WO2020138208A1 - Method for producing hepatocytes - Google Patents

Method for producing hepatocytes Download PDF

Info

Publication number
WO2020138208A1
WO2020138208A1 PCT/JP2019/050926 JP2019050926W WO2020138208A1 WO 2020138208 A1 WO2020138208 A1 WO 2020138208A1 JP 2019050926 W JP2019050926 W JP 2019050926W WO 2020138208 A1 WO2020138208 A1 WO 2020138208A1
Authority
WO
WIPO (PCT)
Prior art keywords
hepatocytes
test substance
cells
producing
medium
Prior art date
Application number
PCT/JP2019/050926
Other languages
French (fr)
Japanese (ja)
Inventor
健二 長船
勝太郎 安田
伸二 上本
Original Assignee
国立大学法人京都大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立大学法人京都大学 filed Critical 国立大学法人京都大学
Priority to JP2020563365A priority Critical patent/JP7452799B2/en
Publication of WO2020138208A1 publication Critical patent/WO2020138208A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/37Digestive system
    • A61K35/407Liver; Hepatocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61LMETHODS OR APPARATUS FOR STERILISING MATERIALS OR OBJECTS IN GENERAL; DISINFECTION, STERILISATION OR DEODORISATION OF AIR; CHEMICAL ASPECTS OF BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES; MATERIALS FOR BANDAGES, DRESSINGS, ABSORBENT PADS OR SURGICAL ARTICLES
    • A61L27/00Materials for grafts or prostheses or for coating grafts or prostheses
    • A61L27/36Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix
    • A61L27/38Materials for grafts or prostheses or for coating grafts or prostheses containing ingredients of undetermined constitution or reaction products thereof, e.g. transplant tissue, natural bone, extracellular matrix containing added animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/867Retroviral vectors
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6851Quantitative amplification
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing

