid body; (B) inducing definitive endoderm by adhering and culturing 10-15 embryoid bodies on a plate using gelatin as a coating agent per well, and (c) adding HGF and DMSO,
  • the present invention relates to a method comprising inducing hepatocyte differentiation by culturing definitive endoderm on a basement membrane substrate.
  • the suspension culture period in step (a) is preferably 5 days.
  • the basement membrane matrix is preferably derived from alveolar type II epithelial cells or liver parenchymal cells.
  • the differentiation inducing method of the present invention functionally superior hepatocytes can be obtained.
  • definitive endoderm can be stably obtained, and differentiation into hepatocytes can be efficiently induced.
  • the differentiated hepatocytes of the present invention are hepatocytes based on iPS cells, ethical problems associated with the establishment and operation of ES cells can be avoided.
  • individual-derived iPS cells individual-specific hepatocytes that do not require immunosuppressive agents can be obtained. Therefore, hepatocytes derived from the iPS cells of the present invention are more suitable as a material for bioartificial liver.
  • FIG. 1 It is a figure which shows the differentiation process from an iPS cell to a hepatocyte.
  • A is a scheme showing the passage of days and the differentiation process.
  • B to E are (B) iPS cells, (C) embryoid bodies (EB), (D) definitive endoderm (DE), and (E to G) iPS in the process of differentiation from iPS cells to hepatocytes, respectively.
  • 2 is a phase contrast micrograph of cell-derived hepatocytes (iPS-Heps).
  • E shows iPS-Heps in Comparative Example 1,
  • F in Example 1
  • G in Example 2. It is a microscope image which shows the expression of albumin by immunohistochemical staining.
  • Example 4 Comparative Example 1, undifferentiated iPS cells, and T3-Alb7-sBM substrate-only sample were used as controls.
  • Microscopic image Alb was expressed using rabbit anti-mouse albumin antibody (Cat. No. 64560, MP biomedicals) as the primary antibody and DyLight54-labeled sheep anti-rabbit antibody (Catalog No. STAR36D549, MorphoSys UK) as the secondary antibody. ) Is dyed.
  • the microscopic image DAPI shows a cell nucleus immunostained with DAPI (Catalog No. H-1200, manufactured by Vector Laboratories Inc.). MERGE indicates a superposition of two microscopic images of Alb and DAPI.
  • FIG. 6B In the strongly magnified image of the phase contrast microscope observation (FIG. 6B), polygonal cells having an abundant circular nucleus and abundant granules in the cytoplasm are clearly observed.
  • FIG. 6C is a diagram showing expression of various genes in cells at each stage of the differentiation induction process of Comparative Example 1.
  • AFP alphafetoprotein
  • Albumin Alb
  • transferrin Trf
  • phosphoenolpyruvate carboxykinase 1 PCK1
  • CPS carbamyl phosphate synthetase
  • Sox17 sex determination gene box 17
  • Lanes 1-4 show iPS cells, embryoid bodies, definitive endoderm and iPS-Heps, respectively.
  • the relative mRNA level in each differentiation process of the comparative example 1 of alpha fetoprotein (AFP), albumin gene (Alb), transferrin (Trf), phosphoenolpyruvate carboxylase 1 (PCK1), and carbamyl phosphate synthetase (CPS) is shown. It is a graph.
  • the present invention relates to a method for inducing differentiation of iPS cells into functional hepatocytes.
  • the method of inducing differentiation of hepatocytes from the iPS cells of the present invention comprises: (a) a step of suspension-culturing iPS cells to form embryoid bodies; (b) transferring embryoid bodies to a plate using gelatin as a coating agent. A step of inducing definitive endoderm by adhering and culturing as 10-15 per well, and (c) hepatic parenchyma by adding HGF and DMSO and culturing definitive endoderm on a basement membrane substrate Inducing cell differentiation.
  • per well when “per well” is described, it means one well of a 12-well culture plate, and the bottom area of one well means 3.6 cm 2 .
  • the iPS cells were preferably suspended in culture for 5 days to form embryoid bodies.
  • the mainstream is that the embryoid body formation period is 2 to 2.5 days.
  • iPS cells have a lower efficiency of differentiation into definitive endoderm, which is a precursor tissue of hepatic parenchymal cells, compared to ES cells, and there is a tendency that sufficient definitive endoderm cannot be obtained in the case of definitive body formation for 2 to 2.5 days. is there. Therefore, for the formation of embryoid bodies, suspension culture for 5 days is preferable in order to increase the efficiency of differentiation into definitive endoderm.
  • Embryoid bodies were cultured in 12-well gelatin-coated plates, and 10 to 15 embryoid bodies per well were induced to start definitive endoderm induction.
  • the differentiation induction process cell growth starts from around the embryoid body and these differentiate into hepatocytes, so it is important to transfer the embryoid body to the plate at an appropriate density.
  • the density of embryoid bodies is low (for example, less than 10 per well), fewer hepatocytes are obtained at the end of differentiation induction.
  • the density of embryoid bodies is high (for example, when there are more than 15 per well), differentiation into cardiomyocytes is likely to be induced, so that the purity of the obtained hepatocytes decreases.
  • cells tend to be killed due to lack of medium. Therefore, it was important to start definitive endoderm induction with 10 to 15 embryoid bodies per well.
  • a serum-free medium that is, knockout DMEM and knockout serum substitute (KSR) was used.
  • a serum medium has been used in the differentiation induction process of ES cells.
  • the serum-free medium showed higher cell proliferation ability than the serum medium.
  • HGF hepatocyte growth factor
  • DMSO dimethyl sulfoxide
  • hepatocytes can be induced from iPS cells effectively and stably.
  • IPS cells are preferably derived from mammals, and examples of mammals include humans, primates such as cynomolgus monkeys, and mice.
  • mouse iPS cells are available from the RIKEN Cell Bank.
  • iPS cells are passaged, maintained and proliferated under non-differentiating conditions (hereinafter, this step may be referred to as a preparation step), and a part thereof is used as necessary. Induces differentiation into hepatocytes.
  • a container coated with feeder cells as a container for culturing iPS cells from the viewpoint of proliferation ability.
  • feeder cells include mouse embryonic fibroblasts irradiated with ⁇ rays.
  • mouse-derived iPS cells it is preferable to use a culture vessel coated with feeder cells.
  • the same conditions as for normal ES cells can be employed.
  • the main points of the preparation process include changing the medium every 24 hours to prevent nutrient depletion, and subculturing before the cell density increases and the colonies adhere to each other.
  • the culture temperature is preferably in the range of 36 to 37 ° C.
  • the pH is preferably in the range of 7.3 to 7.4.
  • leukemia inhibitory factor LIF is generally used to suppress differentiation.
  • hepatic parenchymal cells induced to differentiate from iPS cells by the method of the present invention can be confirmed by observation of morphological characteristics and reverse transcription polymerase chain reaction (RT-PCR).
  • the morphological confirmation method is specific to liver parenchymal cells, such as cells with multiple nuclei observed with a phase contrast microscope and abundant granules in the cytoplasm observed with an electron microscope, especially glycogen granules. Specific morphological features.
  • the expression of induced hepatic parenchymal cells can be evaluated from the expression of the gene by RT-PCR. If it can be confirmed by RT-PCR or the like that the albumin gene is expressed and that the undifferentiated ⁇ -fetoprotein is not expressed, it can be said that differentiation is induced.
  • albumin carbamyl phosphate synthetase (CPS), an enzyme related to urea synthesis, phosphoenolpyruvate carboxykinase 1 (PCK1), transferrin (Trf), an enzyme that regulates gluconeogenesis It is preferable that gene expression such as is observed.
  • hepatocyte determines whether or not it can actually function as a hepatocyte. Moreover, whether or not it can actually function as a hepatocyte is largely determined by whether it can be metabolized by adding ammonia, lidocaine, diazepam, or the like to the medium. Moreover, if albumin and urea are produced in the medium, it can be said that they have differentiated into sufficiently functional hepatocytes.
  • the basement membrane substrate used in the present invention various substrates can be used and are not particularly limited.
  • MAST polymer maleic ⁇ anhydride-styrene copolymer
  • Matrigel is a basement membrane preparation extracted from the Engelbreth-Holm-Swarm tumor matrix (J. Exp. Med. 145, 204-220, 1977) and may affect extracellular matrix synthesis. In addition to various cytokines, laminin-111, entactin, type IV collagen, perlecan and the like are included (Exp. Cell Res. 202, 1-8, 1992).
  • Various cells such as epithelial cells, vascular endothelial cells, and glia can be used as cells having the ability to form a basement membrane.
  • cells derived from lung such as alveolar type II epithelial cells
  • cells derived from liver such as liver parenchymal cells, or 293 cells are genetically modified to stabilize hLN-511.
  • rLN-10 cells that are expressed and secreted in large quantities extracellularly, and most preferably hepatocytes.
  • established cells communityrous and immortalized cell lines
  • Laminin the main glycoprotein of the basement membrane, is a complex composed of three types of ⁇ , ⁇ , and ⁇ subunits, and there are various isoforms such as laminin-111 ( ⁇ 1 ⁇ 1 ⁇ 1) and laminin-511 ( ⁇ 5 ⁇ 1 ⁇ 1). .
  • hepatic parenchymal cells can be improved.
  • hepatic sinusoidal endothelial cells bile duct epithelial cells, hepatic stellate cells, and the like can be used, and human hepatic sinusoidal endothelial cell lines are preferred.
  • a preferred embodiment of the present invention is a method for inducing differentiation from iPS cells to hepatocytes, wherein (a) the iPS cells are cultured in suspension to form embryoid bodies, (b) as a coating agent A step of inducing definitive endoderm by attaching 10-15 embryoid bodies to a plate using gelatin and culturing; and (c) adding HGF and DMSO on a basement membrane substrate
  • the present invention relates to a method comprising a step of inducing hepatocyte differentiation by co-culturing definitive endoderm with hepatic sinusoidal endothelial cells.
  • iPS cells are differentiated into definitive endoderm, pooled to the required number of cells, and then differentiated into hepatocytes as necessary, so that a large number of safe hepatocytes can be produced without causing ethical problems. Can supply.
  • the development of a bioartificial liver that can benefit everyone by using this as the cell source for bioartificial liver is greatly expected.
  • liver parenchymal cells induced to differentiate from iPS cells can be used as a cell source for bioartificial liver.
  • a bioartificial liver is an artificial liver device that mimics the liver in the living body, in which liver parenchymal cells are incorporated into a carrier, fixed, and made into a reactor. The patient's blood is guided into the device, and the metabolic capacity of liver parenchymal cells is used.
  • the apparatus can remove toxins in blood and supply physiologically active substances such as coagulation factors derived from liver cells.
  • bioartificial liver examples include a hybrid type artificial liver combining a hollow fiber type reactor (device) and separated / cultured cells.
  • bioartificial livers There are three types of bioartificial livers: those that are attached outside the body and connected to blood vessels, those that are placed in the body and connected to blood vessels, and those that are placed in the abdominal cavity without being connected to blood vessels.
  • the hepatocytes of the present invention can be used in any form of bioartificial liver, but are preferably in vitro from the viewpoint of avoiding the risks associated with cell transfer.
  • Bioreactors used in bioartificial livers include Cedars-Sinai Medical Center (Los Angeles, California, USA) with the support of Circe Biomedical Inc. (Lexington, Massachusetts, USA). HepatAssist (Hui T, ⁇ ⁇ ⁇ Rozga J, Demetriou AA. J Hepatobiliary Pancreat Surg 2001; 8: 1-15) for the treatment of bioartificial liver using porcine liver parenchymal cells centered on Demetriou et al. .) And German MERLach et al.'S MELS (Modular Extracorporporeal Liver System) using porcine liver parenchymal cells are known.
  • a reactor composed of a hollow fiber and a non-woven fabric is more preferably used so as to provide a scaffold for hepatocytes (Naoya Kobayashi, et al., “Hybrid Artificial Liver”, Hepatobiliary Pancreas, 2003, 46, p381-393) .
  • hepatocytes particularly human hepatocytes, induced to differentiate from iPS cells by the method of the present invention are useful as cells for drug metabolism tests. That is, it is useful as a test cell in new drug development, safety test, toxicity test and the like.
  • Liver parenchymal cells are the center of various drug metabolisms, and in order to test the toxicity and safety of drugs, it is necessary to administer drugs to hepatic parenchymal cells and examine how they are metabolized. And since there is a big difference between humans and experimental animals in substance metabolism, the final test is indispensable using human hepatocytes. In this respect, the present invention that can provide a large amount of human hepatocytes of uniform quality is useful. As a device used for such a metabolic test, a device having the same structure as the bioartificial liver can be used.
  • hepatocytes particularly human hepatocytes, induced to differentiate from iPS cells by the method of the present invention can be used for the production of physiologically active substances.
  • physiologically active substances include albumin and various blood coagulation factors.
  • a device for producing such a physiologically active substance one having the same structure as that of the bioartificial liver can be used.
  • Examples 1 to 4 Induction of differentiation from iPS cells to hepatocytes (1) Cultivation of undifferentiated iPS cells Mouse iPS cells (Cell no. APS0001, Cell name iPS-MEF-Ng) obtained from the RIKEN BioResource Center through the Ministry of Education, Culture, Sports, Science and Technology's National BioResource Project -20D-17, Lot no. 006). Mouse iPS cells are feeders of mouse embryo fibroblasts (MEF) (Dainippon Sumitomo Pharma Co., Ltd.) inactivated by mitomycin C (Catalog No. GR-311, Biomol, Enzo Life Sciences International).
  • MEF mouse embryo fibroblasts
  • Embryoid body (EB) formation Day 0 to Day 4
  • IPS cells cultured on feeder cells were separated from the plate using trypsin-EDTA (Catalog No. T4049, Sigma-Aldrich Japan), and then centrifuged at 800 rpm for 3 minutes and collected as a pellet. Thereafter, knockout serum replacement (KSR, catalog number 10828, GIBCO, Invitrogen) 15%, non-essential amino acids (catalog number 1681049, MP biomedicals) 1%, 2- Mercaptoethanol (Catalog No. 21985-023, Gibco, Invitrogen) 1%, Penicillin / Streptomycin (Catalog No.
  • Stage 2 Induction of definitive endoderm (DE) (Day 5 to Day 7)
  • the medium containing embryoid bodies was collected from one well and centrifuged at 1,000 rpm for 3 minutes to collect the embryoid bodies as pellets.
  • the embryoid body was then penicillin / streptomycin 1%, L-glutamine 1%, activin A (catalog number 338-AC, R & D Systems) 100 ng / ml, bFGF (catalog number 100-18B, Peprotech) 100 ng Suspended in 12 ml of knockout DMEM to which / ml was added.
  • a gelatin-coated 12-well plate (catalog number 353503, Nippon Becton Dickinson) was prepared, and 10 to 15 embryoid bodies were transferred per well.
  • the cells were further cultured for 3 days. KSR was added at 0.2% on the 6th day and 2% on the 7th day. The medium was changed in half the amount every day.
  • mLN-111 isoform basement membrane (mLN111-sBM) substrate Rat immortalized alveolar type II epithelial cells (SV40-T2 cells: Dr. A. Clement, Hopital Armand Trousseau) , Paris (see Reference 6) alveolar type II epithelial cells (collected from rats transfected with SV40-large T antigen gene) according to the method described in Reference 7 (Catalog No. 356234, By culturing in the presence of Nippon Becton Decktonson, a mLN-111 isoform-type basement membrane (mLN111-sBM) structure was prepared on a 12-well culture plate insert (Catalog No.
  • T3-Alb7-sBM Human immortalized liver parenchymal cells
  • T3-Alb7 cells drug-inducible Cre recombinase expressing immortalized human hepatocyte cell line TTNT-16-T3 (Depositing organization: IPOD, National Institute of Advanced Industrial Science and Technology, Patent Biological Depositary Center, Address: 1-chome, East 1-chome, Tsukuba, Ibaraki, Japan, Chuo 6 (postal code 305-8666), Date of deposit: October 2003 1 day, accession number: FERM BP-08501, Totsugawa T, et al., Survival of liver failure pigs by transplantation of reversibly immortalized human hepatocytes with Tamoxifen-mediated self-recombination.
  • T3-Alb7 cells were treated with a D-PBS ( ⁇ ) solution containing 50 mM NH 3 , 0.1% Triton X-100, and protease inhibitor cocktail (PIC, manufactured by Peptide Laboratories).
  • PIC protease inhibitor cocktail
  • iPS-Heps differentiation into iPS cell-derived hepatocytes (iPS-Heps) (from day 8 to day 16)
  • definitive endoderm was separated from the plate using trypsin-EDTA (catalog number T4049, Sigma-Aldrich Japan), then centrifuged at 800 rpm for 3 minutes and collected as a pellet.
  • Example 1 1.5 ml of medium was added to one well of an insert-compatible 12-well culture plate (catalog number 353503, Nippon Becton Dickinson). This was designated Example 1.
  • TMNT-1 human immortalized hepatic sinusoidal endothelial cell line
  • FBS fetal bovine serum
  • Example 2 Add 1.5 ml of the medium to the well, and place the insert containing the definitive endoderm, mLN111-sBM substrate and 0.5 ml of the medium. This was designated Example 2.
  • HGF-free medium that is, knockout DMEM supplemented with 10% KSR, 1% non-essential amino acid, 1% L-glutamine, and 1% DMSO. Exchanged. Differentiation induction was completed on the 16th day.
  • a greater number of iPS cell-derived hepatocytes were obtained (FIG. 1 (EG)).
  • Example 4 1.5 ml of medium was added to one well of a 12-well culture plate corresponding to insert, and an insert containing definitive endoderm, T3-Alb7-sBM substrate and 0.5 ml of medium was placed on the plate and cultured. On the bottom of the 12-well culture plate for inserts, 5.0 ⁇ 10 5 TMNK-1s are seeded beforehand and cultured for 1 day or more, 1.5 ml of medium is added to the wells, definitive endoderm, T3- An insert containing Alb7-sBM substrate and 0.5 ml of medium was placed and cultured, and this was designated as Example 4.
  • HGF-free medium ie, knockout DMEM supplemented with 1% KSR, 1% non-essential amino acid, 1% L-glutamine, and 1% DMSO, and thereafter the entire medium was replaced every 2 days. Exchanged. Differentiation induction was completed on the 16th day. Compared to Comparative Example 1 in which no basement membrane substrate was used, in Examples 3 and 4, a greater number of iPS cell-derived hepatic parenchymal cells were obtained (FIGS. 2 and 5).
  • Test example 1 Functional Evaluation of iPS Cell-Derived Liver Parenchymal Cells
  • A Ammonia Metabolism Ability According to Undifferentiated iPS Cells, iPS Cell-Derived Liver Parenchymal Cells Differentiated According to the Methods of Example 1 and Example 2, 0.56 mM ammonia sulfate (catalog number 02619-15, Sigma-Aldrich) was added to iPS cell-derived hepatocytes derived from differentiation, and the culture supernatant was collected after 24 hours to measure ammonia metabolic capacity. did. The ammonia concentration was measured using Ammonia Test-Wako (catalog number 277-14401, manufactured by Wako Pure Chemical Industries).
  • the iPS cell-derived hepatic parenchymal cells of the present invention exhibited 133 ⁇ 153 ng urea production ability per well in Example 1, and 167 ⁇ 153 ng urea production ability per well in Example 2. This showed a tendency for the amount of urea production to be large compared to 33.3 ⁇ 57.7 ng of the iPS cell-derived liver parenchymal cells of Comparative Example 1.
  • Example 1 albumin production was not shown, and in Example 2, albumin production of 514 ⁇ 890 pg was observed. Compared to these, albumin production was significantly higher at 2320 ⁇ 64 pg in Example 3 and 2635 ⁇ 111 pg in Example 4.
  • the total number of cells and the number of Alb-expressing cells were measured by immunostaining the cell nuclei with DAPI (catalog number H-1200, manufactured by Vector Laboratories Inc.), and the number of nuclei stained with Alb-positive cells / DAPI.
  • the positive rate of Alb expression was calculated. The results are shown in FIG. In the sample of undifferentiated iPS cells and the control T3-Alb7-sBM substrate alone, the Alb positive rate was 0%.
  • the iPS cell-derived liver parenchymal cells of Comparative Example 1 showed an Alb positive rate of 20.6%. On the other hand, in Example 4, a significantly high Alb positive rate of 31.8% was observed.
  • Comparative Example 1 The same procedures as in Example 1 (1) to (3) were performed to obtain definitive endoderm.
  • iPS-Heps differentiation into iPS cell-derived liver parenchymal cells (Days 8 to 16)
  • the medium was replaced with knockout DMEM supplemented with 10% KSR, 1% non-essential amino acids, 1% L-glutamine, 1% DMSO, 100 ng / ml HGF (Catalog No. 100-39, Peprotech).
  • the medium was changed in half the amount every day. Differentiation induction was completed on the 16th day.
  • Reference example 1 Cell morphology of iPS cell-derived liver parenchymal cells (iPS-Heps) in Comparative Example 1
  • the cell morphology of iPS cell-derived liver parenchymal cells in Comparative Example 1 was observed using a phase contrast microscope and an electron microscope.
  • FIG. 6B phase contrast microscope observation
  • polygonal cells having an abundant circular nucleus and abundant granules in the cytoplasm are clearly observed. It is positive for PAS staining, indicating that intracellular granules are glycogen (FIG. 6C).
  • observation by an electron microscope also shows a glycogen field (FIG. 6E), which is a site where glycogen is gathered, and a luminal formation image with microvilli (FIG. 6F), indicating a cell structure consistent with liver parenchymal cells.
  • FIG. 6E glycogen field
  • FIG. 6F luminal formation image with microvilli
  • Reference example 2 Gene expression in the process of inducing differentiation from iPS cells to liver parenchymal cells RT-PCR was performed at each stage of the process of inducing differentiation in Comparative Example 1.
  • RNA was extracted using TRI zol (catalog number 15596-026, Invitrogen).
  • cDNA was prepared from 2 ⁇ g of RNA using MuLV reverse transcriptase (Catalog No. N8080018, Applied Biosystems) and RNase inhibitor (Catalog No. N808119, Applied Biosystems) using a Takara Thermal Cycler. Created.
  • RT-PCR was performed using AmpliTaq Gold DNA polymerase, GeneAmp PCR Gold Buffer, MgCl 2 solution (Catalog No. 606080, Applied Biosystems).
  • the genes include alphafetoprotein (AFP), albumin (Alb), transferrin (Trf), phosphoenolpyruvate carboxykinase 1 (PCK1), carbamyl phosphate synthetase (CPS), sex-determining gene box 17 (Sox17) and endogenous
  • AFP alphafetoprotein
  • albumin Alb
  • transferrin Trf
  • PCK1 carboxykinase 1
  • CPS carbamyl phosphate synthetase
  • Sox17 sex-determining gene box 17
  • the ⁇ -actin gene as a control was examined.
  • the primers used in RT-PCR are shown in Table 1. PCR products were electrophoresed on 2.5% agarose gel and visualized using ethidium bromide (FIG. 7A).
  • lanes 1 to 4 are iPS cells, EB, DE, and iPS-Heps, respectively.
  • Real-time RT-PCR was performed using the Light Cycler 1.5 (Roche Applied Science) kit using the Light Cycler Fast DNA Master SYBR Green I (Catalog No. 03003230001, Roche Applied Science, IN) kit.
  • the genes include alphafetoprotein (AFP), albumin (Alb), transferrin (Trf), phosphoenolpyruvate carboxykinase 1 (PCK1), carbamyl phosphate synthetase (CPS) and the endogenous control ⁇ -actin gene. Examined. Table 2 shows the primers used in real-time RT-PCR.
  • Reference example 3 The effect of embryoid body formation period on the definitive endoderm differentiation efficiency was investigated.
  • EBs formed over 5 days as in the first stage of Example 1 were subjected to endoderm differentiation induction over 5 days under the same conditions as in the second stage of Example 1 (Sample 1).
  • EBs formed in the same manner as in the first stage of Example 1 except that the formation period was 2 days were subjected to endoderm differentiation induction over 5 days under the same conditions as in the second stage of Example 1 (Sample 2). ).
  • the gene expression of the obtained cells was analyzed by real-time PCR as in Example 2.
  • the genes examined were sex-determining gene box 17 (Sox17), chemokine (C—X—C motif) receptor 4 (chemokine (CXC motif) receptor 4) (Cxcr4) and forkhead box A2 (forkhead box A2) (Foxa2) Table 3 shows the primers used.
  • panendoderm markers Sox17 and Foxa2 are 1.4 and 2.8-fold, respectively, and the expression of definitive endoderm marker Cxcr4 is 1.1-fold.
  • SEQ ID NO: 1 PCR forward primer for detecting AFP gene
  • SEQ ID NO: 2 PCR reverse primer for detecting AFP gene
  • SEQ ID NO: 3 PCR forward primer for detecting Alb gene
  • SEQ ID NO: 4 Alb PCR reverse primer for detecting gene
  • SEQ ID NO: 5 PCR forward primer for detecting Trf gene
  • SEQ ID NO: 6 PCR reverse primer for detecting Trf gene
  • SEQ ID NO: 7 for detecting PCK1 gene
  • SEQ ID NO: 8 PCR reverse primer for detecting PCK1 gene
  • SEQ ID NO: 9 PCR forward primer for detecting CPS gene
  • SEQ ID NO: 10 PCR reverse primer for detecting CPS gene Limer
  • SEQ ID NO: 12 PCR reverse primer for detecting the Sox17 gene
  • SEQ ID NO: 13 PCR forward primer SEQ ID NO: for detecting the ⁇ -actin gene 14: Reverse primer for PCR for detecting

