WO2022102742A1 - Marqueur de surface cellulaire pour une purification à haut rendement de cellules de lignée de muscle squelettique et de cellules souches de muscle squelettique, et son utilisation - Google Patents

Marqueur de surface cellulaire pour une purification à haut rendement de cellules de lignée de muscle squelettique et de cellules souches de muscle squelettique, et son utilisation Download PDF

Info

Publication number
WO2022102742A1
WO2022102742A1 PCT/JP2021/041709 JP2021041709W WO2022102742A1 WO 2022102742 A1 WO2022102742 A1 WO 2022102742A1 JP 2021041709 W JP2021041709 W JP 2021041709W WO 2022102742 A1 WO2022102742 A1 WO 2022102742A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
skeletal muscle
cell
positive
cdh13
Prior art date
Application number
PCT/JP2021/041709
Other languages
English (en)
Japanese (ja)
Inventor
英俊 櫻井
ハルチウン ミナス ナルバンディアン
明明 趙
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 JP2022562200A priority Critical patent/JPWO2022102742A1/ja
Publication of WO2022102742A1 publication Critical patent/WO2022102742A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues

Definitions

  • iPS cells have greater proliferative and differentiating potential and are therefore considered to be a very attractive cell source for generating various cell lines for cell therapy.
  • Non-Patent Document 1 a method for inducing skeletal muscle stem cells capable of muscle regeneration from human iPS cells (Non-Patent Document 1), focusing on the MYF5 gene known as a satellite cell marker, and MYF5 positive.
  • MYF5-negative cells When cells and MYF5-negative cells were transplanted into the shin muscles of immunodeficient mice, it was suggested that Myf5-positive cells had a higher degree of muscle fiber formation after transplantation and had higher muscle regeneration ability.
  • the present invention provides a biomarker for obtaining a highly pure skeletal muscle lineage cell population and a skeletal muscle stem cell population, and provides a method for improving the purity of skeletal muscle lineage cells and skeletal muscle stem cells using the biomarker and a method for producing the same.
  • the challenge is to provide.
  • the present invention includes the following inventions in order to solve the above problems.
  • a method for improving the purity of skeletal muscle lineage cells in a cell population which comprises recovering cell surface marker CDH13-positive cells in the cell population.
  • the cell population is a cell population in the process of inducing differentiation from pluripotent stem cells to skeletal muscle cells.
  • the pluripotent stem cell is an iPS cell.
  • a method for producing skeletal muscle stem cells which is (1) a step of initiating differentiation induction from pluripotent stem cells to skeletal muscle cells, and (2) positive for the cell surface marker FGFR4 from a cell population in the process of inducing differentiation.
  • a production method comprising the steps of recovering cells, CDH13-positive cells or FGFR4-positive and CDH13-positive cells.
  • step (1) is a step of inducing differentiation of skeletal muscle lineage cells from pluripotent stem cells so as to mimic the developmental process of a fetus.
  • step (1) is a step of inducing differentiation of skeletal muscle lineage cells from pluripotent stem cells so as to mimic the developmental process of a fetus.
  • the pluripotent stem cell is an iPS cell.
  • the skeletal muscle stem cells are for living-donor transplantation.
  • Human iPS cells (201B7) were used to induce differentiation into skeletal muscle stem cells, and on day 84, the cells were collected and stained with anti-FGFR4 antibody, and FGFR4 positive cells and FGFR4 negative cells were sorted by FACS, respectively. It is a figure which shows the result of staining with the CDH13 antibody and analyzing the CDH13 positive rate by FACS.
  • Human iPS cells (201B7) were used to induce differentiation into skeletal muscle stem cells, cells were harvested on day 84, CDH13-positive cells and FGFR4-positive cells were sorted by FACS, and CDH13-positive cells, FGFR4-positive cells and non-CDH13-positive cells were sorted.