Definitions

  • the present invention relates to cell-related technology, and more particularly to a method for producing functionally mature mammalian hepatocytes.
  • hepatocyte transplantation therapy liver regenerative medicine
  • liver reconstruction liver disease model preparation
  • drug discovery drug hepatotoxicity evaluation system construction
  • Non-patent document 1 reports that induced hepatocytes were prepared by introducing Hnf4a and the genes of Foxa1, Foxa2, or Foxa3 into mouse fetal and adult fibroblasts.
  • Non-Patent Document 2 reports that induced hepatocytes were produced by introducing Gata4, Hnf1a and Foxa3 genes into adult mouse fibroblasts.
  • Non-Patent Document 3 reports that induced hepatocytes were prepared by introducing Hnf1b and Foxa3 genes into mouse fetal fibroblasts.
  • Non-Patent Document 4 reports that induced hepatocytes were produced by introducing FOXA3, HNF1A and HNF4A genes into human fibroblasts.
  • Non-Patent Document 5 reports that induced hepatocytes were prepared by introducing genes for HNF1A, HNF4A, HNF6, ATF5, PROX1 and CEBPA into human fibroblasts.
  • the induced hepatocytes that have been reported so far have a low expression amount of a liver marker, are functionally immature, and cannot be used for the purpose of regenerative medicine as described above.
  • An object of the present invention is to provide a method for efficiently producing functionally mature hepatocytes.
  • Hnf4a Hepatocyte nuclear factor4alpha
  • Foxa3 Foxa3
  • Cebpa CAAT/enhancer-binding protein alpha
  • Cebpd CAAT/enhancer-binding protein delta
  • Hnf6 Hepatocyte nuclear factor6
  • the gist of the present invention is as follows.
  • a method for producing hepatocytes which comprises a step of preparing somatic cells introduced with Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2, and a step of culturing the somatic cells to induce hepatocytes.
  • [4] The method for producing hepatocytes according to any one of [1] to [3], wherein Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are derived from human or mouse.
  • [5] The method for producing hepatocytes according to any one of [1] to [4], wherein Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are introduced using a retrovirus vector.
  • [6] The method for producing hepatocytes according to any one of [1] to [5], wherein Myc is further introduced into the somatic cells.
  • [7] The method for producing hepatocytes according to any one of [1] to [6], wherein the culturing step is performed for 20 days or more.
  • the medium contains a TGF ⁇ (Transforming growth factor beta) receptor inhibitor.
  • TGF ⁇ Transforming growth factor beta
  • [11] A cell preparation for regenerative medicine containing the hepatocyte according to [10].
  • a method for evaluating toxicity of a test substance which comprises the following steps in the order of (i)-(iii): (I) a step of contacting a test substance with hepatocytes produced by the method according to any one of [1] to [9], (Ii) the step of measuring the survival rate of the hepatocytes or the amount of the liver injury marker in the culture supernatant, and (iii) the survival rate of the hepatocytes contacted with the test substance does not contact with the test substance.
  • hepatocytes can be efficiently produced.
  • hepatocyte transplantation therapy for intractable liver diseases liver regenerative medicine
  • liver disease model preparation for intractable liver diseases (liver regenerative medicine)
  • drug development using hepatocytes can be performed with higher accuracy than ever before.
  • the hepatocytes obtained by the method of the present invention can also be used for the purpose of elucidating the differentiation and maturation mechanism of hepatocytes.
  • 6 is a graph showing the expression levels of Vimentin, Cola1 (collagen 1a1), and Afp (Alpha-fetoprotein) when hepatocytes induced by introducing 6 factors were cultured with or without A83-01.
  • 6 is a graph showing the amount of albumin secreted when hepatocytes induced by introducing 6 factors were cultured with or without A83-01.
  • the figure which shows the result of having measured the albumin secretion amount in a culture medium per 24 hours by ELISA. Albumin secretion was corrected by the number of cells that became Albumin-positive by immunostaining. N 3, and the mean value ⁇ standard deviation is shown. **P ⁇ 0.01 (one-way ANOVA with Tukey’s correction).
  • the figure which shows the result of having compared the enzyme activity of Cyp3a11 in each cell. N 3, and the mean value ⁇ standard deviation is shown.
  • FIG. 6 is a graph showing CYP gene expression fluctuations in hepatocytes induced by introducing 6 factors+Myc when stimulated with each drug.
  • the arrow indicates a gene whose expression level is remarkably increased.
  • Graph showing the expression level (n 3, mean ⁇ SD) of Cps1 (Carbamoylphosphate synthase I) gene in hepatocytes induced by introducing 2 to 6 factors.
  • the value in hepatocytes induced by introducing 2 factors (Hnf4a+Foxa3) after standardization with ⁇ -actin (Actb) was expressed as 1.
  • the figure (photograph) which shows the immunostaining result of the induced hepatocyte obtained by introduce
  • the method for producing hepatocytes of the present invention includes a step of preparing somatic cells introduced with Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2, and a step of culturing the somatic cells to induce hepatocytes. ..
  • Step of preparing 6-factor introduced somatic cells is not limited to the case where 6 factors are introduced into somatic cells and newly prepared, and the case where all the remaining factors are introduced into somatic cells in which a part of 6 factors has already been introduced, or 6 It also includes the case of procuring somatic cells into which the factor has been introduced.
  • somatic cells are not particularly limited as long as they are somatic cells other than hepatocytes, for example fibroblasts, epithelial cells (skin epidermal cells, oral mucosal epithelial cells, respiratory mucosal epithelial cells, intestinal mucosal epithelial cells, etc.), Examples thereof include epidermal cells, gingival cells (gingival fibroblasts, gingival epithelial cells), pulp cells, white adipocytes, subcutaneous fat, visceral fat, muscle, blood cells and the like.
  • a body prepared by inducing or dedifferentiating or reprogramming somatic stem cells such as mesenchymal stem cells (MSCs), intestinal stem cells, skin stem cells, hair follicle stem cells, and pigment cell stem cells
  • somatic stem cells such as mesenchymal stem cells (MSCs), intestinal stem cells, skin stem cells, hair follicle stem cells, and pigment cell stem cells
  • MSCs mesenchymal stem cells
  • intestinal stem cells such as intestinal stem cells, skin stem cells, hair follicle stem cells, and pigment cell stem cells
  • pigment cell stem cells also included are cells.
  • Somatic cells may be of adult, pediatric or fetal origin. Somatic cells may be those isolated from mammals other than humans such as humans, mice, rats and pigs, or subcultured cells thereof.
  • fibroblasts for example, fetal fibroblasts, tail-end fibroblasts, cardiac fibroblasts, foreskin fibroblasts, skin fibroblasts, lung fibroblasts, etc. can be used.
  • the somatic cells used for the production of the hepatocytes are the same or substantially the same HLA genotype of the transplant destination individual from the viewpoint that a rejection reaction does not occur. It is desirable that the somatic cells are the same.
  • “substantially the same” means that the HLA genotypes are matched to the extent that the immunoreactivity of the transplanted cells can be suppressed by the immunosuppressant, and for example, the transplant recipient individual and HLA- It is a somatic cell having an HLA type in which 3 loci of A, HLA-B and HLA-DR or 4 loci including HLA-C are matched.
  • ⁇ 6 factors> Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are used as 6 factors.
  • Hnf4a is called Hepatocyte nuclear factor4alpha, and is known to regulate the expression of many genes involved in hepatocyte function, such as glucose metabolism, lipid metabolism, and drug metabolism.
  • the amino acid sequence of human HNF4A protein is registered, for example, in GenBank Accession No. NP_849180 (SEQ ID NO: 1), and the amino acid sequence of mouse HNF4a protein is registered, for example, in GenBank Accession No. NP_032287 (SEQ ID NO: 2).
  • it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences.
  • the base sequence encoding the human HNF4a protein is registered, for example, in GenBank Accession No. NM_178849, and the base sequence encoding the mouse HNF4a protein is registered, for example, in GenBank Accession No. NM_008261.
  • Foxa3 is called Forkhead box A3 and is known to regulate the expression of genes involved in gluconeogenesis in hepatocytes.
  • the amino acid sequence of human FOXA3 protein is registered, for example, in GenBank Accession No. NP_004488 (SEQ ID NO: 3), and the amino acid sequence of mouse Foxa3 protein is registered, for example, in GenBank Accession No. NP_032286 (SEQ ID NO: 4).
  • it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence.
  • nucleotide sequence encoding human FOXA3 protein is registered in, for example, GenBank Accession No. NM_004497, and the nucleotide sequence encoding mouse Foxa3 protein is registered in, for example, GenBank Accession No. NM_008260.
  • Cebpa is called CCAAT/enhancer-binding protein alpha and is known to regulate hepatocyte differentiation and glucose/lipid metabolism in hepatocytes.
  • the amino acid sequence of human CEBPA protein is registered, for example, in GenBank Accession No. NP_004355 (SEQ ID NO: 5), and the amino acid sequence of mouse Cebpa protein is registered, for example, in GenBank Accession No. NP_031704 (SEQ ID NO: 6).
  • it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence.
  • the nucleotide sequence encoding the human CEBPA protein is registered, for example, in GenBank Accession No. NM_004364, and the nucleotide sequence encoding the mouse Cebpa protein is registered, for example, in GenBank Accession No. NM_007678.
  • Cebpd is called CCAAT/enhancer-binding protein delta, which is located downstream of the IL-6 signal in hepatocytes and is known to be involved in the acute phase reaction.
  • the amino acid sequence of human CEBPD protein is registered, for example, in GenBank Accession No. NP_005186 (SEQ ID NO: 7), and the amino acid sequence of mouse Cebpd protein is registered, for example, in GenBank Accession No. NP_031705 (SEQ ID NO: 8).
  • it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences.
  • the nucleotide sequence encoding the human CEBPD protein is registered, for example, in GenBank Accession No. NM_005195, and the nucleotide sequence encoding the mouse Cebpd protein is registered, for example, in GenBank Accession No. NM_007679.
  • Hnf6 is called Hepatocyte nuclear factor 6 and is known to regulate hepatocyte differentiation and glucose/lipid metabolism in hepatocytes.
  • the amino acid sequence of human HNF6 protein is registered, for example, in GenBank Accession No. NP_004489 (SEQ ID NO: 9), and the amino acid sequence of mouse Hnf6 protein is registered, for example, in GenBank Accession No. NP_032288 (SEQ ID NO: 10).
  • it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence.
  • the nucleotide sequence encoding the human HNF6 protein is registered in, for example, GenBank Accession No. NM_004498, and the nucleotide sequence encoding the mouse Hnf6 protein is registered in, for example, GenBank Accession No. NM_008262.
  • Onecut2 is called Onecut domain family member2 and is known to be involved in hepatocyte differentiation and metabolism such as inducing the expression of Foxa2 gene.
  • the amino acid sequence of human ONECUT2 protein is registered, for example, in GenBank Accession No. NP_004843 (SEQ ID NO: 11), and the amino acid sequence of mouse Onecut2 protein is registered, for example, in GenBank Accession No. NP_919244 (SEQ ID NO: 12).
  • it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence.
  • nucleotide sequence encoding the human ONECUT2 protein is registered, for example, in GenBank Accession No. NM_004852, and the nucleotide sequence encoding the mouse Onecut2 protein is registered, for example, in GenBank Accession No. NM_194268.
  • Myc is introduced.
  • the amino acid sequence of human MYC protein is registered in, for example, GenBank Accession No. NP_002458 (SEQ ID NO: 13), and the amino acid sequence of mouse Myc protein is registered in, for example, GenBank Accession No. NP_034979 (SEQ ID NO: 14).
  • it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence.
  • the nucleotide sequence encoding the human MYC protein is registered in, for example, GenBank Accession No. NM_002467, and the nucleotide sequence encoding the mouse Myc protein is registered in, for example, GenBank Accession No. NM_010849.
  • the above 6 factors and Myc may be introduced simultaneously, or individual factors or multiple factors may be introduced sequentially.
  • the expression levels (gene level or protein level) of these transcription factors are preferably maintained at 5 times or more, preferably 10 times or more the expression levels in the cells before the introduction. Is more preferable.
  • the period in which the expression level is maintained at the level is, for example, at least 7 days, preferably 10 days, more preferably 14 days, and particularly preferably the period until the differentiation into hepatocytes is completed. This enables highly efficient differentiation induction into mature hepatocytes. It is further preferable that the differentiation into hepatocytes is completed and the differentiation is continued until it is subjected to the experiment. As a result, highly efficient differentiation induction into mature hepatocytes and the function of the obtained mature hepatocyte function may be favorably maintained.
  • the above transcription factor may be introduced into somatic cells at the gene level or may be introduced into somatic cells at the protein level.
  • the transcription factor if the transcription factor is expressed endogenously, the expression of the endogenous transcription factor may be induced. That is, the introduction of a transcription factor involves the induction of an endogenous transcription factor.
  • a vector such as virus, plasmid or artificial chromosome can be introduced into somatic cells by a technique such as lipofection, liposome, microinjection and the like.
  • the introduction may be a transient introduction or a stable introduction.
  • viral vectors include retrovirus vectors, lentivirus vectors, adenovirus vectors, adeno-associated virus vectors, Sendai virus vectors and the like.
  • artificial chromosome vectors include human artificial chromosomes (HAC), yeast artificial chromosomes (YAC), bacterial artificial chromosomes (BAC, PAC), and the like.
  • a plasmid for mammalian cells can be used.
  • the vector can contain regulatory sequences such as a promoter, an enhancer, a ribosome binding sequence, a terminator, and a polyadenylation site so that the transgene can be expressed, and further, if necessary, a drug resistance gene (for example, kanamycin). Resistance gene, ampicillin resistance gene, puromycin resistance gene, etc.), thymidine kinase gene, diphtheria toxin gene and other selectable marker sequences, fluorescent protein, ⁇ -glucuronidase (GUS), FLAG, and other reporter gene sequences.
  • regulatory sequences such as a promoter, an enhancer, a ribosome binding sequence, a terminator, and a polyadenylation site so that the transgene can be expressed, and further, if necessary, a drug resistance gene (for example, kanamycin). Resistance gene, ampicillin resistance gene, puromycin resistance gene, etc.), thymidine kin
  • SV40 promoter As a promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (Rous sarcoma virus) promoter, MoMuLV (Moloney mouse leukemia virus) LTR, HSV-TK (herpes simplex virus thymidine kinase) promoter, EF- ⁇ promoter, CAG Examples are a promoter and a TRE promoter (CMV minimal promoter having a Tet response element in which the tetO sequence is continuous 7 times).
  • RNA When introduced in the form of RNA, it may be introduced into pluripotent stem cells by a technique such as electroporation, lipofection, or microinjection.
  • the expression of factor 6 be sustained for at least 7 days, preferably 10 days, more preferably 14 days, and particularly preferably a period until the differentiation into hepatocytes is completed. This enables highly efficient differentiation induction into mature hepatocytes. Furthermore, it may be maintained until the differentiation is completed and the experiment is performed. Thereby, the function of the obtained mature hepatocytes may be favorably maintained.
  • the culture medium used in the step of culturing the cells into which the transcription factor has been introduced is not particularly limited, but can be prepared by using a medium used for culturing animal cells as a basal medium and adding appropriate components.
  • the basal medium includes, for example, Iscove's Modified Dulbecco's Medium (IMDM) medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, ⁇ MEM medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, RPMI 1640 medium, Fischer's medium. , Neurobasal Medium (Life Technologies) and mixed media thereof.
  • a medium for hepatocytes such as HCM BulletKit Medium, William's E Medium (Thermo Fisher Scientific), Leibovitz's L-15 Medium (Thermo Fisher Scientific) can also be preferably used.
  • the medium may contain serum or may be serum-free.
  • the basal medium may include, for example, albumin, insulin, transferrin, selenium, fatty acids, trace elements, 2-mercaptoethanol, thiolglycerol, lipids, amino acids, L-glutamine, non-essential amino acids, vitamins, growth factors, low levels.
  • cytokines include hepatocyte growth factor (HGF) and oncostatin M (OSM), which are added at a concentration of 1 to 100 ng/mL, for example.
  • HGF hepatocyte growth factor
  • OSM oncostatin M
  • a TGF ⁇ receptor inhibitor is a substance that inhibits the signal transduction from the binding of TGF ⁇ to the receptor and subsequent to SMAD, a substance that inhibits the binding to the ALK family of receptors, or a phosphorylation of SMAD by the ALK family.
  • substances that inhibit oxidation include Lefty-1 (NCBI Accession No. is exemplified by mouse: NM_010094 and human: NM_020997), SB431542, SB202190 (above, RKLindemann et al., Mol. Cancer, 2003).
  • the TGF ⁇ receptor inhibitor used in the present invention may preferably be A83-01.
  • the concentration of A83-01 in the culture medium is not particularly limited as long as it is a concentration that inhibits ALK5, and is, for example, 500 nM to 5 ⁇ M.
  • the culture may be adherent culture or suspension culture, and can be appropriately selected depending on the type of somatic cells used.
  • suspension culture it is desirable to culture cells by forming aggregates (also referred to as spheres) in a culture container in a non-adhesive state.
  • aggregates also referred to as spheres
  • Such culture is not particularly limited, but it is possible to adhere to cells.
  • Culture vessel that is not artificially treated for example, coating treatment with extracellular matrix etc. for the purpose of improving the property, or treatment that artificially suppresses adhesion (for example, polyhydroxyethyl methacrylic acid (poly-HEMA))
  • a culture vessel coated with a nonionic surface-active polyol for example, a nonionic surface-active polyol (Pluronic F-127 etc.) or a phospholipid-like structure (for example, a water-soluble polymer (Lipidure) containing 2-methacryloyloxyethylphosphorylcholine as a constituent unit)
  • a nonionic surface-active polyol for example, a phospholipid-like structure
  • Lipidure a water-soluble polymer containing 2-methacryloyloxyethylphosphorylcholine as a constituent unit
  • the extracellular matrix is a supramolecular structure existing outside the cell, and may be of natural origin or artificial (recombinant). Examples thereof include substances such as polylysine, polyornithine, collagen, proteoglycan, fibronectin, hyaluronic acid, tenascin, entactin, elastin, fibrillin and laminin, and fragments thereof.
  • These extracellular matrices may be used in combination, and may be, for example, preparations from cells such as BD MatrigelTM.
  • Culture conditions are not particularly limited, but for example, about 37°C to about 42°C, preferably about 37 to about 39°C. Culturing is carried out in an atmosphere of CO 2 -containing air, and the CO 2 concentration is preferably about 2-5%. In addition, those skilled in the art can appropriately determine the culture period while monitoring the cell number and the like.
  • the number of days is not particularly limited, but is, for example, 10 days or more, 12 days or more, 14 days or more, 16 days or more, 18 days or more, 20 days or more, 25 days or more.
  • Hepatocytes can be evaluated by measuring the expression of albumin and the like.
  • Hepatocytes may be obtained as a cell population containing other cell types or may be purified.
  • the cell population obtained by the method of the present invention contains, for example, 3% or more, 5% or more, 10% or more or 20% or more hepatocytes, but preferably 50% or more, 60% or more, 70% or more, or Contains 80% or more hepatocytes.
  • a method of concentrating by using the method is exemplified.
  • the hepatocytes obtained in the present invention have the function as adult hepatocytes by introducing the above 6 factors, preferably Myc.
  • hepatocytes have, in addition to albumin secretion, one or more of hepatocyte functions such as cytochrome P450 activity, drug metabolism, glycogen accumulation, low-density lipoprotein (LDL) uptake, ammonia metabolism and urea synthesis.
  • the present invention provides a cell preparation for regenerative medicine, which comprises the hepatocytes obtained by the above-mentioned method and is used for treatment of liver diseases and the like.
  • a method of administering a cell preparation to a patient for example, a method of forming the obtained hepatocytes into a sheet and affixing it to the patient's liver, suspending the obtained hepatocytes in physiological saline, etc.
  • Examples thereof include a method of direct transplantation, a method of three-dimensionally culturing on a scaffold composed of Matrigel and the like, and a method of transplanting the obtained hepatocyte mass.
  • the number of hepatocytes contained in the cell preparation is not particularly limited as long as the graft can be engrafted after administration, and may be adjusted appropriately according to the size of the affected area or the size of the body.
  • the liver cells prepared by the method of the present invention can be used to test the metabolism of a test substance in vitro.
  • the metabolism test of the test substance includes, for example, the step of (i) contacting the test substance with the hepatocytes obtained by the method of the present invention, and (ii) detecting a metabolite of the test substance.
  • You can The "contact" in step (i) can be performed by adding a test substance to the medium for culturing hepatocytes produced by the present invention.
  • the timing of addition of the test compound is not particularly limited. Therefore, after starting the culture in the medium not containing the test substance, the test substance may be added at a certain point in time, or the culture may be started in advance in the medium containing the test substance.
  • the test substance includes, for example, existing components such as pharmaceuticals and nutritional foods or candidate components.
  • the test substance may be derived from a natural product or may be synthetic. In the latter case, an efficient assay system can be constructed by using, for example, a combinatorial synthesis method.
  • the period of contact with the test substance can be set arbitrarily.
  • the contact period is, for example, 10 minutes to 3 days, preferably 1 hour to 1 day.
  • the contact may be divided into a plurality of times.
  • the metabolism of the test substance examples include chemical modification of the test substance by hydrolysis of ester and the like, oxidation reaction (particularly oxidation by cytochrome P450), reduction reaction, and the like. Can be detected.
  • the expected metabolite is qualitatively or quantitatively measured using the obtained culture solution as a sample.
  • the measurement method may be selected appropriately depending on the test substance and its predicted metabolite, and examples thereof include mass spectrometry, liquid chromatography, immunological methods (eg, fluorescence immunoassay (FIA method), Enzyme-linked immunosorbent assay (EIA method) etc. can be adopted.
  • the amount of metabolism of the test substance can be evaluated according to the amount of metabolites.
  • the hepatocytes obtained in the present invention can be used to confirm the induction of expression of drug-metabolizing enzymes (eg, cytochrome, UGT).
  • drug-metabolizing enzymes eg, cytochrome, UGT.
  • Expression of drug-metabolizing enzymes can be assessed at the mRNA or protein level. For example, when an increase in the mRNA level of a drug-metabolizing enzyme is observed, it can be determined that “the test substance is suspected of causing a drug interaction”.
  • the toxicity of the test substance can also be tested using the hepatocytes prepared by the method of the present invention.
  • the method comprises (i) a step of contacting a test substance with the hepatocytes obtained by the method of the present invention, (ii) a step of examining the state of the hepatocytes after the step (i), and (iii) (ii) ), the step of evaluating the toxicity of the test substance based on the result of 4).
  • Step (i) is the same as the metabolism of the test substance.
  • step (ii) the state of hepatocytes after contact with the test substance is examined, and the toxicity of the test substance is evaluated in step (iii).
  • the state of hepatocytes can be ascertained by measuring viability, observing cell morphology, measuring liver damage markers (GOT, GPT, etc.) in the culture solution (culture supernatant), and the like. For example, when a decrease in survival rate is observed due to contact with a test substance, it can be determined that "the test substance has liver toxicity". Similarly, when an abnormal cell morphology is observed due to contact with a test substance, or when the amount of a liver damage marker in the culture solution is increased, it can be similarly determined that "the test substance has hepatotoxicity". it can.
  • Quantitative determination may be performed depending on the degree of decrease in survival rate and the amount of liver damage marker.
  • the "reduction in survival rate” is, as a guide, 90% or less, 80% or less, 70% or less, 60% or less with respect to the survival rate of hepatocytes (control cells) not contacted with the test substance.
  • the survival rate may be 50% or less.
  • the "increased amount of liver damage marker” is, as a guide, a marker amount of 110% or more, 120% or more, 130% or less, 140% or more, or 150% or more with respect to the survival rate of control cells. It may be.
  • the TC 50 of the test substance may be analyzed in the step (ii), and in the step (iii), it may be determined that the test substance has a hepatotoxicity with respect to the test substance having a TC50 value of a specific value or less. .. By examining the TC 50 of the test substance for hepatocytes, it is possible to contribute to the prediction of the proper dose of the test substance to the human body.
  • Example 1 Induced hepatocytes (iHeps) were prepared by introducing genes into fibroblasts using a retrovirus vector according to the following procedure, and the obtained iHeps was used to analyze the expression of hepatocyte markers.
  • mice Hnf4a gene, Foxa3 gene, Cebpa gene, Cebpd gene, Hnf6 gene and Onecut2 gene were each incorporated into pMXs plasmid to prepare a retroviral vector for introducing each gene.
  • a retrovirus vector for introducing the Myc gene of mouse was also prepared in the same manner.
  • pMXs was transferred from Professor Toshio Kitamura of the University of Tokyo (Exp Hematol. 2003 Nov;31(11):1007-14.).
  • Virus supernatant preparation 1. Plate-E (Cell Biolabs, Inc., Cat# RV-101) with 10% FBS/DMEM (FBS (Thermo Fisher Scientific), DMEM (Nacalai tesque, Cat# 08459-64)) containing no antibiotics. The cells were suspended in a sol, and seeded (5.0 ⁇ 10 5 cells/1.5 ml/well) on a gelatin-coated 6-well plate (Greiner), and incubated overnight at 37°C, 5% CO 2 .
  • Plat-E was subjected to lipofection of each transcription factor plasmid. 4.5 ⁇ l of FuGENE 6 (Promega, Cat# E2692) was added to 50 ⁇ l of Opti-MEM (Thermo Fisher Scientific, Cat# 31985062) at 37° C., and the mixture was allowed to stand at room temperature for 5 minutes after vortexing. Add 500 ng/ ⁇ l of retroviral vector (pMXs) of each transcription factor to it, leave it at room temperature for 20 minutes, then add to each well of Plat-E prepared in 1 above, and then overnight at 37°C 5% CO 2 Incubated.
  • FuGENE 6 Promega, Cat# E2692
  • Opti-MEM Thermo Fisher Scientific, Cat# 31985062
  • the medium was exchanged with 1.6 ml/well of 10% FBS/DMEM containing no antibiotic, and the mixture was incubated overnight at 37° C. and 5% CO 2 and used as a virus supernatant.
  • MEFs mouse embryonic fibroblasts
  • E13.5 Obtained from a fetus of a pregnant mouse (C57BL/6J). Without passage, Cell Reservoir One (Nacalai tesque, Cat# 07485-44) was used as a cell preservation solution and frozen and preserved at -80°C.
  • iHep induction 1 Viral transduction The day before (day-1), the MEFs that had been cryopreserved were put to sleep and put on a 12-well plate (Greiner) coated with Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat# 354230). Seeding (5.0 ⁇ 10 4 cells/well) was performed.
  • HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ ml, A83-01 (Merck, Cat# 616454-2MGCN) 1 ⁇ M), and the medium was exchanged once every two days. It was subjected to experiments such as gene expression analysis around Day 25.
  • Cdx2 is a mid-intestinal/hind-gut marker gene (intestinal marker gene liver derived from posterior foregut), and iHeps produced from two factors in the previous report (Morris et al Cell 2014 Aug 14;158(4):889-902) Although it was shown that the cells were Cdx2 positive cells, the expression of Cdx2 was markedly reduced by using 6 factors. Although Afp is a hepatoblast marker, the use of factor 6 also reduced Afp expression, indicating that iHeps closer to that of more mature hepatocytes was obtained.
  • FIG. 4 shows the results of immunostaining of iHeps (6TF-Myc iHeps) obtained by forced expression of 6 factors and Myc. It can be seen that the albumin-positive epithelial-like cells (6TF-Myc iHeps) spread like cobblestones.
  • 6TF-Myc iHeps was used to measure the amount of albumin secreted into the medium for 24 hours by ELISA.
  • the amount of Albumin secretion was corrected and shown by the number of cells that become Albumin-positive by immunostaining.
  • Myc was combined with 6 factors, the amount of albumin secretion in the obtained induced hepatocytes was remarkably higher than that of 6 factors, which was comparable to that of primary cultured hepatocytes.
  • 6TF-Myc iHeps was used to examine the expression induction of the CYP gene for various drugs.
  • Cif3a11 was induced in Rifampicin
  • Cyp1a2 was induced in Omeprazole
  • Cyp2b10 was induced in TCP0B0P
  • the obtained induced hepatocytes activated CYP corresponding to the drug and had a response profile of normal hepatocytes. I found out.
  • Example 2 Examination of necessity of 6 factors 2 to 6 factors out of 6 factors were introduced by the following procedure to induce hepatocyte differentiation.
  • iHep induction 1. Viral transduction The day before (day-1), the MEFs that had been cryopreserved were put to sleep and placed on a 12-well plate (Greiner) coated with Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat# 354230). Seeding (5.0 ⁇ 10 4 cells/well). 2.
  • HCM BulletKit Medium Longza, Cat# CC-3198
  • HGF Peprotech, Cat# 100-39
  • OSM Peprotech, Cat# 300-10
  • hepatocyte differentiation was induced by the following procedure using human somatic cells.
  • iHep induction 1. Viral transduction The day before (day-1), the frozen HDF-Slc7a1 (Yamakawa, Stem Cells 2016; gift from Okita lab.) was put to sleep, and Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat Seed (5.0 ⁇ 10 4 cells/well) on a 12-well plate (Greiner) coated with #354230). 2.
  • Incubation was carried out overnight at 37°C, 5% CO 2 to complete the virus. 5.
  • the next day (day 1) the medium was replaced (10% FBS/DMEM). 6.
  • the medium was exchanged (10% FBS/DMEM). 7.
  • reseeding was performed at a ratio of 1:10 on a 12-well plate coated with Matrigel.
  • Hepatic specification medium based on 10% FBS/DMEM/F12, GlutaMAX (DMEM/F12, GlutaMAX (Thermo Fisher Scientific, Cat# 10565042) 360 ml, FBS 40 ml, Nicotinamide (STEMCELL Technologies, Cat# 07154) 10 mM , ITS-X (Thermo Fisher Scientific, Cat# 51500056) 400 ⁇ l, Dexamethasone (Merck, Cat# D4902-25MG) 0.1 ⁇ M, 2-Mercaptoethanol (Thermo Fisher Scientific, Cat# 21985023) 50 ⁇ M, Penicillin-Streptomycin (Thermo Fisher The medium was replaced with Scientific, Cat# 15140122) 2 ml). 8.
  • HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ ml) and the medium was exchanged once every two days. It was used for experiments around Day 20.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Analytical Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Plant Pathology (AREA)
  • Oral & Maxillofacial Surgery (AREA)
  • Botany (AREA)
  • Food Science & Technology (AREA)
  • Physiology (AREA)
  • General Physics & Mathematics (AREA)

Abstract

A method for producing hepatocytes including a step for preparing somatic cells into which Hnf4a (hepatocyte nuclear factor 4 alpha), Foxa3 (forkhead box A3), Cebpa (CCAAT/enhancer-binding protein alpha), Cebpd (CCAAT/enhancer-binding protein delta), Hnf6 (hepatocyte nuclear factor 6), and onecut2 (onecut domain family member 2) have been introduced and a step for culturing the somatic cells for induction into hepatocytes.