Landscapes

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

Abstract

Provided is a method for inducing differentiation of iPS cells into hepatic parenchymal cells. Said method yields sufficiently functional, safe hepatic parenchymal cells and can produce large quantities. The provided method includes: (a) a step in which iPS cells are cultured in a suspension culture and embryoid bodies are formed; (b) a step in which 10 to 15 embryoid bodies per well are adhered to a gelatin-covered plate and cultured, inducing embryonic endoderm cells; and (c) a step in which HFG and DMSO are added and the embryonic endoderm cells are cultured on a basement membrane substrate, inducing differentiation into hepatic parenchymal cells.

Description

iPS細胞から肝実質細胞への分化誘導方法Method for inducing differentiation from iPS cells to hepatocytes

 本発明は、iPS細胞(人工多能性幹(induced pluripotent stem)細胞)から機能的な肝実質細胞への分化誘導方法およびそれにより分化された肝実質細胞に関する。 The present invention relates to a method for inducing differentiation from iPS cells (induced pluripotent stem cells) to functional hepatocytes and hepatocytes differentiated thereby.

 従来の技術として、胚性幹(embryonic stem:ES)細胞から肝実質細胞を分化誘導する方法についてはいくつかの報告がなされている。 As a conventional technique, several reports have been made on methods for inducing differentiation of hepatocytes from embryonic stem (ES) cells.

 たとえば、ES細胞での肝実質細胞分化誘導に関する既報告では、胚葉体の形成期間を2~2.5日間とするものが主流である(非特許文献1~3)。 For example, in the reports on the induction of hepatocyte differentiation in ES cells, the mainstream is that the embryoid body formation period is 2 to 2.5 days (Non-Patent Documents 1 to 3).

 一方、iPS細胞から肝実質細胞を分化誘導する具体的な技術は現在のところ報告されていない。 On the other hand, no specific technique for inducing differentiation of hepatocytes from iPS cells has been reported at present.

 ES細胞は多能性を有し、三胚葉系のいずれの細胞にも分化しうる。また、ES細胞は、実験系において無限の増殖能を有する。したがって、ES細胞から多数の幹細胞を分化誘導し、バイオ人工肝臓の素材として用いることにより、肝疾患患者の治療が可能となる。しかし、ES細胞由来の肝実質細胞で細胞治療を行う際には、長期的な免疫抑制剤の使用が不可欠であり、感染症などの合併症をきたしうる。またES細胞を得るには受精卵の胚を必要とするため、ES細胞の樹立および運用には倫理的な問題が伴う。 ES cells have pluripotency and can differentiate into any of the three germ layer cells. Moreover, ES cells have infinite proliferation ability in an experimental system. Therefore, it is possible to treat patients with liver diseases by inducing differentiation of many stem cells from ES cells and using them as a material for bioartificial liver. However, when cell therapy is performed with ES cell-derived hepatocytes, long-term use of immunosuppressive agents is indispensable, and complications such as infections may occur. In addition, since embryos of fertilized eggs are required to obtain ES cells, ethical problems are involved in the establishment and operation of ES cells.

 iPS細胞では、受精卵の胚を必要とせず、さらには患者自身の体細胞を使用することが可能であるため、倫理的な問題および拒絶反応の両面において有望な細胞ソースとなり得ると期待されている。 Since iPS cells do not require embryos of fertilized eggs, and because it is possible to use the patient's own somatic cells, it is expected to be a promising cell source for both ethical issues and rejection. Yes.

 しかしながら、iPS細胞の分化誘導に関する研究の現状はまだ充分なものではない。また、iPS細胞の分化誘導においては、培養時の細胞密度と培養液の選択が難しいことや、iPS細胞はES細胞に比し肝実質細胞の前駆組織である胚体内胚葉(definitive endoderm)への分化効率が低いこと、さらには、得られる肝実質細胞の機能が充分でないといった問題がある。 However, the current state of research on iPS cell differentiation induction is not yet sufficient. In addition, in the differentiation induction of iPS cells, it is difficult to select the cell density and culture medium at the time of culturing, and iPS cells can be transferred to definitive endoderm which is a precursor tissue of hepatic parenchymal cells compared to ES cells. There are problems that differentiation efficiency is low, and further, the function of the obtained hepatocytes is not sufficient.

 したがって、iPS細胞から機能的な肝実質細胞を分化誘導する技術の開発が切望されている。 Therefore, development of a technique for inducing differentiation of functional hepatocytes from iPS cells is eagerly desired.

 一方、基底膜は、上皮組織の内側下面である基底面(basal surface)直下に存在する、ラミニン、エンタクチン、IV型コラーゲンなどのタンパク質およびプロテオグリカンなどの細胞外基質(ECM)からなり、細胞を含まない50~100nmの構造体であり、未成熟な上皮細胞が増殖し、成熟した細胞に分化して、本来の形態や、機能を発現するのに必須なものと考えられている。基底膜については、これまで、線維芽細胞を包埋したコラーゲンマトリックス上で上皮細胞を培養する方法や、マトリゲル(Matrigel(登録商標)、ベクトンデッキンソン)を加えたコラーゲン線維基質上で上皮細胞を培養する方法などインビトロでの作製方法が開発され、培養基材としての使用や、人工組織の作製への使用などの例が報告されている(特許文献1~3)。 On the other hand, the basement membrane is composed of proteins such as laminin, entactin, type IV collagen, and extracellular matrix (ECM) such as proteoglycan, which are directly below the basal basal surface, the inner lower surface of epithelial tissue, and contain cells. It is a structure of 50 to 100 nm, and is considered essential for immature epithelial cells to proliferate and differentiate into mature cells to express their original form and function. For the basement membrane, epithelial cells have been cultured on a collagen matrix in which fibroblasts are embedded, or on a collagen fiber substrate to which Matrigel (Matrigel (registered trademark), Becton Dickinson) has been added. In vitro production methods such as a culture method have been developed, and examples of use as a culture substrate and production of artificial tissue have been reported (Patent Documents 1 to 3).