  • Human iPS cells were used to induce differentiation into skeletal muscle stem cells, cells were harvested on day 84, CDH13-positive and FGFR4-positive cells were sorted by FACS, and CDH13-positive, FGFR4-positive and non-CDH13-positive cells were sorted. It is a figure which shows the result of having measured the expression level of MYF5, PAX7, MYOD1 in a sorting cell.
  • Human iPS cells (201B7, CKI and DMD) were used to induce differentiation into skeletal muscle stem cells, and on day 84, the cells were collected and stained with anti-CDH13 antibody, and CDH13-positive cells and CDH13-negative cells were isolated by FACS, respectively.
  • FIG. 1 It is a figure which shows the result of having examined the expression of myosin heavy chain (MHC) by sorting and further inducing the differentiation of CDH13 positive cell and CDH13 negative cell into skeletal muscle cells, and (A) is the result of immunostaining MHC (A).
  • B) is a diagram showing the results of calculating the differentiation index (MHC positive cell rate) by image analysis.
  • Human iPS cells (201B7, CKI and DMD) were used to induce differentiation into skeletal muscle stem cells, and on day 84, the cells were collected and stained with anti-FGFR4 antibody, and FGFR4 positive cells and FGFR4 negative cells were separated by FACS, respectively.
  • the transplantation site was collected, stained with anti-histopeclin antibody, anti-laminin ⁇ 2 antibody, anti-human nuclear antibody and DAPI, and observed under a confocal microscope. It is a figure which shows the result. It is a figure which shows the result of having measured the number of engraftment cells in the transplantation site collected from the above-mentioned NOG-mdx mouse 4 weeks after transplantation.
  • the above-mentioned transplantation site collected from NOG-mdx mice 4 weeks after transplantation was stained with anti-PAX7 antibody, anti-MYOD antibody, anti-h-LAMIN A / C antibody and DAPI, and observed with a confocal microscope. be.
  • Human iPS cells (DMD, CKI) were used to induce differentiation into skeletal muscle stem cells, cells were collected on day 84, and CD56-positive cell rate, CD82-positive cell rate, and NGFR-positive cell rate were used using FACS.
  • the present invention provides a method for improving the purity of skeletal muscle lineage cells.
  • Skeletal muscle lineage cells are a broad concept that includes cells capable of skeletal muscle differentiation and cells that have finally differentiated into skeletal muscle, and are skeletal muscle stem cells, myoblasts, myotube cells, mature myotube cells, and skeletal muscle cells ( Includes all muscle fibers).
  • the present inventors have found that skeletal muscle lineage cells express CDH13 on the cell surface, and have found that a high-purity skeletal muscle lineage cell population can be obtained by using the cell surface marker CDH13. Therefore, the method for improving the purity of skeletal muscle lineage cells of the present invention is characterized by using the cell surface marker CDH13.
  • CDH13 is also referred to as cadherin-13, T-cadherin, and H-cadherin.
  • the method for improving the purity of skeletal muscle lineage cells of the present invention may be any method including a step of recovering cell surface marker CDH13-positive cells from a cell population.
  • the cell population is not particularly limited as long as it is a cell population that may contain skeletal muscle lineage cells.
  • a cell suspension prepared from a surgical specimen containing skeletal muscle can be mentioned.
  • the cell population may be a human cell population or a cell population of a non-human organism.
  • Organisms other than humans are not particularly limited and may be, for example, mammals. Examples of mammals include monkeys, chimpanzees, dogs, cats, cows, horses, pigs, rabbits, mice, rats and the like.
  • the cell population may be a cell population in the process of inducing differentiation from pluripotent stem cells to skeletal muscle cells.
  • a known method for inducing differentiation from pluripotent stem cells to skeletal muscle cells is appropriately selected to initiate differentiation induction, and before the final differentiation is reached.
  • Cell population can be used.
  • a known method for inducing differentiation of pluripotent stem cells into skeletal muscle cells for example, the method described in Non-Patent Document 1 (Zhao et al., Stem Cell Reports, Vol. 15 1-15 July 14, 2020.). , International release WO2016 / 108288 The method described in A1, Hicks et al. (Nat Cell Biol. 2018 Jan; 20 (1): 46-57.), Etc. can be mentioned.