Description

肝細胞の製造方法Hepatocyte production method
本発明は細胞関連技術に関し、より詳しくは機能的に成熟した哺乳動物肝細胞の製造方法に関する。 The present invention relates to cell-related technology, and more particularly to a method for producing functionally mature mammalian hepatocytes.
現在、人工多能性幹(iPS)細胞や胚性幹(ES)細胞から分化誘導される肝細胞や、線維芽細胞など肝細胞以外の細胞種に転写因子の組み合わせを導入することによって作製される誘導肝細胞を用いた、肝細胞移植療法(肝臓再生医療)、肝臓の再構築、肝疾患モデル作製、創薬、薬剤肝毒性評価系の構築などが期待されている。しかし、現在までのところ、機能的に未熟な胎児期の肝細胞に相当する肝細胞の作製にとどまり、成体の体内に存在する肝細胞に近い機能を有する肝細胞を作製することは可能となっていない。 Currently produced by introducing a combination of transcription factors into hepatocytes that are induced to differentiate from induced pluripotent stem (iPS) cells and embryonic stem (ES) cells, and into cell types other than hepatocytes such as fibroblasts. Hepatocyte transplantation therapy (liver regenerative medicine) using induced hepatocytes, liver reconstruction, liver disease model preparation, drug discovery, and drug hepatotoxicity evaluation system construction are expected. However, to date, it has become possible to produce hepatocytes that are functionally immature and correspond to hepatocytes in the fetal period, and to produce hepatocytes that have a function similar to that of hepatocytes present in the adult body. Not not.
非特許文献1には、マウス胎仔および成体線維芽細胞にHnf4aと、Foxa1、Foxa2またはFoxa3の遺伝子を導入することで誘導肝細胞を作製したことが報告されている。
非特許文献2には、マウス成体線維芽細胞にGata4、Hnf1aおよびFoxa3の遺伝子を導入することで誘導肝細胞を作製したことが報告されている。
非特許文献3には、マウス胎仔線維芽細胞にHnf1bおよびFoxa3の遺伝子を導入することで誘導肝細胞を作製したことが報告されている。
非特許文献4には、ヒト線維芽細胞にFOXA3、HNF1AおよびHNF4Aの遺伝子を導入することで誘導肝細胞を作製したことが報告されている。
非特許文献5には、ヒト線維芽細胞にHNF1A、HNF4A、HNF6、ATF5、PROX1およびCEBPAの遺伝子を導入することで誘導肝細胞を作製したことが報告されている。
しかしながら、これまでに報告されている誘導肝細胞は肝マーカーの発現量が少なく、機能的に未熟で、上記のような再生医療等の目的に使用できるものではなかった。
Non-patent document 1 reports that induced hepatocytes were prepared by introducing Hnf4a and the genes of Foxa1, Foxa2, or Foxa3 into mouse fetal and adult fibroblasts.
Non-Patent Document 2 reports that induced hepatocytes were produced by introducing Gata4, Hnf1a and Foxa3 genes into adult mouse fibroblasts.
Non-Patent Document 3 reports that induced hepatocytes were prepared by introducing Hnf1b and Foxa3 genes into mouse fetal fibroblasts.
Non-Patent Document 4 reports that induced hepatocytes were produced by introducing FOXA3, HNF1A and HNF4A genes into human fibroblasts.
Non-Patent Document 5 reports that induced hepatocytes were prepared by introducing genes for HNF1A, HNF4A, HNF6, ATF5, PROX1 and CEBPA into human fibroblasts.
However, the induced hepatocytes that have been reported so far have a low expression amount of a liver marker, are functionally immature, and cannot be used for the purpose of regenerative medicine as described above.
本発明は機能的に成熟した肝細胞を効率よく製造する方法を提供することを課題とする。 An object of the present invention is to provide a method for efficiently producing functionally mature hepatocytes.
上記課題を解決するために、発明者らは、マウス成体肝細胞に発現する遺伝子の発現プロファイルを探索し、iPS干渉法(Hikichi T. 2013 Apr 16;110(16):6412-7)を用いて成熟肝細胞を誘導する転写因子の組み合わせの絞り込みを行った。その結果、Hnf4a(Hepatocyte nuclear factor 4 alpha)、Foxa3(Forkhead Box A3)、Cebpa(CCAAT/enhancer-binding protein alpha)、Cebpd(CCAAT/enhancer-binding protein delta)、Hnf6(Hepatocyte nuclear factor 6)およびOnecut2(One cut domain family member 2)の6種類の転写因子の組み合わせ(6因子)の遺伝子導入により、マウスおよびヒトの線維芽細胞から最も機能的に成熟した肝細胞を作製することに成功した。また、前述の6因子に加えMycを導入すること、およびTGF-β受容体阻害剤を用いて培養することにより、機能的成熟度をさらに上げることができることを見出した。以上の発見に基づき、本発明を完成させた。 In order to solve the above problems, the inventors searched for an expression profile of a gene expressed in adult mouse hepatocytes, and used iPS interference method (Hikichi T. 2013 2013 Apr 16;110(16):6412-7). The combination of transcription factors that induce mature hepatocytes was narrowed down. As a result, Hnf4a (Hepatocyte nuclear factor4alpha), Foxa3 (Forkhead Box A3), Cebpa (CCAAT/enhancer-binding protein alpha), Cebpd (CCAAT/enhancer-binding protein delta), Hnf6 (Hepatocyte nuclear factor6) and Onecut2. We succeeded in producing the most functionally mature hepatocytes from mouse and human fibroblasts by gene transfer of a combination of 6 types of transcription factors (6 factors) (One cut domain family member 2). It was also found that the functional maturity can be further increased by introducing Myc in addition to the above-mentioned 6 factors and culturing using a TGF-β receptor inhibitor. The present invention has been completed based on the above findings.
したがって、本発明の要旨は以下のとおりである。
[1]Hnf4a、Foxa3、Cebpa、Cebpd、Hnf6およびOnecut2が導入された体細胞を用意する工程、および当該体細胞を培養して肝細胞へと誘導する工程、を含む、肝細胞の製造方法。
[2]体細胞が線維芽細胞である、[1]に記載の肝細胞の製造方法。
[3]体細胞がヒトまたはマウス由来である、[1]または[2]に記載の肝細胞の製造方法。
[4]Hnf4a、Foxa3、Cebpa、Cebpd、Hnf6およびOnecut2がヒトまたはマウス由来である、[1]~[3]のいずれかに記載の肝細胞の製造方法。
[5]Hnf4a、Foxa3、Cebpa、Cebpd、Hnf6およびOnecut2がレトロウイルスベクターを用いて導入された、[1]~[4]のいずれかに記載の肝細胞の製造方法。
[6]前記体細胞は、さらにMycが導入されている、[1]~[5]のいずれかに記載の肝細胞の製造方法。
[7]前記培養工程が20日以上行われる、[1]~[6]のいずれかに記載の肝細胞の製造方法。
[8]前記培養工程において培地はTGFβ(Transforming growth factor beta)受容体阻害剤を含む、[1]~[7]のいずれかに記載の肝細胞の製造方法。
[9]TGFβ受容体阻害剤はA83-01である、請求項8に記載の肝細胞の製造方法。
[10] [1]~[9]のいずれかに記載の方法によって製造された肝細胞。
[11] [10]に記載の肝細胞を含む、再生医療用細胞製剤。
[12]下記工程を(i)-(iii)の順番で含む、被検物質の毒性評価方法;
(i)[1]~[9]のいずれかに記載の方法によって製造された肝細胞に被検物質を接触させる工程、
(ii)前記肝細胞の生存率または培養上清中の肝障害マーカー量を測定する工程、および
(iii)前記被検物質と接触させた肝細胞の生存率が、被検物質と接触させなかった肝細胞(対照細胞)の生存率よりも低値、または、前記被検物質と接触させた肝細胞の培養上清中の肝障害マーカー量が、対照細胞の培養上清中の肝障害マーカー量よりも高値である被検物質を、肝毒性を有すると評価する工程。
 