特開2003-93053号公報JP 2003-93053 A 特開2003-169846号公報JP 2003-169846 A 特開2003-93050号公報JP 2003-93050 A 再表2004-085606号公報Table 2004-085606

Basma H, Soto-Gutieerrez A, Yannam GR, Liu L, Ito R, Yamamoto T, Ellis E, Carson SD, Sato S, Chen Y, Muirhead D, Navarro-Alvarez N, Wong RJ, Roy-Chowdhury J, Platt JL, Mercer DF, Miller JD, Strom SC, Kobayashi N, Fox IJ. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology 2009;136:990-999.Basma H, Soto-Gutieerrez A, Yannam GR, Liu L, Ito R, Yamamoto T, Ellis E, Carson SD, Sato S, Chen Y, Muirhead D, Navarro-Alvarez N, Wong RJlatyJL , Mercer DF, Miller JD, Strom SC, Kobayashi N, Fox IJ. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology 2009; 136: 990-999. Rambhatla L, Chiu CP, Kundu P, Peng Y, Carpenter MK. Generation of hepatocyte-like cells from human embryonic stem cells. Cell Transplant 2003;12:1-11.Rambhatla L, Chiu CP, Kundu P, Peng Y, Carpenter MK. Generation of hepatocyte-like cells from human embryonic stem cells. Cell Transplant 2003; 12: 1-11. Soto-Gutieerrez A, Kobayashi N, Rivas-Carrillo JD, Navarro-Alvarez N, Zhao D, Okitsu T, Noguchi H, Basma H, Tabata Y, Chen Y, Tanaka K, Narushima M, Miki A, Ueda T, Jun HS, Yoon JW, Lebkowski J, Tanaka N, Fox IJ. Reversal of mouse hepatic failure using an implanted liver-assist device containing ES cell-derived hepatocytes. Nat Biotechnol 2006;24:1412-1419.Soto-Gutieerrez A, Kobayashi N, Rivas-Carrillo JD, Navarro-Alvarez N, Zhao D, Okitsu T, Noguchi H, Basma H, Tabata Y, Chen Y, Tanaka K, Narushima M, U , Yoon JW, Lebkowski J, Tanaka N, Fox IJ. Reversal of mouse hepatic failure using an implanted liver-assist device containing ES cell-derived hepatocytes. Nat Biotechnol 2006; 24: 1412-1419.

 本発明の課題は、iPS細胞から効率的に機能的に優れた肝実質細胞を分化誘導する方法を提供すること、そして、倫理的問題を引き起こすことなく、バイオ人工肝臓の素材としての機能的肝実質細胞を提供することである。 An object of the present invention is to provide a method for efficiently inducing differentiation of functionally superior liver parenchymal cells from iPS cells, and functional liver as a bioartificial liver material without causing ethical problems. Providing parenchymal cells.

 そこでわれわれは、以下の点を工夫し、組み合わせることによりiPS細胞から肝実質細胞を分化誘導することに成功した。 Therefore, we succeeded in inducing differentiation of hepatocytes from iPS cells by devising and combining the following points.

 本発明は、iPS細胞から肝実質細胞への分化誘導方法であって、
(a)iPS細胞を浮遊培養し、胚様体(embryoid body)を形成する工程、
(b)被覆剤としてゼラチンを使用したプレートへ胚様体を1ウェルあたり10~15個として接着させ培養することにより、胚体内胚葉を誘導する工程、および
(c)HGFおよびDMSOを添加し、基底膜基質上にて胚体内胚葉を培養することにより肝実質細胞分化を誘導する工程
を含む方法に関する。
The present invention is a method for inducing differentiation from iPS cells to hepatocytes,
(A) a step of suspension culture of iPS cells to form an embryoid body;
(B) inducing definitive endoderm by adhering and culturing 10-15 embryoid bodies on a plate using gelatin as a coating agent per well, and (c) adding HGF and DMSO, The present invention relates to a method comprising inducing hepatocyte differentiation by culturing definitive endoderm on a basement membrane substrate.

 本発明の方法において、(a)工程の浮遊培養期間は5日間が好ましい。 In the method of the present invention, the suspension culture period in step (a) is preferably 5 days.

 また、基底膜基質は、肺胞II型上皮細胞または肝実質細胞由来のものが好ましい。 The basement membrane matrix is preferably derived from alveolar type II epithelial cells or liver parenchymal cells.

 本発明の分化誘導方法によれば、機能的に優れた肝実質細胞を得ることができる。また、安定的に胚体内胚葉を得ることができ、効率よく肝実質細胞への分化誘導が可能となる。本発明の分化肝実質細胞は、iPS細胞に基づく肝実質細胞であるため、ES細胞の樹立および運用に伴う倫理的問題を回避することができる。さらに個体由来のiPS細胞を用いることにより、免疫抑制剤が不要な個体特異的肝実質細胞を得ることができる。したがって、本発明のiPS細胞から分化誘導された肝実質細胞は、バイオ人工肝臓の素材としてより好適なものである。 According to the differentiation inducing method of the present invention, functionally superior hepatocytes can be obtained. In addition, definitive endoderm can be stably obtained, and differentiation into hepatocytes can be efficiently induced. Since the differentiated hepatocytes of the present invention are hepatocytes based on iPS cells, ethical problems associated with the establishment and operation of ES cells can be avoided. Furthermore, by using individual-derived iPS cells, individual-specific hepatocytes that do not require immunosuppressive agents can be obtained. Therefore, hepatocytes derived from the iPS cells of the present invention are more suitable as a material for bioartificial liver.

iPS細胞から肝実質細胞への分化過程を示す図である。Aは、日数経過と分化過程を示すスキームである。B~EはそれぞれiPS細胞から肝実質細胞への分化過程における、(B)iPS細胞、(C)胚様体(EB)、(D)胚体内胚葉(DE)、および(E~G)iPS細胞由来肝実質細胞(iPS-Heps)の位相差顕微鏡写真である。(E)は比較例1、(F)は実施例1および(G)は実施例2のiPS-Hepsを示す。It is a figure which shows the differentiation process from an iPS cell to a hepatocyte. A is a scheme showing the passage of days and the differentiation process. B to E are (B) iPS cells, (C) embryoid bodies (EB), (D) definitive endoderm (DE), and (E to G) iPS in the process of differentiation from iPS cells to hepatocytes, respectively. 2 is a phase contrast micrograph of cell-derived hepatocytes (iPS-Heps). (E) shows iPS-Heps in Comparative Example 1, (F) in Example 1 and (G) in Example 2. アルブミンの発現を免疫組織染色により示す顕微鏡像である。実施例4、比較例1、未分化iPS細胞および対照としてT3-Alb7-sBM基質のみの試料を用いた。顕微鏡像Albは、ラビット抗マウスアルブミン抗体(カタログ番号64560、MP biomedicals製)を一次抗体とし、DyLight54標識ヒツジ抗ラビット抗体(カタログ番号STAR36D549、MorphoSys UK製)を二次抗体とし、発現したアルブミン(Alb)を染色したものを示す。顕微鏡像DAPIは、細胞の核をDAPI(カタログ番号H-1200、Vector Laboratories Inc,製)で免疫染色したものを示す。また、MERGEはAlbおよびDAPIの2つの顕微鏡像を重ね合わせたものを示す。It is a microscope image which shows the expression of albumin by immunohistochemical staining. Example 4, Comparative Example 1, undifferentiated iPS cells, and T3-Alb7-sBM substrate-only sample were used as controls. Microscopic image Alb was expressed using rabbit anti-mouse albumin antibody (Cat. No. 64560, MP biomedicals) as the primary antibody and DyLight54-labeled sheep anti-rabbit antibody (Catalog No. STAR36D549, MorphoSys UK) as the secondary antibody. ) Is dyed. The microscopic image DAPI shows a cell nucleus immunostained with DAPI (Catalog No. H-1200, manufactured by Vector Laboratories Inc.). MERGE indicates a superposition of two microscopic images of Alb and DAPI. 本発明のiPS細胞由来肝実質細胞のアンモニア除去率を示すグラフである。It is a graph which shows the ammonia removal rate of the iPS cell origin hepatocyte of this invention. 本発明のiPS細胞由来肝実質細胞の尿素産生量を示すグラフである。It is a graph which shows the urea production amount of the iPS cell origin hepatocyte of this invention. 本発明のiPS細胞由来肝実質細胞のアルブミン産生量を示すグラフである。It is a graph which shows the albumin production amount of the iPS cell origin hepatocyte of this invention. 比較例1によりiPS細胞より分化誘導した肝実質細胞の位相差顕微鏡写真(A~C)および電子顕微鏡写真(D~F)である。位相差顕微鏡観察の強拡大像(図6B)では、類円形の核および細胞質内に豊富な顆粒をもつ多角形の細胞が明瞭に観察される。PAS染色陽性であり、細胞内顆粒がグリコーゲンであることを示す(図6C)。また電子顕微鏡観察ではグリコーゲンの集簇した部位であるグリコーゲン野(図6E)や、微絨毛を伴う管腔形成像(図6F)も認め、肝実質細胞に一致した細胞構造を示している。2 is a phase contrast micrograph (AC) and an electron micrograph (DF) of hepatic parenchymal cells induced to differentiate from iPS cells according to Comparative Example 1. FIG. In the strongly magnified image of the phase contrast microscope observation (FIG. 6B), polygonal cells having an abundant circular nucleus and abundant granules in the cytoplasm are clearly observed. It is positive for PAS staining, indicating that intracellular granules are glycogen (FIG. 6C). In addition, observation by an electron microscope also shows a glycogen field (FIG. 6E), which is a site where glycogen is gathered, and a luminal formation image with microvilli (FIG. 6F), indicating a cell structure consistent with liver parenchymal cells. 比較例1の分化誘導過程の各段階の細胞における各種遺伝子の発現を示す図である。調べた遺伝子は、アルファフェトタンパク質(AFP)、アルブミン(Alb)、トランスフェリン(Trf)、ホスホエノールピルビン酸カルボキシキナーゼ1(PCK1)、カルバミル燐酸シンテターゼ(CPS)、性決定遺伝子ボックス17(Sex determining region Y, Box 17)(Sox17)およびβ-アクチンの遺伝子である。レーン1~4は、それぞれiPS細胞、胚様体、胚体内胚葉およびiPS-Hepsを示す。FIG. 3 is a diagram showing expression of various genes in cells at each stage of the differentiation induction process of Comparative Example 1. The genes examined were alphafetoprotein (AFP), albumin (Alb), transferrin (Trf), phosphoenolpyruvate carboxykinase 1 (PCK1), carbamyl phosphate synthetase (CPS), sex determination gene box 17 (Sex determining region Y , Box 17) (Sox17) and β-actin gene. Lanes 1-4 show iPS cells, embryoid bodies, definitive endoderm and iPS-Heps, respectively. アルファフェトタンパク質(AFP)、アルブミン遺伝子(Alb)、トランスフェリン(Trf)、ホスホエノールピルビン酸カルボキシナーゼ1(PCK1)、カルバミル燐酸シンテターゼ(CPS)の比較例1の各分化過程における相対的mRNAレベルを示すグラフである。The relative mRNA level in each differentiation process of the comparative example 1 of alpha fetoprotein (AFP), albumin gene (Alb), transferrin (Trf), phosphoenolpyruvate carboxylase 1 (PCK1), and carbamyl phosphate synthetase (CPS) is shown. It is a graph.

 本発明は、iPS細胞を機能的肝実質細胞へ分化誘導する方法に関する。 The present invention relates to a method for inducing differentiation of iPS cells into functional hepatocytes.

 本発明のiPS細胞から肝実質細胞を分化誘導する方法は、(a)iPS細胞を浮遊培養し、胚様体を形成する工程、(b)被覆剤としてゼラチンを使用したプレートへ胚様体を1ウェルあたり10~15個として接着させ培養することにより、胚体内胚葉を誘導する工程、および(c)HGFおよびDMSOを添加し、基底膜基質上にて胚体内胚葉を培養することにより肝実質細胞分化を誘導する工程を含む。 The method of inducing differentiation of hepatocytes from the iPS cells of the present invention comprises: (a) a step of suspension-culturing iPS cells to form embryoid bodies; (b) transferring embryoid bodies to a plate using gelatin as a coating agent. A step of inducing definitive endoderm by adhering and culturing as 10-15 per well, and (c) hepatic parenchyma by adding HGF and DMSO and culturing definitive endoderm on a basement membrane substrate Inducing cell differentiation.

 本明細書において、「1ウェルあたり」と記載する場合、12ウェル培養プレートの1ウェルを指し、1ウェルの底面積は3.6cm2を意味する。 In the present specification, when “per well” is described, it means one well of a 12-well culture plate, and the bottom area of one well means 3.6 cm 2 .

 次に、本発明の分化誘導方法の一実施態様を示す。 Next, an embodiment of the differentiation induction method of the present invention is shown.

1) iPS細胞を好ましくは5日間浮遊培養し、胚様体を形成した。ES細胞での肝実質細胞分化誘導に関する既報告では、胚葉体の形成期間を2~2.5日間とするものが主流である。しかしiPS細胞はES細胞に比し肝実質細胞の前駆組織である胚体内胚葉への分化効率が低く、2~2.5日間の胚葉体形成では、充分な胚体内胚葉が得られない傾向がある。そこで胚様体の形成には5日間浮遊培養することが、胚体内胚葉への分化効率を高めるうえで好ましい。 1) The iPS cells were preferably suspended in culture for 5 days to form embryoid bodies. In the previous reports on induction of hepatocyte differentiation in ES cells, the mainstream is that the embryoid body formation period is 2 to 2.5 days. However, iPS cells have a lower efficiency of differentiation into definitive endoderm, which is a precursor tissue of hepatic parenchymal cells, compared to ES cells, and there is a tendency that sufficient definitive endoderm cannot be obtained in the case of definitive body formation for 2 to 2.5 days. is there. Therefore, for the formation of embryoid bodies, suspension culture for 5 days is preferable in order to increase the efficiency of differentiation into definitive endoderm.

2) 胚様体をゼラチン被覆プレートに移したのち、アクチビンA 100ng/mlおよび塩基性線維芽細胞増殖因子(bFGF)100ng/ml、低濃度の血清代替物(serum replacement)を添加のうえ3日間培養し、胚体内胚葉を誘導した。5日間かけて形成した胚様体は、2~2.5日かけて形成したものに比し、プレートへの接着能が低下しているため、被覆剤なしではプレートへ接着し分化を開始することができない。そこでわれわれはゼラチンを被覆剤として使用することで良好な接着性を得た。 2) After transferring the embryoid body to a gelatin-coated plate, add activin A 100 ng / ml and basic fibroblast growth factor (bFGF) 100 ng / ml, low concentration serum replacement (serumsereplacement) for 3 days Cultured and induced definitive endoderm. Embryoid bodies formed over 5 days have a lower ability to adhere to the plate than those formed over 2 to 2.5 days, and thus adhere to the plate and start to differentiate without a coating. I can't. Therefore, we obtained good adhesion by using gelatin as a coating agent.

3) 胚様体の培養は12ウェルのゼラチン被覆プレートで行い、1ウェルあたりの胚様体は10~15個として、胚体内胚葉の誘導を開始した。分化誘導過程において、胚様体周囲より細胞増殖が開始し、これらが肝実質細胞へと分化するため、胚様体を適切な密度でプレートに移すことが重要である。胚様体の密度が低い場合(たとえば1ウェルあたり10個より少ない場合)、分化誘導の終了時点で得られる肝実質細胞が少なくなる。一方、胚様体の密度が高い場合(たとえば1ウェルあたり15個より多い場合)には、心筋細胞への分化が誘導されやすくなるため、得られる肝実質細胞の純度が低下する。また培地不足による細胞の死滅をきたしやすい。したがって1ウェルあたりの胚様体を10~15個として胚体内胚葉の誘導を開始することが重要であった。 3) Embryoid bodies were cultured in 12-well gelatin-coated plates, and 10 to 15 embryoid bodies per well were induced to start definitive endoderm induction. In the differentiation induction process, cell growth starts from around the embryoid body and these differentiate into hepatocytes, so it is important to transfer the embryoid body to the plate at an appropriate density. When the density of embryoid bodies is low (for example, less than 10 per well), fewer hepatocytes are obtained at the end of differentiation induction. On the other hand, when the density of embryoid bodies is high (for example, when there are more than 15 per well), differentiation into cardiomyocytes is likely to be induced, so that the purity of the obtained hepatocytes decreases. In addition, cells tend to be killed due to lack of medium. Therefore, it was important to start definitive endoderm induction with 10 to 15 embryoid bodies per well.