  • the method for recovering CDH13-positive cells from the cell population is not particularly limited, and examples thereof include a method using a cell sorter and a method using affinity chromatography.
  • a cell sorter for example, a cell suspension of a cell population is prepared, an anti-CDH13 antibody is added to the cell suspension, the anti-CDH13 antibody is bound to the CDH13-positive cells, and the cells are subjected to the cell sorter to obtain the CDH13-positive cells. Can be sorted and collected.
  • a cell suspension of a cell population is prepared, and the CDH13-positive cells are bound to the anti-CDH13 antibody by passing the cell suspension through a column filled with a carrier to which the anti-CDH13 antibody is bound. After that, CDH13-positive cells can be recovered by dissociating the binding with the anti-CDH13 antibody.
  • the method for improving the purity of skeletal muscle lineage cells of the present invention reduces the proportion of CDH13-negative cells in the original cell population and increases the proportion of CDH13-positive cells, so that the resulting cell population improves the purity of skeletal muscle lineage cells. It is a cell population.
  • the pluripotent stem cell that can be used in the present invention is not particularly limited as long as it is a stem cell that has pluripotency capable of differentiating into all cells existing in a living body and also has proliferative ability.
  • embryonic stem (ES) cells embryonic stem (ntES) cells derived from cloned embryos obtained by nuclear transplantation
  • sperm stem (GS) cells embryonic reproductive (EG) cells
  • artificial pluripotent stem (iPS) cells derived from cultured fibroblasts and bone marrow stem cells.
  • Pluripotent cells are ES cells, ntES cells, and iPS cells.
  • Embryonic stem cells ES cells are pluripotent and self-replicating stem cells established from the inner cell mass of early embryos (eg, blastocysts) of mammals such as humans and mice.
  • ES cells are embryo-derived stem cells derived from the inner cell mass of the scutellum vesicle, which is the embryo after the morula at the 8-cell stage of the fertilized egg, and have the ability to differentiate into all the cells that make up the adult, so-called polymorphism. It has the ability and the ability to proliferate by self-replication.
  • ES cells were discovered in mice in 1981 (M.J. Evans and M.H. Kaufman (1981), Nature 292: 154-156), and then ES cell lines were established in primates such as humans and monkeys (J.A. Thomson et). al. (1998), Science 282: 1145-1147; J.A. Thomson et al. (1995), Proc. Natl. Acad. Sci.
  • ES cells For the establishment of ES cells, a method known in this field is used. For example, it can be established by removing the inner cell mass from the blastocyst of a fertilized egg of a target animal and culturing the inner cell mass on a fibroblast feeder. For cell maintenance by subculture, use a culture medium supplemented with substances such as leukemia inhibitory factor (LIF) and basic fibroblast growth factor (bFGF). It can be carried out.
  • LIF leukemia inhibitory factor
  • bFGF basic fibroblast growth factor
  • a culture method for producing ES cells a method known in the art is used.
  • the culture medium use DMEM / F-12 culture medium supplemented with, for example, 0.1 mM 2-mercaptoethanol, 0.1 mM non-essential amino acid, 2 mM L-glutamic acid, 20% KSR (KnockOut Serum Replacement, Invitrogen) and 4 ng / ml bFGF.
  • human ES cells can be maintained in a moist atmosphere of 37 ° C, 2% CO 2 / 98% air (O. Fumitaka et al. (2008), Nat. Biotechnol., 26: 215-224).
  • ES cells may be passaged every 3-4 days, using 0.25% trypsin and 0.1 mg / ml collagenase IV in PBS containing, for example, 1 mM CaCl 2 and 20% KSR. Can be done.
  • ES cells can generally be selected by the Real-Time PCR method using the expression of gene markers such as alkaline phosphatase, Oct-3 / 4, and Nanog as an index.
  • gene markers such as alkaline phosphatase, Oct-3 / 4, and Nanog as an index.