Therefore, the gist of the present invention is as follows.
[1] A method for producing hepatocytes, which comprises a step of preparing somatic cells introduced with Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2, and a step of culturing the somatic cells to induce hepatocytes.
[2] The method for producing hepatocytes according to [1], wherein the somatic cells are fibroblasts.
[3] The method for producing hepatocytes according to [1] or [2], wherein the somatic cells are of human or mouse origin.
[4] The method for producing hepatocytes according to any one of [1] to [3], wherein Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are derived from human or mouse.
[5] The method for producing hepatocytes according to any one of [1] to [4], wherein Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are introduced using a retrovirus vector.
[6] The method for producing hepatocytes according to any one of [1] to [5], wherein Myc is further introduced into the somatic cells.
[7] The method for producing hepatocytes according to any one of [1] to [6], wherein the culturing step is performed for 20 days or more.
[8] The method for producing hepatocytes according to any one of [1] to [7], wherein in the culture step, the medium contains a TGFβ (Transforming growth factor beta) receptor inhibitor.
[9] The method for producing hepatocytes according to claim 8, wherein the TGFβ receptor inhibitor is A83-01.
[10] A hepatocyte produced by the method according to any one of [1] to [9].
[11] A cell preparation for regenerative medicine containing the hepatocyte according to [10].
[12] A method for evaluating toxicity of a test substance, which comprises the following steps in the order of (i)-(iii):
(I) a step of contacting a test substance with hepatocytes produced by the method according to any one of [1] to [9],
(Ii) the step of measuring the survival rate of the hepatocytes or the amount of the liver injury marker in the culture supernatant, and (iii) the survival rate of the hepatocytes contacted with the test substance does not contact with the test substance. Lower than the survival rate of the hepatocytes (control cells), or the amount of the liver injury marker in the culture supernatant of the hepatocytes contacted with the test substance is the liver injury marker in the culture supernatant of the control cells. A step of evaluating a test substance having a higher value than the amount as having hepatotoxicity.
本発明により機能的に成熟した哺乳動物肝細胞を効率よく製造することができる。
本発明の方法により得られる哺乳動物成熟肝細胞を用いることにより、難治性肝疾患に対する肝細胞移植療法(肝臓再生医療)、肝疾患モデル作製、肝細胞を用いた創薬、薬剤肝毒性評価等をこれまでにない高い精度で実施可能となる。また、本発明の方法により得られる肝細胞は、肝細胞の分化および成熟化機構の解明の目的にも使用できる。
According to the present invention, functionally mature mammalian hepatocytes can be efficiently produced.
By using the mature mammalian hepatocytes obtained by the method of the present invention, hepatocyte transplantation therapy for intractable liver diseases (liver regenerative medicine), liver disease model preparation, drug development using hepatocytes, drug hepatotoxicity evaluation, etc. Can be performed with higher accuracy than ever before. The hepatocytes obtained by the method of the present invention can also be used for the purpose of elucidating the differentiation and maturation mechanism of hepatocytes.
2因子(Hnf4a+Foxa3)または6因子(Hnf4a+Foxa3+Cebpa+Cebpd+Hnf6+Onecut2)を導入して誘導された肝細胞における各遺伝子の発現量を示すグラフ。The graph which shows the expression level of each gene in the hepatocyte induced by introducing 2 factors (Hnf4a+Foxa3) or 6 factors (Hnf4a+Foxa3+Cebpa+Cebpd+Hnf6+Onecut2). 6因子を導入して誘導された肝細胞をA83-01のあるなしで培養した時のVimentin、Cola1(コラーゲン1a1)、Afp(Alpha-fetoprotein)の発現量を示すグラフ。6 is a graph showing the expression levels of Vimentin, Cola1 (collagen 1a1), and Afp (Alpha-fetoprotein) when hepatocytes induced by introducing 6 factors were cultured with or without A83-01. 6因子を導入して誘導された肝細胞をA83-01のあるなしで培養した時のアルブミンの分泌量を示すグラフ。6 is a graph showing the amount of albumin secreted when hepatocytes induced by introducing 6 factors were cultured with or without A83-01. 6因子およびMycの強制発現により得られた誘導肝細胞の免疫染色結果を示す図(写真)。略称は次の通り:Alb, Albumin. E-cad, E-cadherin. Ho, Hoechst 33342。The figure (photograph) which shows the immunostaining result of the induced hepatocyte obtained by forced expression of 6 factors and Myc. Abbreviations are as follows: Alb, Albumin. E-cad, E-cadherin. Ho, Hoechst 33342. 培地中への24時間あたりのAlbumin分泌量をELISAにより測定した結果を示す図。Albumin分泌量は、免疫染色でAlbumin陽性となる細胞数で補正した。N=3で行い、平均値±標準偏差で示した。**P < 0.01(one-way ANOVA with Tukey’s correction)。The figure which shows the result of having measured the albumin secretion amount in a culture medium per 24 hours by ELISA. Albumin secretion was corrected by the number of cells that became Albumin-positive by immunostaining. N=3, and the mean value ± standard deviation is shown. **P<0.01 (one-way ANOVA with Tukey’s correction). 各細胞においてCyp3a11の酵素活性を比較した結果を示す図。N=3で行い、平均値±標準偏差で示した。*P < 0.05, **P < 0.01(one-way ANOVA with Tukey’s correction)The figure which shows the result of having compared the enzyme activity of Cyp3a11 in each cell. N=3, and the mean value ± standard deviation is shown. *P<0.05, **P<0.01 (one-way ANOVA with Tukey’s correction) 6因子+Mycを導入して誘導された肝細胞において、各薬剤で刺激した時の、CYP遺伝子の発現変動を示すグラフ。矢印は発現量が顕著に増加した遺伝子を指す。6 is a graph showing CYP gene expression fluctuations in hepatocytes induced by introducing 6 factors+Myc when stimulated with each drug. The arrow indicates a gene whose expression level is remarkably increased. 2~6因子を導入して誘導された肝細胞におけるCps1(Carbamoylphosphate synthase I)遺伝子の発現量(n = 3, mean ± SD)を示すグラフ。βアクチン(Actb)で標準化したのち、2因子(Hnf4a+Foxa3)を導入して誘導された肝細胞における値を1として表した。Graph showing the expression level (n = 3, mean ± SD) of Cps1 (Carbamoylphosphate synthase I) gene in hepatocytes induced by introducing 2 to 6 factors. The value in hepatocytes induced by introducing 2 factors (Hnf4a+Foxa3) after standardization with β-actin (Actb) was expressed as 1. 6因子およびMycをヒト細胞に導入することにより得られた誘導肝細胞の免疫染色結果を示す図(写真)。The figure (photograph) which shows the immunostaining result of the induced hepatocyte obtained by introduce|transducing 6 factors and Myc into a human cell.
本発明の肝細胞の製造方法は、Hnf4a、Foxa3、Cebpa、Cebpd、Hnf6およびOnecut2が導入された体細胞を用意する工程、および当該体細胞を培養して肝細胞へと誘導する工程、を含む。 The method for producing hepatocytes of the present invention includes a step of preparing somatic cells introduced with Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2, and a step of culturing the somatic cells to induce hepatocytes. ..
<6因子導入体細胞を用意する工程>
ここで、用意するとは、体細胞に6因子を導入して新たに用意する場合に限られず、6因子の一部が既に導入された体細胞に残りの因子すべてを導入する場合や、予め6因子が導入された体細胞を調達してくる場合も含む。
<Step of preparing 6-factor introduced somatic cells>
Here, the term “preparation” is not limited to the case where 6 factors are introduced into somatic cells and newly prepared, and the case where all the remaining factors are introduced into somatic cells in which a part of 6 factors has already been introduced, or 6 It also includes the case of procuring somatic cells into which the factor has been introduced.
<体細胞>
本発明において、体細胞は肝細胞以外の体細胞であれば特に限定されないが、例えば線維芽細胞、上皮細胞(皮膚表皮細胞、口腔粘膜上皮細胞、気道粘膜上皮細胞、腸管粘膜上皮細胞など)、表皮細胞、歯肉細胞(歯肉線維芽細胞、歯肉上皮細胞)、歯髄細胞、白色脂肪細胞、 皮下脂肪、内臓脂肪、筋肉、血液細胞などが挙げられる。また、間葉系幹細胞(Mesenchymal stem cell: MSC)、腸幹細胞、皮膚幹細胞、毛包幹細胞、色素細胞幹細胞などの体性幹細胞から分化誘導し、あるいは脱分化させ、あるいはリプログラミングさせて作製した体細胞も挙げられる。 
<Somatic cells>
In the present invention, somatic cells are not particularly limited as long as they are somatic cells other than hepatocytes, for example fibroblasts, epithelial cells (skin epidermal cells, oral mucosal epithelial cells, respiratory mucosal epithelial cells, intestinal mucosal epithelial cells, etc.), Examples thereof include epidermal cells, gingival cells (gingival fibroblasts, gingival epithelial cells), pulp cells, white adipocytes, subcutaneous fat, visceral fat, muscle, blood cells and the like. In addition, a body prepared by inducing or dedifferentiating or reprogramming somatic stem cells such as mesenchymal stem cells (MSCs), intestinal stem cells, skin stem cells, hair follicle stem cells, and pigment cell stem cells Also included are cells.
体細胞の由来は、成体であっても小児であっても胎児であってもよい。体細胞は、ヒト、マウス、ラット、ブタ等のヒト以外の哺乳動物から単離されたものであっても、その継代培養細胞であってもよい。線維芽細胞としては、例えば、胎児線維芽細胞、尾先端部由来線維芽細胞、心臓線維芽細胞、***線維芽細胞、皮膚線維芽細胞、肺線維芽細胞等を用いることができる。 Somatic cells may be of adult, pediatric or fetal origin. Somatic cells may be those isolated from mammals other than humans such as humans, mice, rats and pigs, or subcultured cells thereof. As the fibroblasts, for example, fetal fibroblasts, tail-end fibroblasts, cardiac fibroblasts, foreskin fibroblasts, skin fibroblasts, lung fibroblasts, etc. can be used.
また、得られる肝細胞を移植用細胞の材料として用いる場合、拒絶反応が起こらないという観点から、当該肝細胞の製造に用いる体細胞は、移植先の個体のHLA遺伝子型が同一または実質的に同一である体細胞であることが望ましい。ここで、「実質的に同一」とは、移植した細胞に対して免疫抑制剤により免疫反応が抑制できる程度にHLA遺伝子型が一致していることであり、例えば、移植先の個体とHLA-A、HLA-BおよびHLA-DRの3遺伝子座またはHLA-Cを加えた4遺伝子座が一致するHLA型を有する体細胞である。 Further, when the obtained hepatocytes are used as a material for cells for transplantation, the somatic cells used for the production of the hepatocytes are the same or substantially the same HLA genotype of the transplant destination individual from the viewpoint that a rejection reaction does not occur. It is desirable that the somatic cells are the same. Here, “substantially the same” means that the HLA genotypes are matched to the extent that the immunoreactivity of the transplanted cells can be suppressed by the immunosuppressant, and for example, the transplant recipient individual and HLA- It is a somatic cell having an HLA type in which 3 loci of A, HLA-B and HLA-DR or 4 loci including HLA-C are matched.
<6因子>
6因子として、Hnf4a、Foxa3、Cebpa、Cebpd、Hnf6およびOnecut2を用いる。
<6 factors>
Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are used as 6 factors.
Hnf4aはHepatocyte nuclear factor 4 alphaと呼ばれ、糖代謝や脂質代謝、薬物代謝など、多くの肝細胞機能に関わる遺伝子の発現を調節することが知られている。ヒトHNF4Aタンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_849180(配列番号1)に登録されており、マウスHNF4aタンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_032287(配列番号2)に登録されている。ただし、これら特定のアミノ酸配列を有するタンパク質には限定されず、転写因子としての機能を有する限り、これらの配列と90%以上、好ましくは95%以上、より好ましくは98%以上の同一性を有するアミノ酸配列を有するタンパク質でもよい。ヒトHNF4aタンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_178849に登録されており、マウスHNF4aタンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_008261に登録されている。 Hnf4a is called Hepatocyte nuclear factor4alpha, and is known to regulate the expression of many genes involved in hepatocyte function, such as glucose metabolism, lipid metabolism, and drug metabolism. The amino acid sequence of human HNF4A protein is registered, for example, in GenBank Accession No. NP_849180 (SEQ ID NO: 1), and the amino acid sequence of mouse HNF4a protein is registered, for example, in GenBank Accession No. NP_032287 (SEQ ID NO: 2). However, it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence. The base sequence encoding the human HNF4a protein is registered, for example, in GenBank Accession No. NM_178849, and the base sequence encoding the mouse HNF4a protein is registered, for example, in GenBank Accession No. NM_008261.
Foxa3はForkhead box A3と呼ばれ、肝細胞においては糖新生に関わる遺伝子などの発現を調節することが知られている。ヒトFOXA3タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_004488(配列番号3)に登録されており、マウスFoxa3タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_032286(配列番号4)に登録されている。ただし、これら特定のアミノ酸配列を有するタンパク質には限定されず、転写因子としての機能を有する限り、これらの配列と90%以上、好ましくは95%以上、より好ましくは98%以上の同一性を有するアミノ酸配列を有するタンパク質でもよい。ヒトFOXA3タンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_004497に登録されており、マウスFoxa3タンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_008260に登録されている。 Foxa3 is called Forkhead box A3 and is known to regulate the expression of genes involved in gluconeogenesis in hepatocytes. The amino acid sequence of human FOXA3 protein is registered, for example, in GenBank Accession No. NP_004488 (SEQ ID NO: 3), and the amino acid sequence of mouse Foxa3 protein is registered, for example, in GenBank Accession No. NP_032286 (SEQ ID NO: 4). However, it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence. The nucleotide sequence encoding human FOXA3 protein is registered in, for example, GenBank Accession No. NM_004497, and the nucleotide sequence encoding mouse Foxa3 protein is registered in, for example, GenBank Accession No. NM_008260.
CebpaはCCAAT/enhancer-binding protein alphaと呼ばれ、肝細胞分化や肝細胞における糖・脂質代謝を調節することが知られている。ヒトCEBPA タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_004355(配列番号5)に登録されており、マウスCebpa タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_031704(配列番号6)に登録されている。ただし、これら特定のアミノ酸配列を有するタンパク質には限定されず、転写因子としての機能を有する限り、これらの配列と90%以上、好ましくは95%以上、より好ましくは98%以上の同一性を有するアミノ酸配列を有するタンパク質でもよい。ヒトCEBPAタンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_004364に登録されており、マウスCebpa タンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_007678に登録されている。 Cebpa is called CCAAT/enhancer-binding protein alpha and is known to regulate hepatocyte differentiation and glucose/lipid metabolism in hepatocytes. The amino acid sequence of human CEBPA protein is registered, for example, in GenBank Accession No. NP_004355 (SEQ ID NO: 5), and the amino acid sequence of mouse Cebpa protein is registered, for example, in GenBank Accession No. NP_031704 (SEQ ID NO: 6). However, it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence. The nucleotide sequence encoding the human CEBPA protein is registered, for example, in GenBank Accession No. NM_004364, and the nucleotide sequence encoding the mouse Cebpa protein is registered, for example, in GenBank Accession No. NM_007678.
CebpdはCCAAT/enhancer-binding protein deltaと呼ばれ、肝細胞においてはIL-6によるシグナルの下流に位置し、急性期反応に関わることが知られている。ヒトCEBPDタンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_005186(配列番号7)に登録されており、マウスCebpdタンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_031705(配列番号8)に登録されている。ただし、これら特定のアミノ酸配列を有するタンパク質には限定されず、転写因子としての機能を有する限り、これらの配列と90%以上、好ましくは95%以上、より好ましくは98%以上の同一性を有するアミノ酸配列を有するタンパク質でもよい。ヒトCEBPDタンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_005195に登録されており、マウスCebpdタンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_007679に登録されている。 Cebpd is called CCAAT/enhancer-binding protein delta, which is located downstream of the IL-6 signal in hepatocytes and is known to be involved in the acute phase reaction. The amino acid sequence of human CEBPD protein is registered, for example, in GenBank Accession No. NP_005186 (SEQ ID NO: 7), and the amino acid sequence of mouse Cebpd protein is registered, for example, in GenBank Accession No. NP_031705 (SEQ ID NO: 8). However, it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence. The nucleotide sequence encoding the human CEBPD protein is registered, for example, in GenBank Accession No. NM_005195, and the nucleotide sequence encoding the mouse Cebpd protein is registered, for example, in GenBank Accession No. NM_007679.
Hnf6はHepatocyte nuclear factor 6と呼ばれ、肝細胞分化や肝細胞における糖・脂質代謝を調節することが知られている。ヒトHNF6タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_004489(配列番号9)に登録されており、マウスHnf6タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_032288(配列番号10)に登録されている。ただし、これら特定のアミノ酸配列を有するタンパク質には限定されず、転写因子としての機能を有する限り、これらの配列と90%以上、好ましくは95%以上、より好ましくは98%以上の同一性を有するアミノ酸配列を有するタンパク質でもよい。ヒトHNF6タンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_004498に登録されており、マウスHnf6タンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_008262に登録されている。 Hnf6 is called Hepatocyte nuclear factor 6 and is known to regulate hepatocyte differentiation and glucose/lipid metabolism in hepatocytes. The amino acid sequence of human HNF6 protein is registered, for example, in GenBank Accession No. NP_004489 (SEQ ID NO: 9), and the amino acid sequence of mouse Hnf6 protein is registered, for example, in GenBank Accession No. NP_032288 (SEQ ID NO: 10). However, it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence. The nucleotide sequence encoding the human HNF6 protein is registered in, for example, GenBank Accession No. NM_004498, and the nucleotide sequence encoding the mouse Hnf6 protein is registered in, for example, GenBank Accession No. NM_008262.
Onecut2はOnecut domain family member 2と呼ばれ、Foxa2遺伝子の発現を誘導するなど、肝細胞の分化や代謝に関与することが知られている。ヒトONECUT2タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_004843(配列番号11)に登録されており、マウスOnecut2タンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_919244(配列番号12)に登録されている。ただし、これら特定のアミノ酸配列を有するタンパク質には限定されず、転写因子としての機能を有する限り、これらの配列と90%以上、好ましくは95%以上、より好ましくは98%以上の同一性を有するアミノ酸配列を有するタンパク質でもよい。ヒトONECUT2タンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_004852に登録されており、マウスOnecut2タンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_194268に登録されている。 Onecut2 is called Onecut domain family member2 and is known to be involved in hepatocyte differentiation and metabolism such as inducing the expression of Foxa2 gene. The amino acid sequence of human ONECUT2 protein is registered, for example, in GenBank Accession No. NP_004843 (SEQ ID NO: 11), and the amino acid sequence of mouse Onecut2 protein is registered, for example, in GenBank Accession No. NP_919244 (SEQ ID NO: 12). However, it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence. The nucleotide sequence encoding the human ONECUT2 protein is registered, for example, in GenBank Accession No. NM_004852, and the nucleotide sequence encoding the mouse Onecut2 protein is registered, for example, in GenBank Accession No. NM_194268.
さらに、Mycが導入されることが好ましい。ヒトMYCタンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_002458(配列番号13)に登録されており、マウスMycタンパク質のアミノ酸配列は例えば、GenBank Accession No. NP_034979(配列番号14)に登録されている。ただし、これら特定のアミノ酸配列を有するタンパク質には限定されず、転写因子としての機能を有する限り、これらの配列と90%以上、好ましくは95%以上、より好ましくは98%以上の同一性を有するアミノ酸配列を有するタンパク質でもよい。ヒトMYCタンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_002467に登録されており、マウスMycタンパク質をコードする塩基配列は例えば、GenBank Accession No. NM_010849に登録されている。 Furthermore, it is preferable that Myc is introduced. The amino acid sequence of human MYC protein is registered in, for example, GenBank Accession No. NP_002458 (SEQ ID NO: 13), and the amino acid sequence of mouse Myc protein is registered in, for example, GenBank Accession No. NP_034979 (SEQ ID NO: 14). However, it is not limited to proteins having these specific amino acid sequences, and as long as it has a function as a transcription factor, it has 90% or more, preferably 95% or more, and more preferably 98% or more identity with these sequences. It may be a protein having an amino acid sequence. The nucleotide sequence encoding the human MYC protein is registered in, for example, GenBank Accession No. NM_002467, and the nucleotide sequence encoding the mouse Myc protein is registered in, for example, GenBank Accession No. NM_010849.
上記6因子およびMycは同時に導入されてもよいし、個別因子または複数因子が順次導入されてもよい。 The above 6 factors and Myc may be introduced simultaneously, or individual factors or multiple factors may be introduced sequentially.
これらの転写因子の導入により、これらの転写因子の発現量(遺伝子レベルまたはタンパク質レベル)は、導入前の細胞における発現量の5倍以上に維持されることが好ましく、10倍以上に維持されることがより好ましい。前記発現量が前記レベルに維持される期間としては、例えば、少なくとも7日間、好ましくは10日間、さらに好ましくは14日間、特に好ましくは肝細胞への分化が終了するまでの期間である。これにより、成熟肝細胞への高効率での分化誘導が可能になる。肝細胞への分化が終了して実験に供されるまで持続することがさらに好ましい。これにより、成熟肝細胞への高効率な分化誘導と、得られた成熟肝細胞機能の機能が良好に維持される場合がある。 By the introduction of these transcription factors, the expression levels (gene level or protein level) of these transcription factors are preferably maintained at 5 times or more, preferably 10 times or more the expression levels in the cells before the introduction. Is more preferable. The period in which the expression level is maintained at the level is, for example, at least 7 days, preferably 10 days, more preferably 14 days, and particularly preferably the period until the differentiation into hepatocytes is completed. This enables highly efficient differentiation induction into mature hepatocytes. It is further preferable that the differentiation into hepatocytes is completed and the differentiation is continued until it is subjected to the experiment. As a result, highly efficient differentiation induction into mature hepatocytes and the function of the obtained mature hepatocyte function may be favorably maintained.