4) 一連の分化誘導過程において、無血清培地、すなわちノックアウトDMEMおよびノックアウト血清代替物(KSR)を使用した。従来、ES細胞の分化誘導過程では血清培地が用いられてきたが、iPS細胞の培養および分化誘導においては、無血清培地の方が血清培地に比し高い細胞増殖能を示したため、肝実質細胞の分化誘導において上記の組み合わせを用い、肝実質細胞増殖因子(HGF)とジメチルスルホキシド(DMSO)を添加し、基底膜基質上にて胚体内胚葉を培養することにより、アンモニアを代謝し、尿素を産生することができる機能的な肝実質細胞の分化誘導に成功した。 4) In a series of differentiation induction processes, a serum-free medium, that is, knockout DMEM and knockout serum substitute (KSR) was used. Conventionally, a serum medium has been used in the differentiation induction process of ES cells. However, in the culture and differentiation induction of iPS cells, the serum-free medium showed higher cell proliferation ability than the serum medium. In the differentiation induction, hepatocyte growth factor (HGF) and dimethyl sulfoxide (DMSO) are added, and the definitive endoderm is cultured on the basement membrane substrate, thereby metabolizing ammonia and urea. We succeeded in inducing differentiation of functional hepatocytes that can be produced.

 以上、1)iPS細胞を好ましくは5日間浮遊培養し、胚様体を形成する、2)ゼラチンを被覆剤として使用し、胚様体をプレートへ接着させる、3)1ウェルあたりの胚様体は10~15個として胚体内胚葉の誘導を開始する、4)好ましくは無血清培地、すなわちノックアウトDMEMおよびKSRを使用し、HGFとDMSOを添加し、基底膜基質上にて胚体内胚葉を培養することにより肝実質細胞への分化を始める、上記1)~4)の工程、およびその組み合わせが本願発明の特徴である。 1) Suspension culture of iPS cells preferably for 5 days to form embryoid bodies 2) Use gelatin as a coating agent to adhere embryoid bodies to plates 3) Embryoid bodies per well Initiate definitive endoderm induction as 10-15, 4) preferably using serum-free medium, ie knockout DMEM and KSR, add HGF and DMSO, and culture definitive endoderm on basement membrane substrate Thus, the above-described steps 1) to 4), which start differentiation into hepatocytes, and combinations thereof are the features of the present invention.

 本発明により、効果的かつ安定的にiPS細胞から肝実質細胞を誘導することができる。 According to the present invention, hepatocytes can be induced from iPS cells effectively and stably.

 iPS細胞としては、哺乳類由来のものが好ましく、哺乳類としては、ヒト、カニクイザルなどの霊長類、マウスなどが挙げられる。 IPS cells are preferably derived from mammals, and examples of mammals include humans, primates such as cynomolgus monkeys, and mice.

 iPS細胞の作製方法としては、Okita K, Ichisaka T, Yamanaka S., Generation of germline-competent induced pluripotent stem cells. Nature 2007; 448: 313-317 などを参照することができる。また、たとえばマウスiPS細胞は、理研細胞バンクより入手可能である。 As a method for producing iPS cells, OkitaOK, Ichisaka T, Yamanaka S., Generation of germline-competent induced-pluripotent stem cells. Nature 2007; 448: 313-317 can be referred to. For example, mouse iPS cells are available from the RIKEN Cell Bank.

 本発明においては、必要に応じて、iPS細胞を分化しない条件下に継代、維持、増殖させ(以下、この工程を調製工程という場合がある)、必要に応じてその一部を使用して肝実質細胞に分化誘導する。 In the present invention, if necessary, iPS cells are passaged, maintained and proliferated under non-differentiating conditions (hereinafter, this step may be referred to as a preparation step), and a part thereof is used as necessary. Induces differentiation into hepatocytes.

 調製工程において、iPS細胞の培養を行う容器としては、増殖能の点からフィーダー細胞でコートされたものを使用するのが好ましい。フィーダー細胞としては、γ線で照射処理したマウス胚性繊維芽細胞が挙げられる。マウス由来のiPS細胞の場合、フィーダー細胞でコートした培養容器を使用することが好ましい。 In the preparation step, it is preferable to use a container coated with feeder cells as a container for culturing iPS cells from the viewpoint of proliferation ability. Examples of feeder cells include mouse embryonic fibroblasts irradiated with γ rays. In the case of mouse-derived iPS cells, it is preferable to use a culture vessel coated with feeder cells.

 調製工程における培養条件としては、通常のES細胞と同様の条件を採用することができる。調製工程の要点は、24時間おきに培地を交換して栄養の枯渇を防ぐこと、細胞密度が増加しコロニー同士が接着する前に継代することなどが挙げられる。培養温度は、36~37℃の範囲が好ましく、pHは7.3~7.4の範囲が好ましい。マウスiPS細胞の調製工程では、一般に分化を抑制するために白血病抑制因子(LIF)を使用する。 As the culture conditions in the preparation step, the same conditions as for normal ES cells can be employed. The main points of the preparation process include changing the medium every 24 hours to prevent nutrient depletion, and subculturing before the cell density increases and the colonies adhere to each other. The culture temperature is preferably in the range of 36 to 37 ° C., and the pH is preferably in the range of 7.3 to 7.4. In the process of preparing mouse iPS cells, leukemia inhibitory factor (LIF) is generally used to suppress differentiation.

 本発明の方法によりiPS細胞より分化誘導された肝実質細胞は、形態学的特徴の観察および逆転写ポリメラ-ゼ連鎖反応(RT-PCR)で確認できる。 The hepatic parenchymal cells induced to differentiate from iPS cells by the method of the present invention can be confirmed by observation of morphological characteristics and reverse transcription polymerase chain reaction (RT-PCR).

 形態学的な確認方法としては、位相差型顕微鏡で観察される複数の核を有する細胞および電子顕微鏡で観察される細胞質内に豊富な顆粒、特にグリコーゲン顆粒の存在といった、肝実質細胞に特異的な形態学的特徴を確認することが挙げられる。 The morphological confirmation method is specific to liver parenchymal cells, such as cells with multiple nuclei observed with a phase contrast microscope and abundant granules in the cytoplasm observed with an electron microscope, especially glycogen granules. Specific morphological features.

 また、RT-PCRにより、誘導された肝実質細胞の形質発現を遺伝子の発現から評価することもできる。アルブミン遺伝子が発現していることと、未分化な遺伝子であるα-フェトプロテインが発現していないことがRT-PCRなどで確認できれば、分化が誘導されているということができる。また、肝実質細胞としての成熟度を表わす指標として、アルブミン、尿素合成に関する酵素のカルバミルリン酸シンテターゼ(CPS)、糖新生を調節する酵素のホスホエノールピルビン酸カルボキシキナーゼ1(PCK1)、トランスフェリン(Trf)などの遺伝子発現が認められるのが好ましい。 In addition, the expression of induced hepatic parenchymal cells can be evaluated from the expression of the gene by RT-PCR. If it can be confirmed by RT-PCR or the like that the albumin gene is expressed and that the undifferentiated α-fetoprotein is not expressed, it can be said that differentiation is induced. In addition, as an index indicating the maturity of hepatocytes, albumin, carbamyl phosphate synthetase (CPS), an enzyme related to urea synthesis, phosphoenolpyruvate carboxykinase 1 (PCK1), transferrin (Trf), an enzyme that regulates gluconeogenesis It is preferable that gene expression such as is observed.

 さらには、実際に肝実質細胞として機能できるか否かは、培地中にアンモニア、リドカイン、ジアゼパムなどを添加して代謝できるか否かが大きな点となる。また、培地中にアルブミン、尿素が産生されていれば、充分に機能的な肝実質細胞へと分化しているということができる。 Furthermore, whether or not it can actually function as a hepatocyte is largely determined by whether it can be metabolized by adding ammonia, lidocaine, diazepam, or the like to the medium. Moreover, if albumin and urea are produced in the medium, it can be said that they have differentiated into sufficiently functional hepatocytes.

 本発明に用いる基底膜基質としては、様々なものを使用することができ、特に限定されるものではない。たとえば得られた肝実質細胞の機能が充分に確認されたという点から、特開2003-93053号公報などに記載のマトリゲル存在下に基底膜形成能を有する細胞を培養することにより得られる基底膜基質や、特開2003-93050号公報などに記載の糖鎖β-GlcNAcを結合したMASTポリマー(maleic anhydride-styrene copolymer)をコートしたコラーゲン線維上に基底膜形成能を有する細胞を培養することにより得られる基底膜基質が好ましい。ここで、マトリゲルとは、Engelbreth-Holm-Swarm腫瘍マトリックスから抽出された基底膜調製物であり(J. Exp. Med. 145, 204-220, 1977)、細胞外基質合成に影響を及ぼす可能性のある種々のサイトカインの他に、ラミニン-111、エンタクチン、IV型コラーゲン、パールカンなどを含んでいる(Exp. Cell Res. 202, 1-8, 1992)。 As the basement membrane substrate used in the present invention, various substrates can be used and are not particularly limited. For example, the basement membrane obtained by culturing cells having the ability to form basement membranes in the presence of Matrigel described in JP-A-2003-93053, etc., from the point that the function of the obtained hepatic parenchymal cells has been sufficiently confirmed. By culturing cells having basement membrane-forming ability on collagen fibers coated with a MAST polymer (maleic 結合 anhydride-styrene copolymer) bound to a substrate or sugar chain β-GlcNAc described in JP-A-2003-93050, etc. The resulting basement membrane substrate is preferred. Matrigel is a basement membrane preparation extracted from the Engelbreth-Holm-Swarm tumor matrix (J. Exp. Med. 145, 204-220, 1977) and may affect extracellular matrix synthesis. In addition to various cytokines, laminin-111, entactin, type IV collagen, perlecan and the like are included (Exp. Cell Res. 202, 1-8, 1992).

 基底膜形成能を有する細胞としては、上皮細胞、血管内皮細胞、グリアなど種々のものを使用することができる。肝臓への分化誘導という点からは、例えば、肺胞II型上皮細胞などの肺由来の細胞や、肝実質細胞などの肝臓由来の細胞、あるいは、293細胞を遺伝子改変してhLN-511を安定発現し、細胞外に大量分泌するrLN-10細胞を用いることが好ましく、さらには肝実質細胞が最も好ましい。安定した質の基底膜基質が大量に得られることから株化した細胞(癌化および不死化細胞株)を使用することが好ましく、また安全面を鑑みると不死化した細胞系を使用することがより好ましい。 Various cells such as epithelial cells, vascular endothelial cells, and glia can be used as cells having the ability to form a basement membrane. From the viewpoint of induction of differentiation into the liver, for example, cells derived from lung such as alveolar type II epithelial cells, cells derived from liver such as liver parenchymal cells, or 293 cells are genetically modified to stabilize hLN-511. It is preferable to use rLN-10 cells that are expressed and secreted in large quantities extracellularly, and most preferably hepatocytes. It is preferable to use established cells (cancerous and immortalized cell lines) because a large amount of stable quality basement membrane substrate is obtained, and in view of safety, it is preferable to use immortalized cell lines. More preferred.

 基底膜の主要糖タンパク質であるラミニンは、α、β、γの3種類のサブユニットからなる複合体で、種々のアイソフォーム、ラミニン-111(α1β1γ1)やラミニン-511(α5β1γ1)などが存在する。 Laminin, the main glycoprotein of the basement membrane, is a complex composed of three types of α, β, and γ subunits, and there are various isoforms such as laminin-111 (α1β1γ1) and laminin-511 (α5β1γ1). .

 このような基底膜基質の作製方法としては、たとえば、特開2003-93050号公報または特開2003-93053号公報などに記載の技術を使用することができる。 As a method for producing such a basement membrane substrate, for example, the technique described in Japanese Patent Application Laid-Open No. 2003-93050 or Japanese Patent Application Laid-Open No. 2003-93053 can be used.

 また、基底膜基質上で胚体内胚葉と共に各種の細胞を共培養することで、得られる肝実質細胞の機能を向上させることができる。共培養には、肝類洞内皮細胞や胆管上皮細胞、肝星細胞などを用いることができるが、ヒト肝類洞内皮細胞株などが望ましい。 Also, by co-culturing various cells together with definitive endoderm on the basement membrane matrix, the function of the obtained hepatic parenchymal cells can be improved. For co-culture, hepatic sinusoidal endothelial cells, bile duct epithelial cells, hepatic stellate cells, and the like can be used, and human hepatic sinusoidal endothelial cell lines are preferred.

 よって、本発明の好ましい一実施態様は、iPS細胞から肝実質細胞への分化誘導方法であって、(a)iPS細胞を浮遊培養し、胚様体を形成する工程、(b)被覆剤としてゼラチンを使用したプレートへ胚様体を1ウェルあたり10~15個として接着させ培養することにより、胚体内胚葉を誘導する工程、および(c)HGFおよびDMSOを添加し、基底膜基質上にて胚体内胚葉を肝類洞内皮細胞と共培養することにより肝実質細胞分化を誘導する工程を含む方法に関する。 Therefore, a preferred embodiment of the present invention is a method for inducing differentiation from iPS cells to hepatocytes, wherein (a) the iPS cells are cultured in suspension to form embryoid bodies, (b) as a coating agent A step of inducing definitive endoderm by attaching 10-15 embryoid bodies to a plate using gelatin and culturing; and (c) adding HGF and DMSO on a basement membrane substrate The present invention relates to a method comprising a step of inducing hepatocyte differentiation by co-culturing definitive endoderm with hepatic sinusoidal endothelial cells.

 本発明においては、iPS細胞を胚体内胚葉に分化させ必要細胞数までプールしたのち、必要に応じて肝実質細胞に分化させることにより、倫理的問題を生ずることなく、安全な肝実質細胞が大量に供給できる。これをバイオ人工肝臓の細胞源とすることで万人が恩恵を受けることができるバイオ人工肝臓の開発が大いに期待できる。 In the present invention, iPS cells are differentiated into definitive endoderm, pooled to the required number of cells, and then differentiated into hepatocytes as necessary, so that a large number of safe hepatocytes can be produced without causing ethical problems. Can supply. The development of a bioartificial liver that can benefit everyone by using this as the cell source for bioartificial liver is greatly expected.

 よって、本発明のもう1つの態様は、iPS細胞から分化誘導させた肝実質細胞の用途に関する。これらの肝実質細胞は、バイオ人工肝臓の細胞源に使用することができる。 Therefore, another aspect of the present invention relates to the use of liver parenchymal cells induced to differentiate from iPS cells. These hepatocytes can be used as a cell source for bioartificial liver.

 バイオ人工肝臓とは、肝実質細胞を担体に組み込んで固定しリアクター化した生体内の肝臓を模倣した人工肝臓装置であり、患者の血液を当該装置内に導き、肝実質細胞の代謝能を利用して血液中のトキシンの除去と肝臓細胞に由来する凝固因子などの生理活性物質の供給とを行うことができる装置である。 A bioartificial liver is an artificial liver device that mimics the liver in the living body, in which liver parenchymal cells are incorporated into a carrier, fixed, and made into a reactor. The patient's blood is guided into the device, and the metabolic capacity of liver parenchymal cells is used. Thus, the apparatus can remove toxins in blood and supply physiologically active substances such as coagulation factors derived from liver cells.