  • the expression of gene markers such as OCT-3 / 4, NANOG, and ECAD can be used as an index (E. Croon et al. (2008), Nat. Biotechnol., 26: 443. -452).
  • mouse ES cells various mouse ES cell lines established by inGenious, RIKEN (RIKEN), etc. can be used.
  • human ES cells various human ES cell lines established by the National Institutes of Health (NIH), RIKEN, Kyoto University, and Cellartis can be used.
  • ES cell lines NIH CHB-1 to CHB-12 strains, RUES1 strains, RUES2 strains, HUES1 to HUES28 strains, etc.
  • WisCell Research Institute WA01 (H1) strains, WA09 (H9) strains, RIKEN KhES- One strain, KhES-2 strain, KhES-3 strain, KhES-4 strain, KhES-5 strain, SSES1 strain, SSES2 strain, SSES3 strain and the like can be used.
  • the KhES-1, KhES-2, KhES-3 and KthES11 strains are available from the Institute for Frontier Life and Medical Sciences, Kyoto University (Kyoto, Japan).
  • sperm stem cells are pluripotent stem cells derived from the testis and are the origin cells for spermatogenesis. Similar to ES cells, these cells can be induced to differentiate into various lineages of cells, and have properties such as the ability to produce chimeric mice when transplanted into mouse blastocysts (M. Kanatsu-Shinohara et al. (M. Kanatsu-Shinohara et al.). 2003) Biol. Reprod., 69: 612-616; K. Shinohara et al. (2004), Cell, 119: 1001-1012).
  • GDNF glial cell line-derived neurotrophic factor
  • Embryonic germ cells are cells with pluripotency similar to ES cells, which are established from primordial germ cells in the embryonic period, such as LIF, bFGF, and stem cell factor. It can be established by culturing primordial germ cells in the presence of the substance of (Y. Matsui et al. (1992), Cell, 70: 841-847; JL Resnick et al. (1992), Nature, 359: 550. -551).
  • Induced pluripotent stem cells are similar to ES cells, which can be produced by introducing specific reprogramming factors into somatic cells in the form of DNA or protein.
  • Reprogramming factors are genes that are specifically expressed in ES cells, their gene products or non-cording RNAs, or genes that play an important role in maintaining undifferentiated ES cells, their gene products or non-cording RNAs, or It may be composed of low molecular weight compounds.
  • Genes included in the reprogramming factors include, for example, Oct3 / 4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, Klf2, c-Myc, N-Myc, L-Myc, Nanog, Lin28, Fbx15, ERas, ECAT15. -2, Tcl1, beta-catenin, Lin28b, Sall1, Sall4, Esrrb, Nr5a2, Tbx3, Glis1, etc.
  • initialization factors include WO2007 / 069666, WO2008 / 118820, WO2009 / 007852, WO2009 / 032194, WO2009 / 058413, WO2009 / 057831, WO2009 / 075119, WO2009 / 079007, WO2009 / 091659, WO2009 / 101084, WO2009 / 101407, WO2009 / 102983, WO2009 / 114949, WO2009 / 117439, WO2009 / 126250, WO2009 / 126251, WO2009 / 126655, WO2009 / 157593, WO2010 / 009015, WO2010 / 033906, WO2010 / 033920, WO2010 / 042800, WO2010 / 050626, WO 2010/056831, WO2010 / 068955,
  • the reprogramming factors include histone deacetylase (HDAC) inhibitors [eg, small molecule inhibitors such as valproic acid (VPA), tricostatin A, sodium butyrate, MC 1293, M344, siRNA against HDAC and shRNA (eg).
  • HDAC histone deacetylase
  • HDAC1 siRNA Smartpool (Millipore), HuSH 29mer shRNA Constructs against HDAC1 (OriGene), etc.