本発明において、上記の転写因子は遺伝子レベルで体細胞に導入されてもよいし、タンパク質レベルで体細胞に導入されてもよい。なお、内因性で発現している転写因子であれば内因性転写因子の発現を誘導してもよい。すなわち、転写因子の導入は内因性転写因子の誘導を含む。 In the present invention, the above transcription factor may be introduced into somatic cells at the gene level or may be introduced into somatic cells at the protein level. In addition, if the transcription factor is expressed endogenously, the expression of the endogenous transcription factor may be induced. That is, the introduction of a transcription factor involves the induction of an endogenous transcription factor.
<導入方法>
遺伝子(DNA)の形態で導入する場合、例えば、ウイルス、プラスミド、人工染色体などのベクターをリポフェクション、リポソーム、マイクロインジェクションなどの手法によって体細胞内に導入することができる。導入は一過的導入でもよいし、安定的導入でもよい。
ウイルスベクターとしては、レトロウイルスベクター、レンチウイルスベクター、アデノウイルスベクター、アデノ随伴ウイルスベクター、センダイウイルスベクターなどが例示される。また、人工染色体ベクターとしては、例えばヒト人工染色体(HAC)、酵母人工染色体(YAC)、細菌人工染色体(BAC、PAC)などが含まれる。プラスミドとしては、哺乳動物細胞用プラスミドを使用しうる。ベクターには、導入遺伝子が発現可能なように、プロモーター、エンハンサー、リボゾーム結合配列、ターミネーター、ポリアデニル化サイトなどの制御配列を含むことができるし、さらに、必要に応じて、薬剤耐性遺伝子(例えばカナマイシン耐性遺伝子、アンピシリン耐性遺伝子、ピューロマイシン耐性遺伝子など)、チミジンキナーゼ遺伝子、ジフテリアトキシン遺伝子などの選択マーカー配列、蛍光タンパク質、βグルクロニダーゼ(GUS)、FLAGなどのレポーター遺伝子配列などを含むことができる。プロモーターとして、SV40プロモーター、 LTRプロモーター、CMV (cytomegalovirus)プロモーター、RSV (Rous sarcoma virus)プロモーター、MoMuLV (Moloney mouse leukemia virus) LTR、HSV-TK (herpes simplex virus thymidine kinase)プロモーター、EF-αプロモーター、CAGプロモーターおよびTREプロモーター(tetO 配列が7回連続したTet応答配列をもつCMV 最小プロモーター)が例示される。
<Introduction method>
When introduced in the form of a gene (DNA), for example, a vector such as virus, plasmid or artificial chromosome can be introduced into somatic cells by a technique such as lipofection, liposome, microinjection and the like. The introduction may be a transient introduction or a stable introduction.
Examples of viral vectors include retrovirus vectors, lentivirus vectors, adenovirus vectors, adeno-associated virus vectors, Sendai virus vectors and the like. Examples of artificial chromosome vectors include human artificial chromosomes (HAC), yeast artificial chromosomes (YAC), bacterial artificial chromosomes (BAC, PAC), and the like. As the plasmid, a plasmid for mammalian cells can be used. The vector can contain regulatory sequences such as a promoter, an enhancer, a ribosome binding sequence, a terminator, and a polyadenylation site so that the transgene can be expressed, and further, if necessary, a drug resistance gene (for example, kanamycin). Resistance gene, ampicillin resistance gene, puromycin resistance gene, etc.), thymidine kinase gene, diphtheria toxin gene and other selectable marker sequences, fluorescent protein, β-glucuronidase (GUS), FLAG, and other reporter gene sequences. As a promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, RSV (Rous sarcoma virus) promoter, MoMuLV (Moloney mouse leukemia virus) LTR, HSV-TK (herpes simplex virus thymidine kinase) promoter, EF-α promoter, CAG Examples are a promoter and a TRE promoter (CMV minimal promoter having a Tet response element in which the tetO sequence is continuous 7 times).
RNAの形態で導入する場合、例えばエレクトロポレーション、リポフェクション、マイクロインジェクションなどの手法によって多能性幹細胞内に導入してもよい。 When introduced in the form of RNA, it may be introduced into pluripotent stem cells by a technique such as electroporation, lipofection, or microinjection.
タンパク質の形態で導入する場合、例えばリポフェクション、細胞膜透過性ペプチド(例えば、HIV由来のTATおよびポリアルギニン)との融合、マイクロインジェクションなどの手法によって多能性幹細胞内に導入してもよい。
6因子の発現は、少なくとも7日間、好ましくは10日間、さらに好ましくは14日間、特に好ましくは肝細胞への分化が終了するまでの期間、持続することが望ましい。これにより、成熟肝細胞への高効率での分化誘導が可能になる。さらに、分化が終了して実験に供されるまで持続してもよい。これにより、得られた成熟肝細胞の機能が良好に維持される場合がある。
When introduced in the form of a protein, it may be introduced into pluripotent stem cells by a technique such as lipofection, fusion with a cell membrane-penetrating peptide (eg, TAT derived from HIV and polyarginine), and microinjection.
It is desirable that the expression of factor 6 be sustained for at least 7 days, preferably 10 days, more preferably 14 days, and particularly preferably a period until the differentiation into hepatocytes is completed. This enables highly efficient differentiation induction into mature hepatocytes. Furthermore, it may be maintained until the differentiation is completed and the experiment is performed. Thereby, the function of the obtained mature hepatocytes may be favorably maintained.
<培養工程>
転写因子が導入された細胞を培養する工程に使用する培養液は、特に限定されないが、動物細胞の培養に用いられる培地を基礎培地とし、適切な成分を添加して調製することができる。基礎培地には、例えばIscove's Modified Dulbecco's Medium(IMDM)培地、Medium 199培地、Eagle's Minimum Essential Medium (EMEM)培地、αMEM培地、Dulbecco's modified Eagle's Medium (DMEM)培地、Ham's F12培地、RPMI 1640培地、Fischer's培地、Neurobasal Medium(ライフテクノロジーズ)およびこれらの混合培地などが包含される。HCM BulletKit Medium、William’s E Medium (Thermo Fisher Scientific), Leibovitz’s L-15 Medium (Thermo Fisher Scientific)などの肝細胞用培地も好ましく用いることができる。培地には、血清が含有されていてもよいし、あるいは無血清を使用してもよい。必要に応じて、基礎培地は、例えば、アルブミン、インスリン、トランスフェリン、セレン、脂肪酸、微量元素、2-メルカプトエタノール、チオールグリセロール、脂質、アミノ酸、L-グルタミン、非必須アミノ酸、ビタミン、増殖因子、低分子化合物、抗生物質、抗酸化剤、ピルビン酸、緩衝剤、無機塩類、副腎皮質ホルモン(デキサメタゾンなどのホルモン、サイトカインなどの物質も含有し得る。
サイトカインとしては、肝細胞増殖因子(HGF)やオンコスタチンM(OSM)などが挙げられ、これらは、例えば、1~100ng/mLの濃度で添加される。
<Culture process>
The culture medium used in the step of culturing the cells into which the transcription factor has been introduced is not particularly limited, but can be prepared by using a medium used for culturing animal cells as a basal medium and adding appropriate components. The basal medium includes, for example, Iscove's Modified Dulbecco's Medium (IMDM) medium, Medium 199 medium, Eagle's Minimum Essential Medium (EMEM) medium, αMEM medium, Dulbecco's modified Eagle's Medium (DMEM) medium, Ham's F12 medium, RPMI 1640 medium, Fischer's medium. , Neurobasal Medium (Life Technologies) and mixed media thereof. A medium for hepatocytes such as HCM BulletKit Medium, William's E Medium (Thermo Fisher Scientific), Leibovitz's L-15 Medium (Thermo Fisher Scientific) can also be preferably used. The medium may contain serum or may be serum-free. If necessary, the basal medium may include, for example, albumin, insulin, transferrin, selenium, fatty acids, trace elements, 2-mercaptoethanol, thiolglycerol, lipids, amino acids, L-glutamine, non-essential amino acids, vitamins, growth factors, low levels. It may also contain substances such as molecular compounds, antibiotics, antioxidants, pyruvic acid, buffers, inorganic salts, corticosteroids (hormones such as dexamethasone, cytokines).
Examples of cytokines include hepatocyte growth factor (HGF) and oncostatin M (OSM), which are added at a concentration of 1 to 100 ng/mL, for example.
<TGFβ受容体阻害剤>
培地にはTGFβ受容体阻害剤を添加することが好ましい。TGFβ受容体阻害剤とは、TGFβの受容体への結合からSMADへと続くシグナル伝達を阻害する物質であり、受容体であるALKファミリーへの結合を阻害する物質、またはALKファミリーによるSMADのリン酸化を阻害する物質が挙げられ、例えば、Lefty-1(NCBI Accession No.として、マウス:NM_010094、ヒト:NM_020997が例示される)、SB431542、SB202190(以上、R.K.Lindemann et al., Mol. Cancer, 2003, 2:20)、SB505124 (GlaxoSmithKline)、 NPC30345、SD093、SD908、SD208 (Scios)、LY2109761、LY364947、 LY580276 (Lilly Research Laboratories)、A83-01(WO 2009146408) およびこれらの誘導体などが例示される。本発明で使用されるTGFβ受容体阻害剤は、好ましくは、A83-01であり得る。
<TGFβ receptor inhibitor>
It is preferable to add a TGFβ receptor inhibitor to the medium. A TGFβ receptor inhibitor is a substance that inhibits the signal transduction from the binding of TGFβ to the receptor and subsequent to SMAD, a substance that inhibits the binding to the ALK family of receptors, or a phosphorylation of SMAD by the ALK family. Examples of substances that inhibit oxidation include Lefty-1 (NCBI Accession No. is exemplified by mouse: NM_010094 and human: NM_020997), SB431542, SB202190 (above, RKLindemann et al., Mol. Cancer, 2003). , 2:20), SB505124 (GlaxoSmithKline), NPC30345, SD093, SD908, SD208 (Scios), LY2109761, LY364947, LY580276 (Lilly Research Laboratories), A83-01 (WO 2009146408) and derivatives thereof. The TGFβ receptor inhibitor used in the present invention may preferably be A83-01.
培養液中におけるA83-01の濃度は、ALK5を阻害する濃度であれば特に限定されないが、例えば、500nMから5μMである。 The concentration of A83-01 in the culture medium is not particularly limited as long as it is a concentration that inhibits ALK5, and is, for example, 500 nM to 5 μM.
培養は接着培養でも浮遊培養でもよく、用いる体細胞の種類によって適宜選択できる。浮遊培養によって行われる場合、細胞を培養容器へ非接着の状態で凝集体(スフェアとも言う)を形成させて培養することが望ましく、このような培養は、特に限定はされないが、細胞との接着性を向上させる目的で人工的に処理(例えば、細胞外マトリックス等によるコーティング処理)されていない培養容器、若しくは、人工的に接着を抑制する処理(例えば、ポリヒドロキシエチルメタクリル酸(poly-HEMA)、非イオン性の界面活性ポリオール(Pluronic F-127等)またはリン脂質類似構造物(例えば、2-メタクリロイルオキシエチルホスホリルコリンを構成単位とする水溶性ポリマー(Lipidure))によるコーティング処理した培養容器を使用することによって行うことができる。 The culture may be adherent culture or suspension culture, and can be appropriately selected depending on the type of somatic cells used. When it is carried out by suspension culture, it is desirable to culture cells by forming aggregates (also referred to as spheres) in a culture container in a non-adhesive state. Such culture is not particularly limited, but it is possible to adhere to cells. Culture vessel that is not artificially treated (for example, coating treatment with extracellular matrix etc.) for the purpose of improving the property, or treatment that artificially suppresses adhesion (for example, polyhydroxyethyl methacrylic acid (poly-HEMA)) Use a culture vessel coated with a nonionic surface-active polyol (Pluronic F-127 etc.) or a phospholipid-like structure (for example, a water-soluble polymer (Lipidure) containing 2-methacryloyloxyethylphosphorylcholine as a constituent unit) Can be done by doing.
接着培養によって培養が行われる場合、細胞外基質をコーティング処理された培養容器を用いて培養することによって行うことができる。コーティング処理は、細胞外基質を含有する溶液を培養容器に入れた後、当該溶液を適宜除くことによって行い得る。ここで、細胞外基質とは、細胞の外に存在する超分子構造体であり、天然由来であっても、人工物(組換え体)であってもよい。例えば、ポリリジン、ポリオルニチン、コラーゲン、プロテオグリカン、フィブロネクチン、ヒアルロン酸、テネイシン、エンタクチン、エラスチン、フィブリリン、ラミニンといった物質およびこれらの断片が挙げられる。これらの細胞外基質は、組み合わせて用いられてもよく、例えば、BD Matrigel(商標)などの細胞からの調製物であってもよい。  When culturing is carried out by adhesion culturing, it can be carried out by culturing in a culture vessel coated with an extracellular matrix. The coating treatment can be performed by placing a solution containing an extracellular matrix in a culture vessel and then appropriately removing the solution. Here, the extracellular matrix is a supramolecular structure existing outside the cell, and may be of natural origin or artificial (recombinant). Examples thereof include substances such as polylysine, polyornithine, collagen, proteoglycan, fibronectin, hyaluronic acid, tenascin, entactin, elastin, fibrillin and laminin, and fragments thereof. These extracellular matrices may be used in combination, and may be, for example, preparations from cells such as BD Matrigel™. 
培養条件は、特に限定されないが、例えば、約37℃~約42℃程度、約37~約39℃程度が好ましい。CO2含有空気の雰囲気下で培養が行われ、CO2濃度は、好ましくは約2~5%である。また、培養期間については、当業者であれば細胞数などをモニターしながら、適宜決定することが可能である。日数は特に限定されないが、例えば、10日間以上、12日以上、14日以上、16日以上、18日以上、20日以上、25日以上である。 Culture conditions are not particularly limited, but for example, about 37°C to about 42°C, preferably about 37 to about 39°C. Culturing is carried out in an atmosphere of CO 2 -containing air, and the CO 2 concentration is preferably about 2-5%. In addition, those skilled in the art can appropriately determine the culture period while monitoring the cell number and the like. The number of days is not particularly limited, but is, for example, 10 days or more, 12 days or more, 14 days or more, 16 days or more, 18 days or more, 20 days or more, 25 days or more.
<肝細胞>
肝細胞が得られたことは、アルブミンなどの発現を測定することにより評価できる。
肝細胞は、他の細胞種が含まれる細胞集団として得られてもよく、純化されてもよい。本発明の方法で得られる細胞集団は、例えば、3%以上、5%以上、10%以上もしくは20%以上の肝細胞を含むが、好ましくは、50%以上、60%以上、70%以上または80%以上の肝細胞を含む。なお、肝細胞を純化する方法する方法として、アシアログライコプロテイン(ASGP)受容体に対する抗体などを用いて染色し、染色された細胞をフローサイトメーター(FACS)や磁気細胞分離装置(MACS)を用いて濃縮する方法が例示される。
<Hepatocytes>
Obtainment of hepatocytes can be evaluated by measuring the expression of albumin and the like.
Hepatocytes may be obtained as a cell population containing other cell types or may be purified. The cell population obtained by the method of the present invention contains, for example, 3% or more, 5% or more, 10% or more or 20% or more hepatocytes, but preferably 50% or more, 60% or more, 70% or more, or Contains 80% or more hepatocytes. As a method for purifying hepatocytes, staining with an antibody against asialoglycoprotein (ASGP) receptor or the like and staining the stained cells with a flow cytometer (FACS) or magnetic cell sorter (MACS) A method of concentrating by using the method is exemplified.
本発明で得られた肝細胞は上記6因子、好ましくはさらにMycが導入されたことにより、成体の肝細胞としての機能を有する。例えば、肝細胞はアルブミンの分泌に加え、シトクロムP450活性、薬物代謝、グリコーゲンの蓄積、低比重リポタンパク質(LDL)の取り込み、アンモニア代謝と尿素合成などの肝細胞の機能の1つ以上を有する。 The hepatocytes obtained in the present invention have the function as adult hepatocytes by introducing the above 6 factors, preferably Myc. For example, hepatocytes have, in addition to albumin secretion, one or more of hepatocyte functions such as cytochrome P450 activity, drug metabolism, glycogen accumulation, low-density lipoprotein (LDL) uptake, ammonia metabolism and urea synthesis.
<細胞製剤> 
本発明では、上述した方法により得られた肝細胞を含む、肝疾患治療等に使用される再生医療用細胞製剤を提供する。
患者への細胞製剤の投与方法としては、例えば、得られた肝細胞をシート化して、患者の肝臓に貼付する方法、得られた肝細胞を生理食塩水等に懸濁させ、患者の肝臓に直接移植する方法、マトリゲル等から構成されたスキャフォールド上で三次元培養し、得られた肝細胞塊を移植する方法などが挙げられる。 
<Cell preparation>
The present invention provides a cell preparation for regenerative medicine, which comprises the hepatocytes obtained by the above-mentioned method and is used for treatment of liver diseases and the like.
As a method of administering a cell preparation to a patient, for example, a method of forming the obtained hepatocytes into a sheet and affixing it to the patient's liver, suspending the obtained hepatocytes in physiological saline, etc. Examples thereof include a method of direct transplantation, a method of three-dimensionally culturing on a scaffold composed of Matrigel and the like, and a method of transplanting the obtained hepatocyte mass.
本発明において、細胞製剤に含まれる肝細胞の細胞数は、移植片が投与後に生着できれば特に限定されなく、患部の大きさや体躯の大きさに合わせて適宜増減して調製すればよい。  In the present invention, the number of hepatocytes contained in the cell preparation is not particularly limited as long as the graft can be engrafted after administration, and may be adjusted appropriately according to the size of the affected area or the size of the body. 
<肝細胞を用いた薬物評価>
本発明の方法で調製した肝細胞を用いて被検物質の代謝についてin vitroで試験することができる。
当該被検物質の代謝試験は、例えば、(i)本発明の方法で得られた肝細胞に被検物質を接触させる工程、および(ii)被検物質の代謝産物を検出する工程を含むことができる。
工程(i)での「接触」とは、本発明によって製造された肝細胞を培養する培地に被検物質を添加することによって行われ得る。被検化合物の添加のタイミングは特に限定されない。従って、被検物質を含まない培地で培養を開始した後、ある時点で被検物質を添加することにしても、予め被検物質を含む培地で培養を開始することにしてもよい。 
<Drug evaluation using hepatocytes>
The liver cells prepared by the method of the present invention can be used to test the metabolism of a test substance in vitro.
The metabolism test of the test substance includes, for example, the step of (i) contacting the test substance with the hepatocytes obtained by the method of the present invention, and (ii) detecting a metabolite of the test substance. You can
The "contact" in step (i) can be performed by adding a test substance to the medium for culturing hepatocytes produced by the present invention. The timing of addition of the test compound is not particularly limited. Therefore, after starting the culture in the medium not containing the test substance, the test substance may be added at a certain point in time, or the culture may be started in advance in the medium containing the test substance.
被検物質には、例えば、医薬品や栄養食品等の既存成分或いは候補成分などが包含される。被検物質は天然物由来であっても、或いは合成によるものであってもよい。後者の場合には例えばコンビナトリアル合成の手法を利用して効率的なアッセイ系を構築することができる。  The test substance includes, for example, existing components such as pharmaceuticals and nutritional foods or candidate components. The test substance may be derived from a natural product or may be synthetic. In the latter case, an efficient assay system can be constructed by using, for example, a combinatorial synthesis method. 
被検物質を接触させる期間は任意に設定可能である。接触期間は例えば10分間~3日間、好ましくは1時間~1日間である。接触を複数回に分けて行うことにしてもよい。  The period of contact with the test substance can be set arbitrarily. The contact period is, for example, 10 minutes to 3 days, preferably 1 hour to 1 day. The contact may be divided into a plurality of times. 
被検物質の代謝とは、エステルなどの加水分解、酸化反応(特にシトクロムP450による酸化)、還元反応などによる被検物質の化学修飾が例示され、このような代謝の有無は、例えば、代謝産物の検出によって行うことができる。好ましくは、工程(i)の後、得られた培養液をサンプルとして、予想される代謝産物を定性的又は定量的に測定する。測定方法は被検物質並びにその予測される代謝産物に応じて適切なものを選択すればよいが、例えば、質量分析、液体クロマトグラフィー、免疫学的手法(例えば蛍光免疫測定法(FIA法)、酵素免疫測定法(EIA法))等を採用可能である。  Examples of the metabolism of the test substance include chemical modification of the test substance by hydrolysis of ester and the like, oxidation reaction (particularly oxidation by cytochrome P450), reduction reaction, and the like. Can be detected. Preferably, after the step (i), the expected metabolite is qualitatively or quantitatively measured using the obtained culture solution as a sample. The measurement method may be selected appropriately depending on the test substance and its predicted metabolite, and examples thereof include mass spectrometry, liquid chromatography, immunological methods (eg, fluorescence immunoassay (FIA method), Enzyme-linked immunosorbent assay (EIA method) etc. can be adopted. 
被検物質の代謝産物が検出されたとき、「被検物質が代謝された」と判定する。また、代謝産物の量に応じて被検物質の代謝量を評価することができる。  When the metabolite of the test substance is detected, it is determined that "the test substance has been metabolized." In addition, the amount of metabolism of the test substance can be evaluated according to the amount of metabolites. 
また、本発明で得られた肝細胞を用いて薬物代謝酵素(例えば、シトクロム、UGT)の発現誘導を確認することができる。薬物代謝酵素の発現はmRNAレベル又はタンパク質レベルで評価することができる。例えば、薬物代謝酵素のmRNAレベルに上昇を認めたとき、「被検物質は薬物相互作用を起こす疑いがある」と判定することができる。  In addition, the hepatocytes obtained in the present invention can be used to confirm the induction of expression of drug-metabolizing enzymes (eg, cytochrome, UGT). Expression of drug-metabolizing enzymes can be assessed at the mRNA or protein level. For example, when an increase in the mRNA level of a drug-metabolizing enzyme is observed, it can be determined that “the test substance is suspected of causing a drug interaction”. 
一方、本発明の方法で調製した肝細胞を用いて被検物質の毒性を試験することもできる。当該方法は、(i)本発明の方法で得られた肝細胞に被検物質を接触させる工程と、(ii)工程(i)後の肝細胞の状態を調べる工程と、(iii)(ii)の結果に基づいて被検物質の毒性を評価する工程、とを含むことができる。工程(i)は、前記被検物質の代謝と同様である。 On the other hand, the toxicity of the test substance can also be tested using the hepatocytes prepared by the method of the present invention. The method comprises (i) a step of contacting a test substance with the hepatocytes obtained by the method of the present invention, (ii) a step of examining the state of the hepatocytes after the step (i), and (iii) (ii) ), the step of evaluating the toxicity of the test substance based on the result of 4). Step (i) is the same as the metabolism of the test substance.
工程(ii)では、被検物質を接触させた後の肝細胞の状態を調べ、工程(iii)において被検物質の毒性を評価する。肝細胞の状態は、生存率の測定、細胞形態の観察、培養液(培養上清)中の肝障害マーカー(GOT、GPT等)の測定などによって把握することができる。例えば、被検物質の接触によって生存率の低下を認めたとき、「被検物質は肝毒性を有する」と判定することができる。また、被検物質の接触によって細胞形態の異常を認めたときや、培養液中の肝障害マーカーの量が上昇したときも同様に、「被検物質は肝毒性を有する」と判定することができる。生存率の低下の程度や肝障害マーカーの量に応じて定量的な判定を行ってもよい。なお、前記「生存率の低下」は、目安として、被検物質と接触させなかった肝細胞(対照細胞)の生存率に対し、90%以下、80%以下、70%以下、60%以下、50%以下の生存率であってもよい。また、前記「肝障害マーカーの量が上昇」は、目安として、対照細胞の生存率に対し、110%以上、120%以上、130%以下、140%以上、または150%以上の前記マーカー量であってもよい。
 また、工程(ii)において被検物質のTC50を解析し、工程(iii)において、当該TC50値が特定の値以下の被検物質に対して「肝毒性を有する」と判定してもよい。被検物質の肝細胞に対するTC50を調べることで、人体への被検物質の適正投与量の予測に貢献することができる。
In step (ii), the state of hepatocytes after contact with the test substance is examined, and the toxicity of the test substance is evaluated in step (iii). The state of hepatocytes can be ascertained by measuring viability, observing cell morphology, measuring liver damage markers (GOT, GPT, etc.) in the culture solution (culture supernatant), and the like. For example, when a decrease in survival rate is observed due to contact with a test substance, it can be determined that "the test substance has liver toxicity". Similarly, when an abnormal cell morphology is observed due to contact with a test substance, or when the amount of a liver damage marker in the culture solution is increased, it can be similarly determined that "the test substance has hepatotoxicity". it can. Quantitative determination may be performed depending on the degree of decrease in survival rate and the amount of liver damage marker. The "reduction in survival rate" is, as a guide, 90% or less, 80% or less, 70% or less, 60% or less with respect to the survival rate of hepatocytes (control cells) not contacted with the test substance. The survival rate may be 50% or less. In addition, the "increased amount of liver damage marker" is, as a guide, a marker amount of 110% or more, 120% or more, 130% or less, 140% or more, or 150% or more with respect to the survival rate of control cells. It may be.