 このようなバイオ人工肝臓としては、中空糸型のリアクター(デバイス)と分離・培養細胞を組み合わせたバイブリッド型の人工肝臓などが挙げられる。バイオ人工肝臓は、体外に装着して血管に接続するもの、体内に留置して血管に接続するもの、または血管に接続せずに腹腔内に留置するものの三つの形態がある。本発明の肝実質細胞は、いずれの形態のバイオ人工肝臓にも使用可能であるが、細胞移入などに伴う危険性を回避するという点から体外型であることが好ましい。 Examples of such a bioartificial liver include a hybrid type artificial liver combining a hollow fiber type reactor (device) and separated / cultured cells. There are three types of bioartificial livers: those that are attached outside the body and connected to blood vessels, those that are placed in the body and connected to blood vessels, and those that are placed in the abdominal cavity without being connected to blood vessels. The hepatocytes of the present invention can be used in any form of bioartificial liver, but are preferably in vitro from the viewpoint of avoiding the risks associated with cell transfer.

 バイオ人工肝臓に用いられるバイオリアクターとしては、サーシ バイオメディカル社(Circe Biomedical Inc.)(レキシントン、マサチューセッツ州、米国)の支援下でシダーズサイナイ医療センター(Cedars-Sinai Medical Center)(ロサンジェルス、カリフォルニア州、米国)のディメトリュー(Demetriou)らを中心としたブタ肝実質細胞を用いたバイオ人工肝臓治療用のヘパトアシスト(HepatAssist)(Hui T, Rozga J, Demetriou AA. J Hepatobiliary Pancreat Surg 2001; 8: 1-15.)や、ブタ肝実質細胞を使用したドイツのGerlachらのMELS(モジュラー体外肝臓システム(Modular Extracorporeal Liver System))など、様々なタイプが知られている。これらのリアクターは、肝実質細胞が付着するための足場が無いため、細胞はただ単に、中空糸内スペースか、中空糸外スペースに充填されるのみで浮遊した状態となる傾向がある。肝実質細胞は、浮遊状態では、分化機能が充分に発現されない傾向があり、さらに周りの細胞と衝突し、ストレス刺激を受けやすい。したがって、肝実質細胞に足場が提供できるよう、中空糸と不織布からなるリアクターがより好ましく使用されている(小林直哉、他「ハイブリッド人工肝臓」、肝胆膵、2003年、46巻、p381-393)。 Bioreactors used in bioartificial livers include Cedars-Sinai Medical Center (Los Angeles, California, USA) with the support of Circe Biomedical Inc. (Lexington, Massachusetts, USA). HepatAssist (Hui T, と し た Rozga J, Demetriou AA. J Hepatobiliary Pancreat Surg 2001; 8: 1-15) for the treatment of bioartificial liver using porcine liver parenchymal cells centered on Demetriou et al. .) And German MERLach et al.'S MELS (Modular Extracorporporeal Liver System) using porcine liver parenchymal cells are known. Since these reactors do not have a scaffold for adhering hepatocytes, the cells tend to be in a floating state simply by filling the space inside the hollow fiber or the space outside the hollow fiber. Liver parenchymal cells tend to have insufficient differentiation function in a floating state, and collide with surrounding cells and are susceptible to stress stimulation. Therefore, a reactor composed of a hollow fiber and a non-woven fabric is more preferably used so as to provide a scaffold for hepatocytes (Naoya Kobayashi, et al., “Hybrid Artificial Liver”, Hepatobiliary Pancreas, 2003, 46, p381-393) .

 さらに、本発明の方法によりiPS細胞から分化誘導された肝実質細胞、特にヒト肝実質細胞は、薬剤の代謝試験用細胞として有用である。すなわち、新薬開発や安全性試験、毒性試験などにおける試験用細胞として有用である。 Furthermore, hepatocytes, particularly human hepatocytes, induced to differentiate from iPS cells by the method of the present invention are useful as cells for drug metabolism tests. That is, it is useful as a test cell in new drug development, safety test, toxicity test and the like.

 肝実質細胞は様々な薬物代謝の中心となっており、薬物の毒性や安全性を検定するためには、肝実質細胞に薬物を投与して代謝のされ方を調べることが必要である。そして、物質代謝においては、ヒトと実験動物とでは大きな違いがあるので、最終的なテストはヒト肝実質細胞を使用した試験が不可欠である。この点において、均一な品質のヒト肝実質細胞を大量に提供できる本発明は有用である。このような代謝試験に用いる装置としては、前記のバイオ人工肝臓と同様な構造のものなどが使用できる。 Liver parenchymal cells are the center of various drug metabolisms, and in order to test the toxicity and safety of drugs, it is necessary to administer drugs to hepatic parenchymal cells and examine how they are metabolized. And since there is a big difference between humans and experimental animals in substance metabolism, the final test is indispensable using human hepatocytes. In this respect, the present invention that can provide a large amount of human hepatocytes of uniform quality is useful. As a device used for such a metabolic test, a device having the same structure as the bioartificial liver can be used.

 さらに、本発明の方法によりiPS細胞から分化誘導された肝実質細胞、特にヒト肝実質細胞は、生理活性物質の産生に使用できる。このような生理活性物質としては、アルブミン、各種血液凝固因子などが挙げられる。このような生理活性物質の製造装置としては、前記のバイオ人工肝臓と同様な構造のものなどが使用できる。 Furthermore, hepatocytes, particularly human hepatocytes, induced to differentiate from iPS cells by the method of the present invention can be used for the production of physiologically active substances. Examples of such physiologically active substances include albumin and various blood coagulation factors. As a device for producing such a physiologically active substance, one having the same structure as that of the bioartificial liver can be used.

 以下、本発明をマウス由来のiPS細胞を用いた実施例をあげて説明するが、本発明はこれらに限定されるものではない。 Hereinafter, the present invention will be described with reference to examples using mouse-derived iPS cells, but the present invention is not limited thereto.

実施例1~4
iPS細胞から肝実質細胞への分化誘導
(1) 未分化iPS細胞の培養
 文部科学省ナショナルバイオリソースプロジェクトを介して理研バイオリソースセンターより入手したマウスiPS細胞(Cell no. APS0001, Cell name iPS-MEF-Ng-20D-17, Lot no. 006)を用いた。マウスiPS細胞は、マイトマイシンC(カタログ番号GR-311、バイオモル(Biomol)、エンゾライフサイエンスインターナショナル(Enzo Life Sciences International))により不活化したマウス胚線維芽細胞(MEF)(大日本住友製薬)をフィーダー細胞とし、ゼラチン(カタログ番号ES-006-B、スペシャルティメディア(Specialtymedia)、ケミコンインターナショナル(Chemicon international))被覆プレート上にて37℃、CO2濃度5%の条件下で培養した。培地はComplete ES cell medium(カタログ番号SF001-500P、ミリポア(Millipore)、ケミコンインターナショナル)を用い、白血病抑制因子(recombinant leukemia inhibitory factor:LIF、カタログ番号LIF2010、ミリポア、ケミコンインターナショナル)1000U/mlを添加した。継代数5~20のマウスiPS細胞(図1(B))を分化誘導実験に使用した。
Examples 1 to 4
Induction of differentiation from iPS cells to hepatocytes (1) Cultivation of undifferentiated iPS cells Mouse iPS cells (Cell no. APS0001, Cell name iPS-MEF-Ng) obtained from the RIKEN BioResource Center through the Ministry of Education, Culture, Sports, Science and Technology's National BioResource Project -20D-17, Lot no. 006). Mouse iPS cells are feeders of mouse embryo fibroblasts (MEF) (Dainippon Sumitomo Pharma Co., Ltd.) inactivated by mitomycin C (Catalog No. GR-311, Biomol, Enzo Life Sciences International). Cells were cultured on gelatin (catalog number ES-006-B, Specialty media, Chemicon international) coated plates at 37 ° C. with a CO 2 concentration of 5%. The medium was Complete ES cell medium (catalog number SF001-500P, Millipore, Chemicon International), and leukemia inhibitory factor (LIF, catalog number LIF2010, Millipore, Chemicon International) 1000 U / ml was added. . Mouse iPS cells with passage number 5-20 (FIG. 1 (B)) were used for differentiation induction experiments.

(2) 第1段階:胚様体(EB)の形成(第0日~第4日)
 フィーダー細胞上で培養したiPS細胞を、トリプシン-EDTA(カタログ番号T4049、シグマ-アルドリッチ(Sigma-Aldrich) ジャパン)を用いてプレートより分離したのち、800rpmで3分間遠心分離しペレットとして収集した。その後ノックアウト血清代替因子(Knockout serum replacement:KSR、カタログ番号10828、ギブコ(GIBCO)、インビトロジェン(Invitrogen))15%、非必須アミノ酸(カタログ番号1681049、MP バイオメディカルズ(biomedicals))1%、2-メルカプトエタノール(カタログ番号21985-023、ギブコ、インビトロジェン)1%、ペニシリン/ストレプトマイシン(カタログ番号ES-101-B、スペシャルティメディア、ケミコンインターナショナル)1%、L-グルタミン(カタログ番号DSM202、DSファーマバイオメディカル(DS pharma biomedical))1%を添加したノックアウトDMEM(カタログ番号10829、ギブコ、インビトロジェン)にて懸濁した。この細胞懸濁液を低接着性の6ウェルプレート(カタログ番号3471、コスター(Costar)、コーニング(Corning))へ移し、2×105cells/2ml/ウェルの密度で浮遊培養し、胚様体の形成を開始した。培地は毎日半分の量を交換した。
(2) Stage 1: Embryoid body (EB) formation (Day 0 to Day 4)
IPS cells cultured on feeder cells were separated from the plate using trypsin-EDTA (Catalog No. T4049, Sigma-Aldrich Japan), and then centrifuged at 800 rpm for 3 minutes and collected as a pellet. Thereafter, knockout serum replacement (KSR, catalog number 10828, GIBCO, Invitrogen) 15%, non-essential amino acids (catalog number 1681049, MP biomedicals) 1%, 2- Mercaptoethanol (Catalog No. 21985-023, Gibco, Invitrogen) 1%, Penicillin / Streptomycin (Catalog No. ES-101-B, Specialty Media, Chemicon International) 1%, L-Glutamine (Catalog No. DSM202, DS Pharma Biomedical ( DS pharma biomedical)) was suspended in a knockout DMEM (catalog number 10829, Gibco, Invitrogen) supplemented with 1%. This cell suspension was transferred to a low-adhesion 6-well plate (Catalog No. 3471, Costar, Corning), suspended in suspension at a density of 2 × 10 5 cells / 2 ml / well, and embryoid bodies Began to form. The medium was changed in half the amount every day.

 胚様体の形成開始後、細胞が集合し始め、1日後には球形の細胞集塊を形成した。これらの細胞集塊はその後もさらに結合し、5日後には地図状の形態をなす集塊となった(図1(C))。 After the start of embryoid body formation, the cells began to aggregate, and a day later, spherical cell clusters were formed. These cell agglomerates were further combined, and after 5 days, they became a cluster-like agglomeration (FIG. 1 (C)).

(3)第2段階:胚体内胚葉(DE)の誘導(第5日~第7日)
 第5日に胚様体を含む培地を1ウェルから回収し、1,000rpmで3分間遠心分離しペレットとして胚様体を収集した。胚様体をその後、ペニシリン/ストレプトマイシン 1%、L-グルタミン 1%、アクチビンA(カタログ番号338-AC、R&Dシステムズ)100ng/ml、bFGF(カタログ番号100-18B、ぺプロテック(Peprotech))100ng/mlを添加したノックアウトDMEM 12mlに懸濁した。その後、ゼラチン被覆した12ウェルプレート(カタログ番号353503、日本ベクトン・ディッキンソン)を用意し、1ウェルにつき胚様体を10~15個ずつ移した。胚体内胚葉の形成のため、さらに3日間培養した。KSRは第6日に0.2%、第7日に2%となるよう添加した。培地は毎日半分の量を交換した。
(3) Stage 2: Induction of definitive endoderm (DE) (Day 5 to Day 7)
On day 5, the medium containing embryoid bodies was collected from one well and centrifuged at 1,000 rpm for 3 minutes to collect the embryoid bodies as pellets. The embryoid body was then penicillin / streptomycin 1%, L-glutamine 1%, activin A (catalog number 338-AC, R & D Systems) 100 ng / ml, bFGF (catalog number 100-18B, Peprotech) 100 ng Suspended in 12 ml of knockout DMEM to which / ml was added. Thereafter, a gelatin-coated 12-well plate (catalog number 353503, Nippon Becton Dickinson) was prepared, and 10 to 15 embryoid bodies were transferred per well. In order to form definitive endoderm, the cells were further cultured for 3 days. KSR was added at 0.2% on the 6th day and 2% on the 7th day. The medium was changed in half the amount every day.

 ゼラチン被覆プレートに移された後、胚様体はプレートに接着し、胚様体の周囲より細胞増殖が開始された(図1(D))。 After being transferred to the gelatin-coated plate, the embryoid body adhered to the plate, and cell growth started from around the embryoid body (FIG. 1 (D)).

(4)基底膜基質の作製
a)mLN-111アイソフォーム型基底膜(mLN111-sBM)基質の作製
 ラット不死化肺胞II型上皮細胞(SV40-T2細胞:Dr. A. Clement, Hopital Armand Trousseau, Paris (参考文献6参照)から供与された肺胞II型上皮細胞(SV40-ラージT抗原遺伝子をトランスフェクトしたラットから採取))を参考文献7記載の方法に準じてマトリゲル(カタログ番号356234、日本ベクトン・デッキンソン)の存在下で培養することにより、12ウェル培養プレート用インサート(カタログ番号353494、日本ベクトン・デッキンソン)上にmLN-111アイソフォーム型基底膜(mLN111-sBM)構造体を作製し、タンパク質分解酵素阻害剤の混合液(PIC、ペプチド研究所社製)を添加した等張のリン酸緩衝液(pH7.2;PBS(-))中で、0.1%のトリトンX-100(界面活性剤)2mlを用いて、上皮細胞の脂質成分を溶解・溶出すると同時に、共存する50mM NH3で、基底膜構造体表面に残存するタンパク質を溶解する操作を2回(基底膜構造体のタンパク質までは溶かしてはならない)繰り返した後、再度PICを含むPBS(-)溶液で基底膜構造体から界面活性剤とアルカリとを洗浄することによって、肺胞II型上皮細胞層を剥離して基底膜構造体が露出したmLN-111アイソフォーム型基底膜(mLN111-sBM)基質を作製した。
(4) Preparation of basement membrane substrate a) Preparation of mLN-111 isoform basement membrane (mLN111-sBM) substrate Rat immortalized alveolar type II epithelial cells (SV40-T2 cells: Dr. A. Clement, Hopital Armand Trousseau) , Paris (see Reference 6) alveolar type II epithelial cells (collected from rats transfected with SV40-large T antigen gene) according to the method described in Reference 7 (Catalog No. 356234, By culturing in the presence of Nippon Becton Decktonson, a mLN-111 isoform-type basement membrane (mLN111-sBM) structure was prepared on a 12-well culture plate insert (Catalog No. 353494, Nippon Becton Deckonson). And a mixture of protease inhibitors (PIC, manufactured by Peptide Laboratories) At the same time as dissolving and eluting the lipid component of epithelial cells using 2 ml of 0.1% Triton X-100 (surfactant) in a phosphate buffer (pH 7.2; PBS (−)) After repeating the operation of dissolving the protein remaining on the surface of the basement membrane structure with the coexisting 50 mM NH 3 twice (the protein of the basement membrane structure must not be dissolved), the PBS (−) solution containing PIC again The base material was washed with surfactant and alkali to remove the alveolar type II epithelial cell layer and the basement membrane structure was exposed to reveal the mLN-111 isoform-type basement membrane (mLN111-sBM) substrate Was made.