  • MEK inhibitors eg PD184352, PD98059, U0126, SL327 and PD0325901
  • Glycogen synthase kinase- 3 Against small molecule inhibitors such as inhibitors (eg Bio and CHIR99021), DNA methyltransferase inhibitors (eg 5-azacytidine), histone methyltransferase inhibitors (eg BIX-01294), Suv39hl, Suv39h2, SetDBl and G9a Nucleic acid expression inhibitors such as siRNA and shRNA), L-channel calciumagonist (eg Bayk8644), butyric acid, TGF ⁇ inhibitors or ALK5 inhibitors (eg LY364947, SB431542, 616453 and A-83-01), p53 inhibition Agents (eg siRNA and shRNA for p53), ARID3
  • the reprogramming factor may be introduced into somatic cells by techniques such as lipofection, fusion with cell membrane permeable peptides (eg, HIV-derived TAT and polyarginine), and microinjection.
  • somatic cells by techniques such as lipofection, fusion with cell membrane permeable peptides (eg, HIV-derived TAT and polyarginine), and microinjection.
  • DNA morphology in the case of DNA morphology, it can be introduced into somatic cells by methods such as viruses, plasmids, vectors such as artificial chromosomes, lipofection, liposomes, and microinjection.
  • Viral vectors include retro viral vectors and lentiviral vectors (above, Cell, 126, pp.663-676, 2006; Cell, 131, pp.861-872, 2007; Science, 318, pp.1917-1920, 2007. ), Adenovirus vector (Science, 322, 945-949, 2008), adeno-associated virus vector, Sendai virus vector (WO2010 / 008054) and the like.
  • the artificial chromosome vector includes, for example, a human artificial chromosome (HAC), a yeast artificial chromosome (YAC), a bacterial artificial chromosome (BAC, PAC) and the like.
  • a plasmid a plasmid for mammalian cells can be used (Science, 322: 949-953, 2008).
  • the vector can contain regulatory sequences such as promoters, enhancers, ribosome binding sequences, terminators, polyadenylation sites, etc. so that the nuclear reprogramming substance can be expressed, and further, if necessary, a drug resistance gene (drug resistance gene).
  • canamycin resistance gene for example, ampicillin resistance gene, puromycin resistance gene, etc.
  • thymidin kinase gene diphtheriatoxin gene and other selection marker sequences
  • green fluorescent protein (GFP) green fluorescent protein
  • GUS ⁇ -glucuronidase
  • FLAG FLAG and other reporter gene sequences, etc.
  • the above vector has LoxP sequences before and after introduction into somatic cells in order to excise both the gene encoding the reprogramming factor or the promoter and the gene encoding the reprogramming factor that binds to the promoter. You may.
  • RNA morphology it may be introduced into somatic cells by a method such as lipofection or microinjection, and in order to suppress degradation, RNA incorporating 5-methylcytidine and pseudouridine (TriLink Biotechnologies) is used. It may be (Warren L, (2010) Cell Stem Cell. 7: 618-630).
  • Cultures for iPS cell induction include, for example, DMEM, DMEM / F12 or DME cultures containing 10-15% FBS (these cultures also include LIF, penicillin / streptomycin, puromycin, L-glutamine). , Non-essential amino acids, ⁇ -mercaptoethanol, etc. can be appropriately contained.) Or mouse ES cell culture medium (TX-WES culture medium, Thrombo X), primate ES cell culture medium (primates). Commercially available cultures such as ES / iPS cell culture medium, Reprocell), serum-free pluripotent stem cell maintenance medium (for example, mTeSR (Stemcell Technology), Essential 8 (Life Technologies), StemFit AK03 (AJINOMOTO)) Illustrated.
  • mTeSR StemTeSR
  • Essential 8 Life Technologies
  • StemFit AK03 AJINOMOTO
  • the somatic cells are brought into contact with the reprogramming factor on a DMEM or DMEM / F12 culture medium containing 10% FBS and cultured for about 4 to 7 days. Then, the cells were re-seeded on feeder cells (for example, mitomycin C-treated STO cells, SNL cells, etc.), and about 10 days after the contact between the somatic cells and the reprogramming factor, the culture medium for bFGF-containing primate ES cell culture was used. It can be cultured and give rise to iPS-like colonies about 30-about 45 days or more after the contact.