Further, the TC 50 of the test substance may be analyzed in the step (ii), and in the step (iii), it may be determined that the test substance has a hepatotoxicity with respect to the test substance having a TC50 value of a specific value or less. .. By examining the TC 50 of the test substance for hepatocytes, it is possible to contribute to the prediction of the proper dose of the test substance to the human body.
以下、実施例を挙げて本発明を具体的に説明するが、本発明の態様は以下には限定されない。 Hereinafter, the present invention will be specifically described with reference to examples, but the embodiments of the present invention are not limited to the following.
[実施例1]
以下の手順でレトロウイルスベクターを使用して線維芽細胞に遺伝子導入を行うことにより誘導肝細胞(iHeps)を作製し、得られたiHepsを用いて肝細胞マーカーの発現などを解析した。
[Example 1]
Induced hepatocytes (iHeps) were prepared by introducing genes into fibroblasts using a retrovirus vector according to the following procedure, and the obtained iHeps was used to analyze the expression of hepatocyte markers.
ウイルスベクター作製
マウスのHnf4a遺伝子、Foxa3遺伝子、Cebpa遺伝子、Cebpd遺伝子、Hnf6遺伝子およびOnecut2遺伝子をそれぞれpMXsプラスミドに組み込み、各遺伝子導入用のレトロウイルスベクターをそれぞれ作製した。また、マウスのMyc遺伝子導入用のレトロウイルスベクターも同様に作製した。なお、pMXsは東京大学の北村俊雄教授より譲り受けた(Exp Hematol. 2003 Nov;31(11):1007-14.)。
Preparation of viral vector The mouse Hnf4a gene, Foxa3 gene, Cebpa gene, Cebpd gene, Hnf6 gene and Onecut2 gene were each incorporated into pMXs plasmid to prepare a retroviral vector for introducing each gene. In addition, a retrovirus vector for introducing the Myc gene of mouse was also prepared in the same manner. In addition, pMXs was transferred from Professor Toshio Kitamura of the University of Tokyo (Exp Hematol. 2003 Nov;31(11):1007-14.).
ウイルス上清準備
1. Plat-E (Cell Biolabs, Inc., Cat# RV-101) を、抗生物質を含まない10% FBS/DMEM (FBS (Thermo Fisher Scientific), DMEM (Nacalai tesque, Cat# 08459-64)) に懸濁し、ゼラチンコートした6-well plate (Greiner) に播種 (5.0 × 105 cells/1.5 ml/well)し、37℃ 5% CO2 で一晩インキュベートした。
Virus supernatant preparation
1.Plat-E (Cell Biolabs, Inc., Cat# RV-101) with 10% FBS/DMEM (FBS (Thermo Fisher Scientific), DMEM (Nacalai tesque, Cat# 08459-64)) containing no antibiotics. The cells were suspended in a sol, and seeded (5.0 × 10 5 cells/1.5 ml/well) on a gelatin-coated 6-well plate (Greiner), and incubated overnight at 37°C, 5% CO 2 .
2. Plat-Eに対し、各転写因子のplasmidのlipofectionを行った。
37℃のOpti-MEM (Thermo Fisher Scientific, Cat# 31985062) 50 μlにFuGENE 6 (Promega, Cat# E2692) 4.5 μlを加え、vortex後に5分間室温静置した。そこに各転写因子のレトロウイルスベクター (pMXs) 500 ng/μlを加え、20分間室温静置し、その後、1で用意したPlat-Eの各wellに加え、37℃ 5% CO2 で一晩インキュベートした。
2. Plat-E was subjected to lipofection of each transcription factor plasmid.
4.5 μl of FuGENE 6 (Promega, Cat# E2692) was added to 50 μl of Opti-MEM (Thermo Fisher Scientific, Cat# 31985062) at 37° C., and the mixture was allowed to stand at room temperature for 5 minutes after vortexing. Add 500 ng/μl of retroviral vector (pMXs) of each transcription factor to it, leave it at room temperature for 20 minutes, then add to each well of Plat-E prepared in 1 above, and then overnight at 37°C 5% CO 2 Incubated.
3. 抗生物質を含まない10%FBS/DMEM 1.6 ml/wellで培地交換し、37℃ 5% CO2で一晩インキュベートし、それをウイルス上清として用いた。 3. The medium was exchanged with 1.6 ml/well of 10% FBS/DMEM containing no antibiotic, and the mixture was incubated overnight at 37° C. and 5% CO 2 and used as a virus supernatant.
MEFs (mouse embryonic fibroblasts)準備
E13.5 妊娠マウス (C57BL/6J) の胎仔より得た。継代は行わず、Cell Reservoir One (Nacalai tesque, Cat# 07485-44)を細胞保存液として用いて、-80℃凍結保存した。
Preparation of MEFs (mouse embryonic fibroblasts)
E13.5 Obtained from a fetus of a pregnant mouse (C57BL/6J). Without passage, Cell Reservoir One (Nacalai tesque, Cat# 07485-44) was used as a cell preservation solution and frozen and preserved at -80°C.
iHep induction
1. Viral transduction前日 (day-1)、凍結保存していたMEFsを起眠し、Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat# 354230) でコーティングした12-well plate (Greiner) 上に播種 (5.0 × 10cells/well)した。
iHep induction
1. Viral transduction The day before (day-1), the MEFs that had been cryopreserved were put to sleep and put on a 12-well plate (Greiner) coated with Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat# 354230). Seeding (5.0 × 10 4 cells/well) was performed.
2. Viral transduction当日(day0)、Plat-Eの培養上清をウイルス上清として回収し、filtering (Millex-HP Syringe Filter Unit 0.45 μm, Merck Millipore, Cat# SLHP033RS)した。 2. On the day of Viral transduction (day 0), the culture supernatant of Plat-E was collected as a virus supernatant and subjected to filtering (Millex-HPSyringe Filter Unit 0.45 μm, Merck Millipore, Cat# SLHP033RS).
3. 各転写因子のウイルス上清を各100 μl(c-Mycは12.5 μl), 10% FBS/DMEM 87.5 μl, Polybrene (nacalai tesque, Cat# 12996-81) 0.7 μlを加え、そのうち500 μlを、前日MEFsを播種した12-well plateの1wellあたりに加えた。 3.100 μl of virus supernatant for each transcription factor (12.5 μl for c-Myc), 10% FBS/DMEM 87.5 μl, Polybrene (nacalai tesque, Cat# 12996-81) 0.7 μl, of which 500 μl , MEFs were seeded the day before, per well of a 12-well plate.
4. 37℃, 5% CO2 で一晩インキュベートし、ウイルス感染を行った。 4. Incubated overnight at 37°C, 5% CO 2 to infect the virus.
5. 翌日 (day1) 培地交換 (10% FBS/DMEM)した。 5. The next day (day 1), medium replacement (10% FBS/DMEM) was performed.
6. Day3にも培地交換 (10% FBS/DMEM)した。 6. The medium was changed (10% FBS/DMEM) on Day 3.
7. Day4でMatrigelでコートされた12-well plateに1:10の割合でreplateした。培地は10% FBS/DMEM/F12, GlutaMAXをベースにしたHepatic specification培地 (DMEM/F12, GlutaMAX (Thermo Fisher Scientific, Cat# 10565042) 360 ml, FBS 40 ml, Nicotinamide (STEMCELL Technologies, Cat# 07154) 10mM, ITS-X (Thermo Fisher Scientific, Cat# 51500056) 400μl, Dexamethasone (Merck, Cat# D4902-25MG) 0.1μM, 2-Mercaptoethanol (Thermo Fisher Scientific, Cat# 21985023) 50 μM, Penicillin-Streptomycin (Thermo Fisher Scientific, Cat# 15140122) 2 ml)とし、Day6に培地交換を行った。 7. On Day 4, replate 1:10 on 12-well plate coated with Matrigel. Hepatic specification medium based on 10% FBS/DMEM/F12, GlutaMAX , ITS-X (ThermoFisher Scientific, Cat# 51500056) 400μl, Dexamethasone (Merck, Cat# D4902-25MG) 0.1μM, 2-Mercaptoethanol (Thermo Fisher Fercier , Cat#15140122)2) ml), and changed the medium on Day 6.
8. Day7から、hepatic maturation培地(HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ml, A83-01 (Merck, Cat# 616454-2MGCN) 1 μM)に変更して、2日に一度培地交換した。
Day25前後で遺伝子発現解析等の実験に供した。
8. From Day 7, hepatic maturation medium (HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ ml, A83-01 (Merck, Cat# 616454-2MGCN) 1 μM), and the medium was exchanged once every two days.
It was subjected to experiments such as gene expression analysis around Day 25.
遺伝子発現はRT-qPCRで解析した。
Albuminの分泌量の定量はELISAキット(Mouse Albumin ELISA Kit (BETHYL Laboratories)を使用した。
Cyp3a11の活性測定においては、基質となるMidazolamを培地に加え、Cyp3a11による水酸化により産生されたHydroxy midazolamの24時間あたりの培地中への分泌量を、Mass spectrometryで測定した。
Gene expression was analyzed by RT-qPCR.
An ELISA kit (Mouse Albumin ELISA Kit (BETHYL Laboratories) was used to quantify the amount of Albumin secreted.
In the activity measurement of Cyp3a11, the substrate Midazolam was added to the medium, and the secretory amount of Hydroxy midazolam produced by the hydroxylation by Cyp3a11 into the medium for 24 hours was measured by mass spectrometry.
<結果>
発明者らは、マウス成体肝細胞に発現する遺伝子の発現プロファイルを探索し、iPS干渉法を用いて成熟肝細胞を誘導する転写因子の組み合わせの絞り込みを行った。その結果、Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6, Onecut2の6因子の組み合わせが成熟肝細胞の誘導に最適であることを見出した。
図1に、6因子の優位性を示すため、2因子(Hnf4a, Foxa3)のみを導入して誘導した場合とで、肝細胞マーカー遺伝子の発現を比較した。その結果、6因子の導入は、2因子に比べ、顕著に肝細胞マーカー遺伝子を増加させた。Cdx2は中腸・後腸マーカー遺伝子(腸管マーカー遺伝子 肝臓は後方前腸由来)であり、既報(Morris et al Cell 2014 Aug 14;158(4):889-902)で2因子から作製したiHepsはCdx2 positive細胞であることが示されたが、6因子を用いることで、Cdx2の発現は著明に低下した。また、Afpは肝芽細胞マーカーであるが、6因子を用いることで、Afpの発現も低下したことから、より成熟した肝細胞に近いiHepsが得られたことが示された。
<Results>
The inventors searched for the expression profile of genes expressed in adult mouse hepatocytes and narrowed down the combination of transcription factors that induce mature hepatocytes using iPS interference method. As a result, it was found that the combination of 6 factors of Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6, and Onecut2 is optimal for the induction of mature hepatocytes.
In order to show the superiority of the 6 factors in FIG. 1, the expression of the hepatocyte marker gene was compared with the case of inducing by introducing only 2 factors (Hnf4a, Foxa3). As a result, the introduction of 6 factors markedly increased the hepatocyte marker gene compared to 2 factors. Cdx2 is a mid-intestinal/hind-gut marker gene (intestinal marker gene liver derived from posterior foregut), and iHeps produced from two factors in the previous report (Morris et al Cell 2014 Aug 14;158(4):889-902) Although it was shown that the cells were Cdx2 positive cells, the expression of Cdx2 was markedly reduced by using 6 factors. Although Afp is a hepatoblast marker, the use of factor 6 also reduced Afp expression, indicating that iHeps closer to that of more mature hepatocytes was obtained.
次に、6因子を導入したのちの培養条件を最適化するため、肝細胞を効率的に誘導できる培地成分の検討を行ったところ、TGFβ受容体阻害剤であるA83-01の添加が有効であることを見出した。hepatic maturation培地にA83-01を添加した場合と添加しない場合で培養を行って得られた肝細胞におけるマーカーの発現を調べた結果を図2、3に示す。その結果、A83-01の添加はアルブミン分泌を増加させた。
一方、A83-01はVimentin、Col1a1、Afpなどの脱分化因子の発現を低下させ、肝細胞の分化効率を上げることが分かった。
Next, in order to optimize the culture conditions after the introduction of the 6 factors, we examined the medium components that can efficiently induce hepatocytes, and found that the addition of the TGFβ receptor inhibitor A83-01 was effective. I found that there is. 2 and 3 show the results of investigating the expression of markers in hepatocytes obtained by culturing with and without addition of A83-01 to the hepatic maturation medium. As a result, addition of A83-01 increased albumin secretion.
On the other hand, it was found that A83-01 decreases the expression of dedifferentiation factors such as Vimentin, Col1a1, Afp, etc., and increases the differentiation efficiency of hepatocytes.
以下、6因子とともにMyc遺伝子を導入して誘導し、hepatic maturation培地にA83-01を添加して培養して得られた肝細胞を用いて実験を行った。
図4には、6因子およびMycの強制発現により得られたiHeps (6TF-Myc iHeps) の免疫染色結果を示す。Albumin陽性の上皮様細胞(6TF-Myc iHeps)が敷石状に拡がっていることがわかる。
Hereinafter, experiments were carried out using hepatocytes obtained by introducing the Myc gene together with 6 factors to induce the cells, and adding A83-01 to the hepatic maturation medium for culturing.
FIG. 4 shows the results of immunostaining of iHeps (6TF-Myc iHeps) obtained by forced expression of 6 factors and Myc. It can be seen that the albumin-positive epithelial-like cells (6TF-Myc iHeps) spread like cobblestones.
図5では、6TF-Myc iHepsを用い、培地中への24時間あたりのAlbumin分泌量をELISAにより測定した。なお、Albumin分泌量は、免疫染色でAlbumin陽性となる細胞数で補正して示した。その結果、6因子にMycを組み合わせた場合、得られる誘導肝細胞におけるアルブミン分泌量は6因子の場合に比べても顕著に多く、初代培養肝細胞に匹敵するほどであった。 In FIG. 5, 6TF-Myc iHeps was used to measure the amount of albumin secreted into the medium for 24 hours by ELISA. The amount of Albumin secretion was corrected and shown by the number of cells that become Albumin-positive by immunostaining. As a result, when Myc was combined with 6 factors, the amount of albumin secretion in the obtained induced hepatocytes was remarkably higher than that of 6 factors, which was comparable to that of primary cultured hepatocytes.
図6では、肝細胞による薬物代謝において、主要な役割を果たすCytochrome P450 (CYP)のうち、代表的なCyp3a11の酵素活性を、各誘導肝細胞又は初代培養肝細胞で比較した。Hydroxy midazolamの分泌量は、免疫染色でAlbumin陽性となる細胞数で補正した。代謝産物であるHydroxy midazolamの産生量が多いほど、酵素活性が高いことを意味し、より成熟化した肝細胞機能を有することを示す。図6によれば、6因子にMycを組み合わせた場合、得られる誘導肝細胞における酵素活性は6因子の場合に比べても顕著に高く、初代培養肝細胞に匹敵するほどであった。 In FIG. 6, of the Cytochrome P450s (CYPs) that play a major role in drug metabolism by hepatocytes, the representative enzyme activity of Cyp3a11 was compared in each induced hepatocyte or primary cultured hepatocyte. The amount of hydroxymidazolam secreted was corrected by the number of cells that became albumin positive by immunostaining. The higher the production of the metabolite Hydroxymidazolam, the higher the enzyme activity, indicating that it has a more mature hepatocyte function. According to FIG. 6, when Myc was combined with 6 factors, the enzyme activity in the obtained induced hepatocytes was remarkably higher than that in the case of 6 factors, which was comparable to that of primary cultured hepatocytes.
図7では、6TF-Myc iHepsを用い、種々の薬剤に対するCYP遺伝子の発現誘導を調べた。その結果、RifampicinではCyp3a11が誘導され、OmeprazoleではCyp1a2が誘導され、TCP0B0PではCyp2b10が誘導され、得られた誘導肝細胞は、薬剤に対応したCYPを活性化し、正常肝細胞の応答プロファイルを有していることが分かった。 In FIG. 7, 6TF-Myc iHeps was used to examine the expression induction of the CYP gene for various drugs. As a result, Cif3a11 was induced in Rifampicin, Cyp1a2 was induced in Omeprazole, Cyp2b10 was induced in TCP0B0P, and the obtained induced hepatocytes activated CYP corresponding to the drug and had a response profile of normal hepatocytes. I found out.
[実施例2]
6因子の必要性の検討
以下の手順で6因子のうちの2~6因子を導入し、肝細胞分化を誘導した。
iHep induction
1. Viral transduction前日 (day-1)、凍結保存していたMEFsを起眠し、Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat# 354230) でコーティングした12-well plate (Greiner) 上に播種 (5.0 × 10cells/well)。
2. Viral transduction当日(day0)、Plat-Eの培養上清をウイルス上清として回収し、filtering (Millex-HP Syringe Filter Unit 0.45 μm, Merck Millipore, Cat# SLHP033RS)を行った。
3. 各転写因子のウイルス上清を各100 μl, Polybrene (nacalai tesque, Cat# 12996-81) 0.6 μl を加え、そのうち500 μlを、前日MEFsを播種した12-well plateの1wellあたりに加えた。
4. 37℃, 5% CO2 で一晩インキュベートし、ウイルス完成を行った。
5. 翌日 (day1) 培地交換 (10% FBS/DMEM)した。
6. Day3にも培地交換 (10% FBS/DMEM)した。 
7. Day4で10% FBS/DMEM/F12, GlutaMAXをベースにしたHepatic specification培地 (DMEM/F12, GlutaMAX (Thermo Fisher Scientific, Cat# 10565042) 360 ml, FBS 40 ml, Nicotinamide (STEMCELL Technologies, Cat# 07154) 10mM, ITS-X (Thermo Fisher Scientific, Cat# 51500056) 400 μl, Dexamethasone (Merck, Cat# D4902-25MG) 0.1 μM, 2-Mercaptoethanol (Thermo Fisher Scientific, Cat# 21985023) 50 μM, Penicillin-Streptomycin (Thermo Fisher Scientific, Cat# 15140122) 2 ml)に培地交換した。さらに2日に一度培地交換した。
8. Day11から、hepatic maturation培地(HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ml)に変更して、2日に一度培地交換した。Day19前後で実験に供した。
[Example 2]
Examination of necessity of 6 factors 2 to 6 factors out of 6 factors were introduced by the following procedure to induce hepatocyte differentiation.
iHep induction
1. Viral transduction The day before (day-1), the MEFs that had been cryopreserved were put to sleep and placed on a 12-well plate (Greiner) coated with Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat# 354230). Seeding (5.0 × 10 4 cells/well).
2. On the day of Viral transduction (day 0), the culture supernatant of Plat-E was collected as a virus supernatant and subjected to filtering (Millex-HP Syringe Filter Unit 0.45 μm, Merck Millipore, Cat# SLHP033RS).
3. 100 μl of virus supernatant of each transcription factor and 0.6 μl of Polybrene (nacalai tesque, Cat# 12996-81) were added, and 500 μl of them was added per well of 12-well plate seeded with MEFs the day before. ..
4. Incubation was carried out overnight at 37°C, 5% CO 2 to complete the virus.
5. The next day (day 1), the medium was replaced (10% FBS/DMEM).
6. On Day 3, the medium was exchanged (10% FBS/DMEM).
7. Day 4 Hepatic specification medium based on 10% FBS/DMEM/F12, GlutaMAX (DMEM/F12, GlutaMAX (Thermo Fisher Scientific, Cat# 10565042) 360 ml, FBS 40 ml, Nicotinamide (STEMCELL Technologies, Cat# 07154 ) 10mM, ITS-X (Thermo Fisher Scientific, Cat# 51500056) 400 μl, Dexamethasone (Merck, Cat# D4902-25MG) 0.1 μM, 2-Mercaptoethanol (Thermo Fisher Scientific, Cat# 21985023) 50 μM, Penicillin-Streptomycin ( The medium was replaced with Thermo Fisher Scientific, Cat# 15140122) 2 ml). The medium was exchanged once every two days.
8. From Day 11, hepatic maturation medium (HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ ml) and the medium was exchanged once every two days. It was used for experiments around Day 19.
<結果>
図8に示すように、6因子のうち1つでも欠けると、肝細胞へのconversion 効率が大幅に落ちることが確認できた。なお、本実施例ではMyc遺伝子は導入しておらず、Myc遺伝子はなくとも6因子で肝細胞分化が効率よく実施できることが示された。
<Results>
As shown in FIG. 8, it was confirmed that if even one of the six factors was missing, the conversion efficiency for hepatocytes was significantly reduced. In this Example, the Myc gene was not introduced, and it was shown that hepatocyte differentiation can be efficiently performed with 6 factors without the Myc gene.
[実施例3]
次に、ヒト体細胞を用いて以下の手順で肝細胞分化を誘導した。
iHep induction
1. Viral transduction前日 (day-1)、凍結保存していたHDF-Slc7a1(Yamakawa, Stem Cells 2016; gift from Okita lab.)を起眠し、Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat# 354230) でコーティングした12-well plate (Greiner) 上に播種 (5.0 × 10cells/well)した。
2. Viral transduction当日(day0)、Plat-Eの培養上清をウイルス上清として回収し、filtering (Millex-HP Syringe Filter Unit 0.45 μm, Merck Millipore, Cat# SLHP033RS)した。
3. 各転写因子のウイルス上清を各100 μl, c-Myc 12.5 μl, 10% FBS/DMEM 87.5 μl, Polybrene (nacalai tesque, Cat# 12996-81) 0.7 μl を加え、そのうち500 μlを、前日HDF-Slc7a1を播種した12-well plateの1wellあたりに加えた。
4. 37℃, 5% CO2 で一晩インキュベートし、ウイルス完成を行った。
5. 翌日 (day1) 培地交換 (10% FBS/DMEM)した。
6. Day3にも培地交換 (10% FBS/DMEM)した。 
7. Day4に、Matrigelでコーティングした12-well plate上に1:10の割合で再播種した。培地は10% FBS/DMEM/F12, GlutaMAXをベースにしたHepatic specification培地 (DMEM/F12, GlutaMAX (Thermo Fisher Scientific, Cat# 10565042) 360 ml, FBS 40 ml, Nicotinamide (STEMCELL Technologies, Cat# 07154) 10mM, ITS-X (Thermo Fisher Scientific, Cat# 51500056) 400 μl, Dexamethasone (Merck, Cat# D4902-25MG) 0.1 μM, 2-Mercaptoethanol (Thermo Fisher Scientific, Cat# 21985023) 50 μM, Penicillin-Streptomycin (Thermo Fisher Scientific, Cat# 15140122) 2 ml)に培地交換した。
8. Day6から、hepatic maturation培地(HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ml)に変更して、2日に一度培地交換した。Day20前後で実験に供した。
[Example 3]
Next, hepatocyte differentiation was induced by the following procedure using human somatic cells.
iHep induction
1. Viral transduction The day before (day-1), the frozen HDF-Slc7a1 (Yamakawa, Stem Cells 2016; gift from Okita lab.) was put to sleep, and Matrigel (BD Matrigel Matrix Growth Factor Reduced, BD Biosciences, Cat Seed (5.0 × 10 4 cells/well) on a 12-well plate (Greiner) coated with #354230).
2. On the day of Viral transduction (day 0), the culture supernatant of Plat-E was collected as a virus supernatant and filtered (Millex-HP Syringe Filter Unit 0.45 μm, Merck Millipore, Cat# SLHP033RS).
3. Add 100 μl, c-Myc 12.5 μl, 10% FBS/DMEM 87.5 μl, Polybrene (nacalai tesque, Cat# 12996-81) 0.7 μl of each viral supernatant of each transcription factor, and 500 μl of which was added the day before. HDF-Slc7a1 was added to each well of a 12-well plate seeded.
4. Incubation was carried out overnight at 37°C, 5% CO 2 to complete the virus.
5. The next day (day 1), the medium was replaced (10% FBS/DMEM).
6. On Day 3, the medium was exchanged (10% FBS/DMEM).
7. On Day 4, reseeding was performed at a ratio of 1:10 on a 12-well plate coated with Matrigel. Hepatic specification medium based on 10% FBS/DMEM/F12, GlutaMAX (DMEM/F12, GlutaMAX (Thermo Fisher Scientific, Cat# 10565042) 360 ml, FBS 40 ml, Nicotinamide (STEMCELL Technologies, Cat# 07154) 10 mM , ITS-X (Thermo Fisher Scientific, Cat# 51500056) 400 μl, Dexamethasone (Merck, Cat# D4902-25MG) 0.1 μM, 2-Mercaptoethanol (Thermo Fisher Scientific, Cat# 21985023) 50 μM, Penicillin-Streptomycin (Thermo Fisher The medium was replaced with Scientific, Cat# 15140122) 2 ml).
8. From Day 6, hepatic maturation medium (HCM BulletKit Medium (Lonza, Cat# CC-3198), HGF (Peprotech, Cat# 100-39) 20 ng/ml, OSM (Peprotech, Cat# 300-10) 20 ng/ ml) and the medium was exchanged once every two days. It was used for experiments around Day 20.
<結果>
図9に示すように、ヒト細胞を用いても同様にアルブミン陽性の肝細胞を誘導できることが分かった。
<Results>
As shown in FIG. 9, it was found that albumin-positive hepatocytes can be similarly induced using human cells.