b)T3-Alb7アイソフォーム型基底膜(T3-Alb7-sBM)基質の作製
 ヒト不死化肝実質細胞(T3-Alb7細胞、薬剤誘導性Cre組み換え酵素発現不死化ヒト肝細胞株 TTNT-16-T3(寄託機関:IPOD 独立行政法人産業技術総合研究所 特許生物寄託センター、あて名:日本国茨城県つくば市東1丁目1番地1 中央第6(郵便番号305-8566)、寄託日:平成15年10月1日、受託番号:FERM BP-08501、Totsugawa T, et al., Survival of liver failure pigs by transplantation of reversibly immortalized human hepatocytes with Tamoxifen-mediated self-recombination. J. Hepatol. 47: 74-82, 2007 参照、万人が使用できる))を、特開2003-93050号公報記載の方法に準じて糖鎖β-GlcNAcを結合したMASTポリマー(maleic anhydride-styrene copolymer:特許文献4参照)をコートしたコラーゲン線維上に播種し、約2週間培養することで基底膜構造体を形成させた。基底膜構造体形成後、50mM NH3、0.1% Triton X-100、及び、プロテアーゼインヒビターカクテル(PIC、ペプチド研究所社製)を含むD-PBS(-)溶液でT3-Alb7細胞を処理することで細胞のみを除去し、T3-Alb7細胞が形成した基底膜構造体を傷つけることなく露出させることで、T3-Alb7アイソフォーム型基底膜(T3-Alb7-sBM)基質を作製した。
b) Preparation of T3-Alb7 isoform-type basement membrane (T3-Alb7-sBM) substrate Human immortalized liver parenchymal cells (T3-Alb7 cells, drug-inducible Cre recombinase expressing immortalized human hepatocyte cell line TTNT-16-T3 (Depositing organization: IPOD, National Institute of Advanced Industrial Science and Technology, Patent Biological Depositary Center, Address: 1-chome, East 1-chome, Tsukuba, Ibaraki, Japan, Chuo 6 (postal code 305-8666), Date of deposit: October 2003 1 day, accession number: FERM BP-08501, Totsugawa T, et al., Survival of liver failure pigs by transplantation of reversibly immortalized human hepatocytes with Tamoxifen-mediated self-recombination. See J. Hepatol. 47: 74-82, 2007. MAST polymer (maleic anhydride-s) to which a sugar chain β-GlcNAc is bonded according to the method described in JP-A-2003-93050) A basement membrane structure was formed by seeding on collagen fibers coated with tyrene copolymer (see Patent Document 4) and culturing for about 2 weeks. After formation of the basement membrane structure, T3-Alb7 cells were treated with a D-PBS (−) solution containing 50 mM NH 3 , 0.1% Triton X-100, and protease inhibitor cocktail (PIC, manufactured by Peptide Laboratories). Thus, only the cells were removed, and the basement membrane structure formed by the T3-Alb7 cells was exposed without damaging, thereby preparing a T3-Alb7 isoform-type basement membrane (T3-Alb7-sBM) substrate.

(5)第3段階:iPS細胞由来肝実質細胞(iPS-Heps)への分化(第8日~第16日)
 第8日に、胚体内胚葉をトリプシン-EDTA(カタログ番号T4049、シグマ-アルドリッチジャパン)を用いてプレートより分離したのち、800rpmで3分間遠心分離し、ペレットとして収集した。KSR 10%、非必須アミノ酸 1%、L-グルタミン 1%、ジメチルスルホキシド(DMSO、カタログ番号D4540-100ML、シグマ-アルドリッチ)1%、HGF(カタログ番号100-39、ぺプロテック)100ng/mlを添加したノックアウトDMEMを培地として用意し、1ウェルから収集したペレットに対して0.5mlの培地で懸濁した。この懸濁液を、前記(4)で作製したmLN111-sBM基質またはT3-Alb7-sBM基質を含むインサート内に滴下した。
(5) Third stage: differentiation into iPS cell-derived hepatocytes (iPS-Heps) (from day 8 to day 16)
On the 8th day, definitive endoderm was separated from the plate using trypsin-EDTA (catalog number T4049, Sigma-Aldrich Japan), then centrifuged at 800 rpm for 3 minutes and collected as a pellet. KSR 10%, non-essential amino acid 1%, L-glutamine 1%, dimethyl sulfoxide (DMSO, catalog number D4540-100ML, Sigma-Aldrich) 1%, HGF (catalog number 100-39, Peprotech) 100 ng / ml The added knockout DMEM was prepared as a medium, and suspended in 0.5 ml of the medium collected from the pellet collected from one well. This suspension was dropped into the insert containing mLN111-sBM substrate or T3-Alb7-sBM substrate prepared in (4) above.

 インサート対応12ウェル培養プレート(カタログ番号353503、日本ベクトン・ディッキンソン)の1ウェルに培地1.5mlを加え、これに胚体内胚葉、mLN111-sBM基質および培地0.5mlを含むインサートを載せ培養し、これを実施例1とした。また、インサート対応12ウェル培養プレートのプレート底面に、あらかじめ5.0×105個のヒト不死化肝類洞内皮細胞株 TMNT-1(寄託機関:独立行政法人産業技術総合研究所 特許生物寄託センター、あて名:日本国茨城県つくば市東1丁目1番地1 中央第6(郵便番号305-8566)、寄託日:平成14年4月16日、受託番号:FERM BP-8017、参考文献5参照、万人が使用できる)を播種し、10%FBS(fetal bovine serum)ならびに1%ペニシリンおよびストレプトマイシンを添加した、4.5g/L濃度のグルコースを含むDMEM(DMEM high glucose)中、1日以上培養しておき、このウェルに培地1.5mlを加え、胚体内胚葉、mLN111-sBM基質および培地0.5mlを含むインサートを載せ培養し、これを実施例2とした。それぞれ第10日にHGFを含まない培地、すなわちKSR 10%、非必須アミノ酸 1%、L-グルタミン 1%、DMSO 1%を添加したノックアウトDMEMに交換し、以後は2日おきに全量の培地を交換した。分化誘導は第16日に終了した。基底膜基質を用いない比較例1と比べ、実施例1および2では多数のiPS細胞由来肝実質細胞が得られた(図1(E~G))。 1.5 ml of medium was added to one well of an insert-compatible 12-well culture plate (catalog number 353503, Nippon Becton Dickinson). This was designated Example 1. In addition, 5.0 × 10 5 human immortalized hepatic sinusoidal endothelial cell line TMNT-1 (deposited organization: National Institute of Advanced Industrial Science and Technology, Patent Organism Depositary) , Address: 1-chome, Higashi 1-chome, Tsukuba, Ibaraki, Japan, Central 6 (postal code 305-8656), deposit date: April 16, 2002, deposit number: FERM BP-8017, see reference 5, 10,000 Cultivated in DMEM (DMEM high glucose) containing 4.5 g / L glucose and supplemented with 10% FBS (fetal bovine serum) and 1% penicillin and streptomycin. Add 1.5 ml of the medium to the well, and place the insert containing the definitive endoderm, mLN111-sBM substrate and 0.5 ml of the medium. This was designated Example 2. On the 10th day, the medium was replaced with HGF-free medium, that is, knockout DMEM supplemented with 10% KSR, 1% non-essential amino acid, 1% L-glutamine, and 1% DMSO. Exchanged. Differentiation induction was completed on the 16th day. Compared to Comparative Example 1 in which no basement membrane substrate was used, in Examples 1 and 2, a greater number of iPS cell-derived hepatocytes were obtained (FIG. 1 (EG)).

 インサート対応12ウェル培養プレートの1ウェルに培地1.5mlを加え、これに胚体内胚葉、T3-Alb7-sBM基質および培地0.5mlを含むインサートを載せ培養し、これを実施例3とした。インサート対応12ウェル培養プレートのプレート底面に、あらかじめ5.0×105個のTMNK-1を播種し1日以上培養しておき、このウェルに培地1.5mlを加え、胚体内胚葉、T3-Alb7-sBM基質および培地0.5mlを含むインサートを載せ培養し、これを実施例4とした。それぞれ第10日にHGFを含まない培地、すなわちKSR 1%、非必須アミノ酸 1%、L-グルタミン 1%、DMSO 1%を添加したノックアウトDMEMに交換し、以後は2日おきに全量の培地を交換した。分化誘導は第16日に終了した。基底膜基質を用いない比較例1と比べ、実施例3および4では多数のiPS細胞由来肝実質細胞が得られた(図2および5)。 1.5 ml of medium was added to one well of a 12-well culture plate corresponding to insert, and an insert containing definitive endoderm, T3-Alb7-sBM substrate and 0.5 ml of medium was placed on the plate and cultured. On the bottom of the 12-well culture plate for inserts, 5.0 × 10 5 TMNK-1s are seeded beforehand and cultured for 1 day or more, 1.5 ml of medium is added to the wells, definitive endoderm, T3- An insert containing Alb7-sBM substrate and 0.5 ml of medium was placed and cultured, and this was designated as Example 4. On the 10th day, the medium was replaced with HGF-free medium, ie, knockout DMEM supplemented with 1% KSR, 1% non-essential amino acid, 1% L-glutamine, and 1% DMSO, and thereafter the entire medium was replaced every 2 days. Exchanged. Differentiation induction was completed on the 16th day. Compared to Comparative Example 1 in which no basement membrane substrate was used, in Examples 3 and 4, a greater number of iPS cell-derived hepatic parenchymal cells were obtained (FIGS. 2 and 5).

試験例1
iPS細胞由来肝実質細胞の機能評価
(A)アンモニア代謝能
 未分化iPS細胞ならびに実施例1および実施例2の方法にしたがって分化誘導したiPS細胞由来肝実質細胞および後述の比較例1の方法にしたがって分化誘導したiPS細胞由来肝実質細胞に0.56mM濃度のアンモニア硫酸塩(カタログ番号02619-15、シグマ-アルドリッチ製)をそれぞれ添加し、24時間後に培養上清を回収してアンモニア代謝能を測定した。アンモニア濃度はアンモニアテスト-ワコー(カタログ番号277-14401、和光純薬製)を用いて測定した。
Test example 1
Functional Evaluation of iPS Cell-Derived Liver Parenchymal Cells (A) Ammonia Metabolism Ability According to Undifferentiated iPS Cells, iPS Cell-Derived Liver Parenchymal Cells Differentiated According to the Methods of Example 1 and Example 2, 0.56 mM ammonia sulfate (catalog number 02619-15, Sigma-Aldrich) was added to iPS cell-derived hepatocytes derived from differentiation, and the culture supernatant was collected after 24 hours to measure ammonia metabolic capacity. did. The ammonia concentration was measured using Ammonia Test-Wako (catalog number 277-14401, manufactured by Wako Pure Chemical Industries).

 結果を図3に示す。未分化iPS細胞ではアンモニア濃度はむしろ増加し、除去率は-42.9±10.4%であった。本発明のiPS細胞由来肝実質細胞は、実施例1では培養液中に添加されたアンモニアの63.7±7.3%を代謝し、実施例2では31.9±5.3%を代謝した。これは比較例1のiPS細胞由来肝実質細胞の18.1±5.9%と比較して優れた代謝能を示した。 The results are shown in FIG. In undifferentiated iPS cells, the ammonia concentration rather increased, and the removal rate was −42.9 ± 10.4%. The iPS cell-derived hepatic parenchymal cells of the present invention metabolize 63.7 ± 7.3% of ammonia added to the culture medium in Example 1, and 31.9 ± 5.3% in Example 2. did. This showed an excellent metabolic capacity compared to 18.1 ± 5.9% of the iPS cell-derived hepatic parenchymal cells of Comparative Example 1.

(B)尿素の産生
 未分化iPS細胞ならびに実施例1および実施例2の方法にしたがって分化誘導したiPS細胞由来肝実質細胞および後述の比較例1の方法にしたがって分化誘導したiPS細胞由来肝実質細胞それぞれの尿素産生能を評価するため、培地交換より24時間後に培養液を採取し、尿素の産生量を測定(SRL社に委託)した。結果を図4に示す。未分化iPS細胞は尿素産生能を示さなかった。本発明のiPS細胞由来肝実質細胞は、実施例1では1ウェルあたり133±153ngの尿素産生能を、実施例2では1ウェルあたり167±153ngの尿素産生能を示した。これは比較例1のiPS細胞由来肝実質細胞の33.3±57.7ngと比較して尿素産生量が多い傾向を示した。
(B) Production of urea Undifferentiated iPS cells, iPS cell-derived liver parenchymal cells induced to differentiate according to the methods of Examples 1 and 2, and iPS cell-derived liver parenchymal cells induced to differentiate according to the method of Comparative Example 1 described later In order to evaluate each urea production ability, the culture solution was collected 24 hours after the medium exchange, and the production amount of urea was measured (consigned to SRL). The results are shown in FIG. Undifferentiated iPS cells did not show urea-producing ability. The iPS cell-derived hepatic parenchymal cells of the present invention exhibited 133 ± 153 ng urea production ability per well in Example 1, and 167 ± 153 ng urea production ability per well in Example 2. This showed a tendency for the amount of urea production to be large compared to 33.3 ± 57.7 ng of the iPS cell-derived liver parenchymal cells of Comparative Example 1.

(C)アルブミンの産生
 未分化iPS細胞ならびに実施例2~4の方法にしたがって分化誘導したiPS細胞由来肝実質細胞および後述の比較例1の方法にしたがって分化誘導したiPS細胞由来肝実質細胞それぞれのアルブミン産生量を評価するため、培地交換より24時間後に培養液を採取し、AssayMaxマウスアルブミンELISAキット(カタログ番号EMA3201-1、Gentaur製)を用いてアルブミン量を測定した。結果を図5に示す。未分化iPS細胞はアルブミン産生能を示さなかった。比較例1のiPS細胞由来肝実質細胞は53±38pgのアルブミン産生能を示した。実施例1ではアルブミン産生量を示さず、実施例2では514±890pgのアルブミン産生がみられた。これらに比べ、実施例3では2320±64pg、実施例4では2635±111pgと有意に高いアルブミン産生がみられた。
(C) Albumin production Each of undifferentiated iPS cells, iPS cell-derived liver parenchymal cells induced to differentiate according to the methods of Examples 2 to 4, and iPS cell-derived liver parenchymal cells induced to differentiate according to the method of Comparative Example 1 described later. In order to evaluate the amount of albumin produced, the culture solution was collected 24 hours after the medium exchange, and the amount of albumin was measured using an AssayMax mouse albumin ELISA kit (catalog number EMA3201-1, manufactured by Gentaur). The results are shown in FIG. Undifferentiated iPS cells did not show albumin producing ability. The iPS cell-derived hepatocytes of Comparative Example 1 showed an albumin producing ability of 53 ± 38 pg. In Example 1, albumin production was not shown, and in Example 2, albumin production of 514 ± 890 pg was observed. Compared to these, albumin production was significantly higher at 2320 ± 64 pg in Example 3 and 2635 ± 111 pg in Example 4.

(D)アルブミン発現の陽性率
 未分化iPS細胞、実施例4の方法にしたがって分化誘導したiPS細胞由来肝実質細胞、および後述の比較例1の方法にしたがって分化誘導したiPS細胞由来肝実質細胞のそれぞれのアルブミン発現の陽性率を評価するためラビット抗マウスアルブミン抗体(カタログ番号64560、MP biomedicals製)を一次抗体とし、DyLight54標識ヒツジ抗ラビット抗体(カタログ番号STAR36D549、MorphoSys UK製)を二次抗体とし、発現したアルブミン(Alb)を染色した。また、細胞の核をDAPI(カタログ番号H-1200、Vector Laboratories Inc,製)で免疫染色することにより総細胞数とAlb発現細胞数を測定し、Alb陽性細胞/DAPIで染色された核の数としてAlb発現の陽性率を算出した。結果を図2に示す。未分化iPS細胞および対照のT3-Alb7-sBM基質のみの試料ではAlb陽性率は0%であった。比較例1のiPS細胞由来肝実質細胞は20.6%のAlb陽性率を示した。一方、実施例4では31.8%と有意に高いAlb陽性率がみられた。
(D) Positive rate of albumin expression of undifferentiated iPS cells, iPS cell-derived liver parenchymal cells induced to differentiate according to the method of Example 4, and iPS cell-derived liver parenchymal cells induced to differentiate according to the method of Comparative Example 1 described later Rabbit anti-mouse albumin antibody (Cat. No. 64560, manufactured by MP biomedicals) is used as the primary antibody, and DyLight54-labeled sheep anti-rabbit antibody (Cat. # STAR36D549, manufactured by MorphoSys UK) is used as the secondary antibody to evaluate the positive rate of each albumin expression. The expressed albumin (Alb) was stained. In addition, the total number of cells and the number of Alb-expressing cells were measured by immunostaining the cell nuclei with DAPI (catalog number H-1200, manufactured by Vector Laboratories Inc.), and the number of nuclei stained with Alb-positive cells / DAPI. The positive rate of Alb expression was calculated. The results are shown in FIG. In the sample of undifferentiated iPS cells and the control T3-Alb7-sBM substrate alone, the Alb positive rate was 0%. The iPS cell-derived liver parenchymal cells of Comparative Example 1 showed an Alb positive rate of 20.6%. On the other hand, in Example 4, a significantly high Alb positive rate of 31.8% was observed.

比較例1
 実施例1の(1)~(3)までを同様に行ない胚体内胚葉を得た。
Comparative Example 1
The same procedures as in Example 1 (1) to (3) were performed to obtain definitive endoderm.

(4a)第3段階:iPS細胞由来肝実質細胞(iPS-Heps)への分化(第8日~第16日)
 第8日に培地をKSR 10%、非必須アミノ酸 1%、L-グルタミン 1%、DMSO 1%、HGF(カタログ番号100-39、ぺプロテック)100ng/mlを添加したノックアウトDMEMに交換した。培地は毎日半分の量を交換した。分化誘導は第16日に終了した。形態的に、分化の最終段階でマウスiPS細胞は肝実質細胞に類似した形態を示した。すなわち細胞質内に豊富な顆粒をもつ多角形の細胞となった(図1E、図6A、図6B)。
(4a) Third stage: differentiation into iPS cell-derived liver parenchymal cells (iPS-Heps) (Days 8 to 16)
On day 8, the medium was replaced with knockout DMEM supplemented with 10% KSR, 1% non-essential amino acids, 1% L-glutamine, 1% DMSO, 100 ng / ml HGF (Catalog No. 100-39, Peprotech). The medium was changed in half the amount every day. Differentiation induction was completed on the 16th day. Morphologically, at the final stage of differentiation, mouse iPS cells showed a morphology similar to that of hepatocytes. That is, it became a polygonal cell with abundant granules in the cytoplasm (FIG. 1E, FIG. 6A, FIG. 6B).

参考例1
比較例1のiPS細胞由来肝実質細胞(iPS-Heps)の細胞形態
 比較例1のiPS細胞由来肝実質細胞の細胞形態を位相差顕微鏡および電子顕微鏡を用いて観察した。位相差顕微鏡観察の強拡大像(図6B)では、類円形の核および細胞質内に豊富な顆粒をもつ多角形の細胞が明瞭に観察される。PAS染色陽性であり、細胞内顆粒がグリコーゲンであることを示す(図6C)。また電子顕微鏡観察ではグリコーゲンの集簇した部位であるグリコーゲン野(図6E)や、微絨毛を伴う管腔形成像(図6F)も認め、肝実質細胞に一致した細胞構造を示している。
Reference example 1
Cell morphology of iPS cell-derived liver parenchymal cells (iPS-Heps) in Comparative Example 1 The cell morphology of iPS cell-derived liver parenchymal cells in Comparative Example 1 was observed using a phase contrast microscope and an electron microscope. In the strongly magnified image of the phase contrast microscope observation (FIG. 6B), polygonal cells having an abundant circular nucleus and abundant granules in the cytoplasm are clearly observed. It is positive for PAS staining, indicating that intracellular granules are glycogen (FIG. 6C). In addition, observation by an electron microscope also shows a glycogen field (FIG. 6E), which is a site where glycogen is gathered, and a luminal formation image with microvilli (FIG. 6F), indicating a cell structure consistent with liver parenchymal cells.

参考例2
iPS細胞から肝実質細胞への分化誘導過程における遺伝子発現
 比較例1の分化誘導過程の各段階でRT-PCRを実施した。RNAはTRI zol(カタログ番号15596-026、インビトロジェン)を用いて抽出した。cDNAは2μgのRNAから、MuLV逆転写酵素(カタログ番号N8080018、アプライド バイオシステムズ(Applied Biosystems))およびRNアーゼ阻害剤(カタログ番号N8080119、アプライド バイオシステムズ)を用い、タカラ社性サーマルサイクラーを使用して作成した。RT-PCRはAmpliTaqゴールドDNAポリメラーゼ、GeneAmp PCRゴールドバッファー、MgCl2溶液(カタログ番号606080、アプライド バイオシステムズ)を用いて行った。遺伝子としては、アルファフェトタンパク質(AFP)、アルブミン(Alb)、トランスフェリン(Trf)、ホスホエノールピルビン酸カルボキシキナーゼ1(PCK1)、カルバミル燐酸シンテターゼ(CPS)、性決定遺伝子ボックス17(Sox17)および内因性コントロールであるβ-アクチンの遺伝子を調べた。RT-PCRで使用したプライマーを表1に示す。PCR産物は2.5%アガロースゲルにて電気泳動し、エチジウムブロマイドを用いて可視化した(図7A)。図7A中、レーン1~4はそれぞれ、iPS細胞、EB、DEおよびiPS-Hepsである。
Reference example 2
Gene expression in the process of inducing differentiation from iPS cells to liver parenchymal cells RT-PCR was performed at each stage of the process of inducing differentiation in Comparative Example 1. RNA was extracted using TRI zol (catalog number 15596-026, Invitrogen). cDNA was prepared from 2 μg of RNA using MuLV reverse transcriptase (Catalog No. N8080018, Applied Biosystems) and RNase inhibitor (Catalog No. N808119, Applied Biosystems) using a Takara Thermal Cycler. Created. RT-PCR was performed using AmpliTaq Gold DNA polymerase, GeneAmp PCR Gold Buffer, MgCl 2 solution (Catalog No. 606080, Applied Biosystems). The genes include alphafetoprotein (AFP), albumin (Alb), transferrin (Trf), phosphoenolpyruvate carboxykinase 1 (PCK1), carbamyl phosphate synthetase (CPS), sex-determining gene box 17 (Sox17) and endogenous The β-actin gene as a control was examined. The primers used in RT-PCR are shown in Table 1. PCR products were electrophoresed on 2.5% agarose gel and visualized using ethidium bromide (FIG. 7A). In FIG. 7A, lanes 1 to 4 are iPS cells, EB, DE, and iPS-Heps, respectively.

 リアルタイムRT-PCRはLight Cycler Fast Start DNA Master SYBR Green I(カタログ番号03003230001、 ロッシュアプライドサイエンス(Roche Applied Science)、IN)キットを使用し、ライトサイクラー1.5(ロッシュアプライドサイエンス)を用いて実施した。遺伝子としては、アルファフェトタンパク質(AFP)、アルブミン(Alb)、トランスフェリン(Trf)、ホスホエノールピルビン酸カルボキシキナーゼ1(PCK1)、カルバミル燐酸シンテターゼ(CPS)および内因性コントロールであるβ-アクチンの遺伝子を調べた。リアルタイムRT-PCRで使用したプライマーを表2に示す。 Real-time RT-PCR was performed using the Light Cycler 1.5 (Roche Applied Science) kit using the Light Cycler Fast DNA Master SYBR Green I (Catalog No. 03003230001, Roche Applied Science, IN) kit. . The genes include alphafetoprotein (AFP), albumin (Alb), transferrin (Trf), phosphoenolpyruvate carboxykinase 1 (PCK1), carbamyl phosphate synthetase (CPS) and the endogenous control β-actin gene. Examined. Table 2 shows the primers used in real-time RT-PCR.

 RT-PCRおよびリアルタイムRT-PCRの結果では、分化過程に伴って成熟肝実質細胞の指標であるアルブミンの遺伝子発現が徐々に増加した。また尿素合成に関する酵素のカルバミルリン酸シンテターゼ(CPS)、糖新生を調節する酵素のホスホエノールピルビン酸カルボキシキナーゼ1(PCK1)、トランスフェリン(Trf)など、肝実質細胞で豊富にみられる遺伝子についても経時的に発現が増強していた(図7B)。 In the results of RT-PCR and real-time RT-PCR, the gene expression of albumin, which is an indicator of mature liver parenchymal cells, gradually increased with the differentiation process. The genes that are abundant in liver parenchymal cells such as carbamyl phosphate synthetase (CPS), an enzyme related to urea synthesis, phosphoenolpyruvate carboxykinase 1 (PCK1), an enzyme that regulates gluconeogenesis, and so on are also analyzed over time. Expression was enhanced (FIG. 7B).

Figure JPOXMLDOC01-appb-T000001
Figure JPOXMLDOC01-appb-T000001

Figure JPOXMLDOC01-appb-T000002
Figure JPOXMLDOC01-appb-T000002

参考例3
 胚体内胚葉の分化効率に対する胚様体の形成期間の影響を調べた。
Reference example 3
The effect of embryoid body formation period on the definitive endoderm differentiation efficiency was investigated.

 実施例1の第1段階と同様に5日間かけて形成したEBを、実施例1の第2段階と同様の条件で内胚葉分化誘導を5日間かけて行った(試料1)。形成期間を2日間とした以外は実施例1の第1段階と同様に形成したEBを、実施例1の第2段階と同様の条件で内胚葉分化誘導を5日間かけて行った(試料2)。得られた細胞の遺伝子の発現を実施例2と同様にリアルタイムPCRで分析した。調べた遺伝子は性決定遺伝子ボックス17(Sox17)、ケモカイン(C-X-Cモチーフ)受容体4(chemokine (C-X-C motif) receptor 4)(Cxcr4)およびフォークヘッドボックスA2(forkhead box A2)(Foxa2)であり、使用したプライマーを表3に示す。 EBs formed over 5 days as in the first stage of Example 1 were subjected to endoderm differentiation induction over 5 days under the same conditions as in the second stage of Example 1 (Sample 1). EBs formed in the same manner as in the first stage of Example 1 except that the formation period was 2 days were subjected to endoderm differentiation induction over 5 days under the same conditions as in the second stage of Example 1 (Sample 2). ). The gene expression of the obtained cells was analyzed by real-time PCR as in Example 2. The genes examined were sex-determining gene box 17 (Sox17), chemokine (C—X—C motif) receptor 4 (chemokine (CXC motif) receptor 4) (Cxcr4) and forkhead box A2 (forkhead box A2) (Foxa2) Table 3 shows the primers used.

 試料1では、試料2と比較して、汎内胚葉マーカーであるSox17およびFoxa2の発現はそれぞれ1.4および2.8倍であり、また胚体内胚葉マーカーであるCxcr4の発現は1.1倍であった。 In sample 1, compared to sample 2, the expression of panendoderm markers Sox17 and Foxa2 is 1.4 and 2.8-fold, respectively, and the expression of definitive endoderm marker Cxcr4 is 1.1-fold. Met.

 以上より、胚様体の形成期間は2日間よりも5日間のほうが、その後の胚体内胚葉の分化効率が良好であることが示された。 From the above, it was shown that the differentiation efficiency of the definitive endoderm is better when the embryoid body is formed for 5 days than for 2 days.

Figure JPOXMLDOC01-appb-T000003
Figure JPOXMLDOC01-appb-T000003

[参考文献]
1) Kubo A, Shinozaki K, Shannon JM, Kouskoff V, Kennedy M, Woo S, Fehling HJ, Keller G. Development of definitive endoderm from embryonic stem cells in culture. Development 2004;131:1651-1662.
2) Basma H, Soto-Gutieerrez A, Yannam GR, Liu L, Ito R, Yamamoto T, Ellis E, Carson SD, Sato S, Chen Y, Muirhead D, Navarro-Alvarez N, Wong RJ, Roy-Chowdhury J, Platt JL, Mercer DF, Miller JD, Strom SC, Kobayashi N, Fox IJ. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes. Gastroenterology 2009;136:990-999.
3) Soto-Gutieerrez A, Kobayashi N, Rivas-Carrillo JD, Navarro-Alvarez N, Zhao D, Okitsu T, Noguchi H, Basma H, Tabata Y, Chen Y, Tanaka K, Narushima M, Miki A, Ueda T, Jun HS, Yoon JW, Lebkowski J, Tanaka N, Fox IJ. Reversal of mouse hepatic failure using an implanted liver-assist device containing ES cell-derived hepatocytes. Nat Biotechnol 2006;24:1412-1419.
4) Yasunaga M, Tada S, Torikai-Nishikawa S, Nakano Y, Okada M, Jakt LM, Nishikawa S, Chiba T, Era T, Nishikawa S. Induction and monitoring of definitive and visceral endoderm differentiation of mouse ES cells. Nat Biotechnol 2005;23:1542-1550.
5) Matsumura, T. et al. Establishment of an immortalized human-liver endothelial cell line with SV40T and hTERT. Transplantation 77, 1357-1365 (2004).
6) Clement et al., Exp. Cell Res., 196:198-205, 1991.
7) Hosokawa T, Furuyama A, Katagiri K, Betsuyaku T, Nishimura M, and Mochitate K.   Differentiation of Tracheal Basal Cells to Ciliated Cells and Tissue Reconstruction on the Synthesized Basement Membrane Substratum In Vitro.  Connective Tissue Research 48:9-18, 2007.
8) Totsugawa T, Chen Y, Soto-Gutierrez A, Rivas-Carrillo1 JD, Navarro-Alvarez N, Noguchi H, Okitsu T, Westerman KA, Tanaka N, Leboulch P, and Kobayashi N. Survival of liver failure pigs by transplantation of 3 reversibly immortalized human hepatocytes with Tamoxifen-mediated self-recombination.  J. Hepatol. 47:74-82, 2007.
[References]
1) Kubo A, Shinozaki K, Shannon JM, Kouskoff V, Kennedy M, Woo S, Fehling HJ, Keller G. Development of definitive endoderm from embryonic stem cells in culture.Development 2004; 131: 1651-1662.
2) Basma H, Soto-Gutieerrez A, Yannam GR, Liu L, Ito R, Yamamoto T, Ellis E, Carson SD, Sato S, Chen Y, Muirhead D, Navarro-Alvarez N, Wong RJ, Roy-Chowdhury J, Platt JL, Mercer DF, Miller JD, Strom SC, Kobayashi N, Fox IJ. Differentiation and transplantation of human embryonic stem cell-derived hepatocytes.Gastroenterology 2009; 136: 990-999.
3) Soto-Gutieerrez A, Kobayashi N, Rivas-Carrillo JD, Navarro-Alvarez N, Zhao D, Okitsu T, Noguchi H, Basma H, Tabata Y, Chen Y, Tanaka K, Narushima M, Miki A, Ueda T, Jun HS, Yoon JW, Lebkowski J, Tanaka N, Fox IJ.Reversal of mouse hepatic failure using an implanted liver-assist device containing ES cell-derived hepatocytes. Nat Biotechnol 2006; 24: 1412-1419.
4) Yasunaga M, Tada S, Torikai-Nishikawa S, Nakano Y, Okada M, Jakt LM, Nishikawa S, Chiba T, Era T, Nishikawa S. Induction and monitoring of definitive and visceral endoderm differentiation of mouse ES cells.Nat Biotechnol 2005; 23: 1542-1550.
5) Matsumura, T. et al. Establishment of an immortalized human-liver endothelial cell line with SV40T and hTERT.Transplantation 77, 1357-1365 (2004).
6) Clement et al., Exp. Cell Res., 196: 198-205, 1991.
7) Hosokawa T, Furuyama A, Katagiri K, Betsuyaku T, Nishimura M, and Mochitate K. Differentiation of Tracheal Basal Cells to Ciliated Cells and Tissue Reconstruction on the Synthesized Basement Membrane Substratum In Vitro. Connective Tissue Research 48: 9-18, 2007.
8) Totsugawa T, Chen Y, Soto-Gutierrez A, Rivas-Carrillo1 JD, Navarro-Alvarez N, Noguchi H, Okitsu T, Westerman KA, Tanaka N, Leboulch P, and Kobayashi N. Survival of liver failure pigs by transplantation of 3 reversibly immortalized human hepatocytes with Tamoxifen-mediated self-recombination. J. Hepatol. 47: 74-82, 2007.