Claims (12)

  1. Hnf4a(Hepatocyte nuclear factor 4 alpha)、Foxa3(Forkhead Box A3)、Cebpa(CCAAT/enhancer-binding protein alpha)、Cebpd(CCAAT/enhancer-binding protein delta)、Hnf6(Hepatocyte nuclear factor 6)およびOnecut2(Onecut domain family member 2)が導入された体細胞を用意する工程、および
    当該体細胞を培養して肝細胞へと誘導する工程、を含む、肝細胞の製造方法。
    Hnf4a (Hepatocyte nuclear factor 4 alpha), Foxa3 (Forkhead Box A3), Cebpa (CCAAT/enhancer-binding protein alpha), Cebpd (CCAAT/enhancer-binding protein delta), Hnf6 (Hepatocyte nuclear factor 6) and Onecut2 (Onecut domain) A method for producing hepatocytes, comprising a step of preparing somatic cells into which family member 2) has been introduced, and a step of culturing the somatic cells to induce them into hepatocytes.
  2. 体細胞が線維芽細胞である、請求項1に記載の肝細胞の製造方法。 The method for producing hepatocytes according to claim 1, wherein the somatic cells are fibroblasts.
  3. 体細胞がヒトまたはマウス由来である、請求項1または2に記載の肝細胞の製造方法。 The method for producing hepatocytes according to claim 1 or 2, wherein the somatic cells are of human or mouse origin.
  4. Hnf4a、Foxa3、Cebpa、Cebpd、Hnf6およびOnecut2がヒトまたはマウス由来である、請求項1~3のいずれか一項に記載の肝細胞の製造方法。 The method for producing hepatocytes according to any one of claims 1 to 3, wherein Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are derived from human or mouse.
  5. Hnf4a、Foxa3、Cebpa、Cebpd、Hnf6およびOnecut2がレトロウイルスベクターを用いて導入された、請求項1~4のいずれか一項に記載の肝細胞の製造方法。 The method for producing hepatocytes according to any one of claims 1 to 4, wherein Hnf4a, Foxa3, Cebpa, Cebpd, Hnf6 and Onecut2 are introduced using a retrovirus vector.
  6. 前記体細胞は、さらにMycが導入されている、請求項1~5のいずれか一項に記載の肝細胞の製造方法。 The method for producing hepatocytes according to any one of claims 1 to 5, wherein Myc is further introduced into the somatic cells.
  7. 前記培養工程が20日以上行われる、請求項1~6のいずれか一項に記載の肝細胞の製造方法。 The method for producing hepatocytes according to any one of claims 1 to 6, wherein the culturing step is performed for 20 days or more.
  8. 前記培養工程において培地はTGFβ(Transforming growth factor beta)受容体阻害剤を含む、請求項1~7のいずれか一項に記載の肝細胞の製造方法。 The method for producing hepatocytes according to any one of claims 1 to 7, wherein in the culture step, the medium contains a TGFβ (Transforming growth factor beta) receptor inhibitor.
  9. TGFβ受容体阻害剤はA83-01である、請求項8に記載の肝細胞の製造方法。 The method for producing hepatocytes according to claim 8, wherein the TGFβ receptor inhibitor is A83-01.
  10. 請求項1~9のいずれか一項に記載の方法によって製造された肝細胞。 A hepatocyte produced by the method according to any one of claims 1 to 9.
  11. 請求項10に記載の肝細胞を含む、再生医療用細胞製剤。 A cell preparation for regenerative medicine, comprising the hepatocyte according to claim 10.
  12. 下記工程を(i)-(iii)の順番で含む、被検物質の毒性評価方法;
    (i)請求項1~9のいずれか一項に記載の方法によって製造された肝細胞に被検物質を接触させる工程、
    (ii)前記肝細胞の生存率または培養上清中の肝障害マーカー量を測定する工程、および
    (iii)前記被検物質と接触させた肝細胞の生存率が、被検物質と接触させなかった肝細胞(対照細胞)の生存率よりも低値、または、前記被検物質と接触させた肝細胞の培養上清中の肝障害マーカー量が、対照細胞の培養上清中の肝障害マーカー量よりも高値である被検物質を、肝毒性を有すると評価する工程。
    A method for evaluating toxicity of a test substance, which comprises the following steps in the order of (i)-(iii);
    (I) a step of bringing a test substance into contact with hepatocytes produced by the method according to any one of claims 1 to 9,
    (Ii) the step of measuring the survival rate of the hepatocytes or the amount of a liver injury marker in the culture supernatant, and (iii) the survival rate of the hepatocytes contacted with the test substance does not contact with the test substance. Lower than the survival rate of the hepatocytes (control cells), or the amount of the liver injury marker in the culture supernatant of the hepatocytes contacted with the test substance is the liver injury marker in the culture supernatant of the control cells. A step of evaluating a test substance having a higher value than the amount as having hepatotoxicity.
PCT/JP2019/050926 2018-12-26 2019-12-25 Method for producing hepatocytes WO2020138208A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2020563365A JP7452799B2 (en) 2018-12-26 2019-12-25 Method for producing hepatocytes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2018-242917 2018-12-26
JP2018242917 2018-12-26

Publications (1)

Publication Number Publication Date
WO2020138208A1 true WO2020138208A1 (en) 2020-07-02

Family

ID=71128692

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2019/050926 WO2020138208A1 (en) 2018-12-26 2019-12-25 Method for producing hepatocytes

Country Status (2)

Country Link
JP (1) JP7452799B2 (en)
WO (1) WO2020138208A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011102532A1 (en) * 2010-02-16 2011-08-25 国立大学法人九州大学 Induced hepatocytes
JP2015527084A (en) * 2012-09-07 2015-09-17 ジェネンテック, インコーポレイテッド Methods and compositions for producing induced hepatocytes
JP2017511150A (en) * 2014-02-12 2017-04-20 北昊干細胞与再生医学研究院有限公司BeiHao Stem Cell and Regenerative Medicine Translational Research Institute Kits and methods for reprogramming non-hepatocytes to hepatocytes

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5722885B2 (en) 2009-06-18 2015-05-27 セレクティス アーベー 3D culture system for growth and differentiation of human pluripotent stem (hPS) cells
WO2012141038A1 (en) 2011-04-15 2012-10-18 国立大学法人鳥取大学 Synthesis and analysis of novel compound capable of inducing differentiation of human mesenchymal stem cell into hepatocyte
WO2013183571A1 (en) 2012-06-08 2013-12-12 公立大学法人名古屋市立大学 Method for inducing differentiation of artificial pluripotent stem cell into hepatocyte

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011102532A1 (en) * 2010-02-16 2011-08-25 国立大学法人九州大学 Induced hepatocytes
JP2015527084A (en) * 2012-09-07 2015-09-17 ジェネンテック, インコーポレイテッド Methods and compositions for producing induced hepatocytes
JP2017511150A (en) * 2014-02-12 2017-04-20 北昊干細胞与再生医学研究院有限公司BeiHao Stem Cell and Regenerative Medicine Translational Research Institute Kits and methods for reprogramming non-hepatocytes to hepatocytes

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DU, Y. ET AL.: "Human Hepatocytes with Drug Metabolic Function Induced from Fibroblasts by Lineage Reprogramming", CELL STEM CELL, vol. 14, 2014, pages 394 - 403, XP055194340, DOI: 10.1016/j.stem.2014.01.008 *
HUANG, P. ET AL.: "Direct Reprogramming of Human Fibroblasts to Functional and Expandable Hepatocytes", CELL STEM CELL, vol. 14, 2014, pages 370 - 384, XP055194338, DOI: 10.1016/j.stem.2014.01.003 *
HUANG, P. ET AL.: "Induction of functional hepatocyte-like cells from mouse fibroblasts by defined factors", NATURE, vol. 475, 2011, pages 386 - 389, XP055086662, DOI: 10.1038/nature10116 *
SEKIYA, S. ET AL.: "Direct conversion of mouse fibroblasts to hepatocyte-like cells by defined factors", NATURE, vol. 475, 2011, pages 390 - 393, XP055086663, DOI: 10.1038/nature10263 *
YASUDA, KATSUTARO.: "Identification of Master Transcription Factor for Inducing Mature Hepatocyte Differentiation with Applied Reprogramming Technology.", PROGRAM ABSTRACTS OF THE 17TH CONGRESS OF THE JAPANESE SOCIETY FOR REGENERATIVE MEDICINE., 23 February 2018 (2018-02-23), pages 439 *
YU , B. ET AL.: "Reprogramming Fibroblasts into Bipotential Hepatic Stem Cells by Defined Factors", CELL STEM CELL, vol. 13, 2013, pages 328 - 340, XP055194339, DOI: 10.1016/j.stem.2013.06.017 *

Also Published As

Publication number Publication date
JP7452799B2 (en) 2024-03-19
JPWO2020138208A1 (en) 2021-11-18

Similar Documents

Publication Publication Date Title
CN108350429B (en) Method for directed differentiation of pluripotent stem cells into immune cells
US9260722B2 (en) Hepatocyte production by forward programming
US20140242595A1 (en) Hepatocyte production via forward programming by combined genetic and chemical engineering
EP3633026A1 (en) Method for inducing differentiation of intermediate mesodermal cell to renal progenitor cell, and method for inducing differentiation of pluripotent stem cell to renal progenitor cell
US20170107486A1 (en) Hepatocyte production via forward programming by combined genetic and chemical engineering
JP5751548B2 (en) Canine iPS cells and production method thereof
US20210147869A1 (en) Reprogramming vectors
JP2021192627A (en) Method for inducing hepatocytes
JP2015146803A (en) Method for inducing differentiation of melanocyte
EP4324917A1 (en) Cell bank composed of ips cells for introducing t cell receptor gene
WO2020138208A1 (en) Method for producing hepatocytes
WO2011111588A1 (en) Method of inducing the differentiation of germline stem cells, method of expanding the cells, and culture media therefor
WO2014168255A1 (en) Megakaryocyte maturation accelerator
EP3984549A1 (en) Medicinal composition
EP4130253A1 (en) T cell progenitor production method
JP2022534927A (en) Compositions and uses of engineered myogenic cells
US20240218332A1 (en) Methods of generating mature hepatocytes
EP4386083A1 (en) Method for producing renal interstitial progenitor cells, erythropoietin-producing cells, and method for producing renin-producing cells
EP4273234A1 (en) Method for producing regenerated t cell via ips cell
TW202417612A (en) Transdifferentiation of non-dermal papilla cells to dermal papilla cells
Guddati Derivation of cardiomyocytes from embryonic stem cells and development of techniques to study cardiac lineage

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: 19905335

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2020563365

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19905335

Country of ref document: EP

Kind code of ref document: A1