配列番号1:AFP遺伝子を検出するためのPCR用フォワードプライマー
配列番号2:AFP遺伝子を検出するためのPCR用リバースプライマー
配列番号3:Alb遺伝子を検出するためのPCR用フォワードプライマー
配列番号4:Alb遺伝子を検出するためのPCR用リバースプライマー
配列番号5:Trf遺伝子を検出するためのPCR用フォワードプライマー
配列番号6:Trf遺伝子を検出するためのPCR用リバースプライマー
配列番号7:PCK1遺伝子を検出するためのPCR用フォワードプライマー
配列番号8:PCK1遺伝子を検出するためのPCR用リバースプライマー
配列番号9:CPS遺伝子を検出するためのPCR用フォワードプライマー
配列番号10:CPS遺伝子を検出するためのPCR用リバースプライマー
配列番号11:Sox17遺伝子を検出するためのPCR用フォワードプライマー
配列番号12:Sox17遺伝子を検出するためのPCR用リバースプライマー
配列番号13:β-アクチン遺伝子を検出するためのPCR用フォワードプライマー
配列番号14:β-アクチン遺伝子を検出するためのPCR用リバースプライマー
配列番号15:AFP遺伝子を検出するためのリアルタイムRT-PCR用フォワードプライマー
配列番号16:AFP遺伝子を検出するためのリアルタイムRT-PCR用リバースプライマー
配列番号17:Alb遺伝子を検出するためのリアルタイムRT-PCR用フォワードプライマー
配列番号18:Alb遺伝子を検出するためのリアルタイムRT-PCR用リバースプライマー
配列番号19:Sox17遺伝子を検出するためのリアルタイムPCR用フォワードプライマー
配列番号20:Sox17遺伝子を検出するためのリアルタイムPCR用リバースプライマー
配列番号21:Cxcr4遺伝子を検出するためのリアルタイムPCR用フォワードプライマー
配列番号22:Cxcr4遺伝子を検出するためのリアルタイムPCR用リバースプライマー
配列番号23:Foxa2遺伝子を検出するためのリアルタイムPCR用フォワードプライマー
配列番号24:Foxa2遺伝子を検出するためのリアルタイムPCR用リバースプライマー
SEQ ID NO: 1: PCR forward primer for detecting AFP gene SEQ ID NO: 2: PCR reverse primer for detecting AFP gene SEQ ID NO: 3: PCR forward primer for detecting Alb gene SEQ ID NO: 4: Alb PCR reverse primer for detecting gene SEQ ID NO: 5: PCR forward primer for detecting Trf gene SEQ ID NO: 6: PCR reverse primer for detecting Trf gene SEQ ID NO: 7: for detecting PCK1 gene PCR forward primer SEQ ID NO: 8: PCR reverse primer for detecting PCK1 gene SEQ ID NO: 9: PCR forward primer for detecting CPS gene SEQ ID NO: 10: PCR reverse primer for detecting CPS gene Limer SEQ ID NO: 11: PCR forward primer for detecting the Sox17 gene SEQ ID NO: 12: PCR reverse primer for detecting the Sox17 gene SEQ ID NO: 13: PCR forward primer SEQ ID NO: for detecting the β-actin gene 14: Reverse primer for PCR for detecting β-actin gene SEQ ID NO: 15: Forward primer for real-time RT-PCR for detecting AFP gene SEQ ID NO: 16: Reverse for real-time RT-PCR for detecting AFP gene Primer SEQ ID NO: 17: Real-time RT-PCR forward primer for detecting Alb gene SEQ ID NO: 18: Real-time RT-PCR reverse primer for detecting Alb gene SEQ ID NO: 19 Real-time PCR forward primer for detecting the ox17 gene SEQ ID NO: 20: Real-time PCR reverse primer for detecting the Sox17 gene SEQ ID NO: 21: Real-time PCR forward primer for detecting the Cxcr4 gene SEQ ID NO: 22: Cxcr4 gene Reverse primer for real-time PCR for detecting NO SEQ ID NO: 23: Forward primer for real-time PCR for detecting Foxa2 gene SEQ ID NO: 24: Reverse primer for real-time PCR for detecting Foxa2 gene

Claims (6)

iPS細胞から肝実質細胞への分化誘導方法であって、
(a)iPS細胞を浮遊培養し、胚様体を形成する工程、
(b)被覆剤としてゼラチンを使用したプレートへ胚様体を1ウェルあたり10~15個として接着させ培養することにより、胚体内胚葉を誘導する工程、および
(c)HGFおよびDMSOを添加し、基底膜基質上にて胚体内胚葉を培養することにより肝実質細胞分化を誘導する工程
を含む方法。
A method for inducing differentiation from iPS cells to hepatocytes,
(A) a step of suspension culture of iPS cells to form an embryoid body,
(B) inducing definitive endoderm by adhering and culturing 10-15 embryoid bodies on a plate using gelatin as a coating agent per well, and (c) adding HGF and DMSO, A method comprising inducing hepatocyte differentiation by culturing definitive endoderm on a basement membrane substrate.
前記(a)の浮遊培養期間が5日間である請求項1記載の方法。 The method according to claim 1, wherein the suspension culture period of (a) is 5 days. 前記基底膜基質が肺胞II型上皮細胞または肝実質細胞由来である請求項1記載の方法。 The method according to claim 1, wherein the basement membrane matrix is derived from alveolar type II epithelial cells or hepatocytes. 前記(c)の分化誘導がさらに肝臓の非実質細胞との共培養によって行われる請求項1記載の方法。 The method according to claim 1, wherein the differentiation induction of (c) is further performed by co-culture with non-parenchymal cells of the liver. 肝臓の非実質細胞が肝類洞内皮細胞である請求項4記載の方法。 The method according to claim 4, wherein the nonparenchymal cells of the liver are hepatic sinusoidal endothelial cells. 請求項1の方法により誘導される肝実質細胞。 Liver parenchymal cells induced by the method of claim 1.
PCT/JP2010/063185 2009-08-04 2010-08-04 METHOD FOR INDUCING DIFFERENTIATION OF iPS CELLS INTO HEPATIC PARENCHYMAL CELLS Ceased WO2011016485A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2011525912A JPWO2011016485A1 (en) 2009-08-04 2010-08-04 Method for inducing differentiation from iPS cells to hepatocytes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2009181891 2009-08-04
JP2009-181891 2009-08-04

Publications (1)

Publication Number Publication Date
WO2011016485A1 true WO2011016485A1 (en) 2011-02-10

Family

ID=43544380

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2010/063185 Ceased WO2011016485A1 (en) 2009-08-04 2010-08-04 METHOD FOR INDUCING DIFFERENTIATION OF iPS CELLS INTO HEPATIC PARENCHYMAL CELLS

Country Status (2)

Country Link
JP (1) JPWO2011016485A1 (en)
WO (1) WO2011016485A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015523063A (en) * 2012-05-23 2015-08-13 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Compositions of endoderm cells and liver parenchymal cells and methods of obtaining and using those cells
WO2019131938A1 (en) * 2017-12-27 2019-07-04 国立研究開発法人国立成育医療研究センター Method for preparing functional liver progenitor cells or liver cells, or preparing functional small-intestinal epithelial progenitor cells or small-intestinal epithelial cells
EP3431586A4 (en) * 2016-02-29 2019-10-23 Yoshikazu Yonemitsu HIGHLY FUNCTIONAL HEPATIC CELLS AND USE THEREOF
CN114377209A (en) * 2020-10-19 2022-04-22 清华大学 Artificial liver structure containing bile duct and liver tissue and its preparation method and application
CN114381420A (en) * 2020-10-19 2022-04-22 清华大学 Hepatic tissue-like structure and preparation method and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003093050A (en) * 2001-09-25 2003-04-02 Japan Science & Technology Corp Preparation method of basement membrane
JP2003093053A (en) * 2001-09-25 2003-04-02 Japan Science & Technology Corp Preparation of basement membrane preparation
WO2008066199A1 (en) * 2006-12-01 2008-06-05 Naoya Kobayashi Method of inducing differentiation of embryonic stem cells into insulin-secreting cells, insulin-secreting cells induced by the method and use of the same
JP2008306987A (en) * 2007-06-15 2008-12-25 Covalent Materials Corp Cell culture carrier and cell culture method
WO2009057831A1 (en) * 2007-10-31 2009-05-07 Kyoto University Nuclear reprogramming method

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003093050A (en) * 2001-09-25 2003-04-02 Japan Science & Technology Corp Preparation method of basement membrane
JP2003093053A (en) * 2001-09-25 2003-04-02 Japan Science & Technology Corp Preparation of basement membrane preparation
WO2008066199A1 (en) * 2006-12-01 2008-06-05 Naoya Kobayashi Method of inducing differentiation of embryonic stem cells into insulin-secreting cells, insulin-secreting cells induced by the method and use of the same
JP2008306987A (en) * 2007-06-15 2008-12-25 Covalent Materials Corp Cell culture carrier and cell culture method
WO2009057831A1 (en) * 2007-10-31 2009-05-07 Kyoto University Nuclear reprogramming method

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BASMA, H. ET AL.: "Differentiation and Transplantation of Human Embryonic Stem Cell- Derives Hepatocytes.", GASTROENTEROLOGY, vol. 163, no. 3, 29 October 2008 (2008-10-29), pages 990 - 999 *
HOSOKAWA, T. ET AL.: "Differentiation of tracheal basal cells to ciliated cells and tissue reconstruction on the synthesized basement membrane substratum in vitro.", CONNECT TISSUE RES., vol. 48, no. 1, 2007, pages 9 - 18 *
IWAMURO, M. ET AL.: "Comparative analysis of endoderm formation efficiency between mouse ES cells and iPS cells.", CELL TRANSPLANT., vol. 19, no. 6, 29 June 2010 (2010-06-29), pages 831 - 839, XP055117434, DOI: doi:10.3727/096368910X508951 *
IWAMURO, M. ET AL.: "Hepatic differentiation of mouse iPS cells in vitro.", CELL TRANSPLANT., vol. 19, no. 6, 29 June 2010 (2010-06-29), pages 841 - 847 *
NAOYA KOBAYASHI ET AL.: "Regeneration medicine of the liver", CLINIC ALL-ROUND, vol. 58, no. 1, January 2009 (2009-01-01), pages 79 - 85 *
SOTO-GUTIERREZ, A. ET AL.: "Reversal of mouse hepatic failure using an implanted liver- assist device containing ES cell-derived hepatocytes.", NAT. BIOTECHNOL., vol. 24, no. 11, 5 November 2006 (2006-11-05), pages 1412 - 1419, XP008127326, DOI: doi:10.1038/nbt1257 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015523063A (en) * 2012-05-23 2015-08-13 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Compositions of endoderm cells and liver parenchymal cells and methods of obtaining and using those cells
JP2017113005A (en) * 2012-05-23 2017-06-29 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Compositions of endoderm cells and hepatic parenchymal cells and methods for obtaining and using these cells
EP3431586A4 (en) * 2016-02-29 2019-10-23 Yoshikazu Yonemitsu HIGHLY FUNCTIONAL HEPATIC CELLS AND USE THEREOF
WO2019131938A1 (en) * 2017-12-27 2019-07-04 国立研究開発法人国立成育医療研究センター Method for preparing functional liver progenitor cells or liver cells, or preparing functional small-intestinal epithelial progenitor cells or small-intestinal epithelial cells
US20200339955A1 (en) * 2017-12-27 2020-10-29 Akihiro Umezawa Method for preparing functional hepatic progenitor cells or hepatic cells, or functional small intestinal epithelial progenitor cells or small intestinal epithelial cells
JPWO2019131938A1 (en) * 2017-12-27 2021-08-26 明弘 梅澤 Method for preparing functional hepatic progenitor cells or hepatocytes or functional small intestinal epithelial progenitor cells or small intestinal epithelial cells
JP7191041B2 (en) 2017-12-27 2022-12-16 明弘 梅澤 Method for preparing functional hepatic progenitor cells or hepatocytes or functional small intestinal epithelial progenitor cells or small intestinal epithelial cells
US12110507B2 (en) 2017-12-27 2024-10-08 Akihiro Umezawa Method for preparing functional hepatic progenitor cells or hepatic cells, or functional small intestinal epithelial progenitor cells or small intestinal epithelial cells
CN114377209A (en) * 2020-10-19 2022-04-22 清华大学 Artificial liver structure containing bile duct and liver tissue and its preparation method and application
CN114381420A (en) * 2020-10-19 2022-04-22 清华大学 Hepatic tissue-like structure and preparation method and application thereof
CN114377209B (en) * 2020-10-19 2022-09-23 清华大学 Artificial liver structure containing bile duct and liver tissue and preparation method and application thereof
CN114381420B (en) * 2020-10-19 2024-01-30 清华大学 Liver-like tissue structure body and preparation method and application thereof

Also Published As

Publication number Publication date
JPWO2011016485A1 (en) 2013-01-10

Similar Documents

Publication Publication Date Title
US20240309330A1 (en) Differentiation and enrichment of islet-like cells from human pluripotent stem cells
EP3397753B1 (en) Microtissue formation using stem cell-derived human hepatocytes
Luo et al. Three-dimensional hydrogel culture conditions promote the differentiation of human induced pluripotent stem cells into hepatocytes
JP5611035B2 (en) Efficient production and use of highly cardiogenic progenitor cells and cardiomyocytes from embryonic stem cells and induced pluripotent stem cells
Xu et al. Three-dimensional culture promotes the differentiation of human dental pulp mesenchymal stem cells into insulin-producing cells for improving the diabetes therapy
US20120231490A1 (en) Method Of Differentiation From Stem Cells To Hepatocytes
KR101861171B1 (en) Cardiomyocyte medium with dialyzed serum
WO2011009294A1 (en) Methods for obtaining hepatic cells, hepatic endoderm cells and hepatic precursor cells by inducing the differentiation
JP2005521405A (en) Dedifferentiated programmable stem cells originating from monocytes and their production and use
CN105296418A (en) Method for long-time in-vitro culturing and proliferating hepatic cells and application of method
EP3132023B1 (en) Method of preparing cells for 3d tissue culture
JPWO2008066199A1 (en) Method for inducing differentiation of embryonic stem cells into insulin-secreting cells, insulin-secreting cells induced by the method, and use thereof
JP6421335B2 (en) Method for culturing hepatic progenitor-like cells and culture thereof
US20250164464A1 (en) Liver organoid-derived cultured hepatocytes
US20050064587A1 (en) Pancreatic small cells and uses thereof
WO2011016485A1 (en) METHOD FOR INDUCING DIFFERENTIATION OF iPS CELLS INTO HEPATIC PARENCHYMAL CELLS
JP6139054B2 (en) Cell culture substrate, cell culture method using the same, and differentiation induction method for pluripotent stem cells
US20230078230A1 (en) Methods for the production of committed cardiac progenitor cells
KR20250075707A (en) Method for producing cardiac fibroblasts
Hefei et al. Morphological characteristics and identification of islet‐like cells derived from rat adipose‐derived stem cells cocultured with pancreas adult stem cells
JP6486619B2 (en) Drug evaluation cell and drug evaluation method
TW201734206A (en) Highly functional liver cells, and use thereof
JP2007014273A (en) Liver tissue / organ and method for producing the same
KR20250103497A (en) A mature hepatocyte cell sheet derived from induced pluripotent stem cell, and a method for treating Hemophilia B using the cell sheet
Pettinato et al. Liver Research

Legal Events

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

Ref document number: 10806484

Country of ref document: EP

Kind code of ref document: A1

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2011525912

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10806484

Country of ref document: EP

Kind code of ref document: A1