  • feeder cells for example, mitomycin C-treated STO cells, SNL cells, etc.
  • DMEM culture medium containing 10% FBS for example, LIF, penicillin / streptomycin, etc.
  • feeder cells eg, mitomycin C-treated STO cells, SNL cells, etc.
  • FBS for example, LIF, penicillin / streptomycin, etc.
  • feeder cells eg, mitomycin C-treated STO cells, SNL cells, etc.
  • FBS for example, LIF, penicillin / streptomycin, etc.
  • feeder cells eg, mitomycin C-treated STO cells, SNL cells, etc.
  • FBS for example, LIF, penicillin / streptomycin, etc.
  • ES-like colonies can be generated after about 25 to about 30 days or more.
  • the reprogrammed somatic cells themselves are used (Takahashi K, et al. (2009), PLoS One. 4: e8067 or WO2010 / 137746), or extracellular matrix (eg, Laminin-). 5 (WO2009 / 123
  • iPS cells may be established under hypoxic conditions (oxygen concentration of 0.1% or more and 15% or less) (Yoshida Y, et al. (2009), Cell Stem Cell. 5: 237. -241 or WO2010 / 013845).
  • the fresh culture solution and the culture solution are exchanged once a day from the second day after the start of the culture.
  • the number of somatic cells used for nuclear reprogramming is not limited, but ranges from about 5 ⁇ 10 3 to about 5 ⁇ 10 6 cells per 100 cm 2 culture dish.
  • the iPS cells can be selected according to the shape of the formed colonies.
  • a drug resistance gene expressed in conjunction with a gene expressed when somatic cells are reprogrammed for example, Oct3 / 4, Nanog
  • a culture medium containing the corresponding drug selection.
  • Established iPS cells can be selected by culturing in a culture medium).
  • iPS cells are selected by observing with a fluorescence microscope when the marker gene is a fluorescent protein gene, by adding a luminescent substrate when it is a luminescent enzyme gene, and by adding a chromogenic substrate when it is a chromogenic enzyme gene. can do.
  • the term "somatic cell” refers to any animal cell (eg, mammalian cell including human) except germline cells such as eggs, egg mother cells, ES cells or totipotent cells. .. Somatic cells include, but are not limited to, fetal (pup) somatic cells, neonatal (pup) somatic cells, and mature healthy or diseased somatic cells, as well as primary cultured cells. , Passed cells, and established cells are all included. Specifically, the somatic cells include, for example, (1) tissue stem cells (somatic stem cells) such as nerve stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells, (2) tissue precursor cells, (3) lymphocytes, and epithelium.
  • tissue stem cells such as nerve stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells
  • tissue precursor cells such as lymphocytes, and epithelium.
  • Endothelial cells muscle cells, fibroblasts (skin cells, etc.), hair cells, hepatocytes, gastric mucosal cells, intestinal cells, splenocytes, pancreatic cells (pancreatic exocrine cells, etc.), brain cells, lung cells, renal cells And differentiated cells such as fat cells are exemplified.
  • the HLA genotypes of the transplanted individuals are the same or substantially the same from the viewpoint that rejection does not occur. It is desirable to use cells.
  • substantially the same HLA type means that the HLA genotypes match to the extent that the transplanted cells can engraft when the cells are transplanted.
  • the main HLA HLA
  • HLA- It is a somatic cell having an HLA type that matches the 3 loci of A, HLA-B and HLA-DR or the 4 loci with HLA-C).
  • iPS cell lines established by NIH, RIKEN, Kyoto University, etc. may be used.
  • ES cells derived from cloned embryos obtained by nuclear transplantation ntES cells are ES cells derived from cloned embryos produced by nuclear transplantation technology and have almost the same characteristics as ES cells derived from fertilized eggs (T. Wakayama et al. (2001), Science, 292: 740-743; S. Wakayama et al. (2005), Biol. Reprod., 72: 932-936; J. Byrne et al. (2007), Nature, 450: 497-502).
  • ES cells established from the inner cell mass of a blastocyst derived from a cloned embryo obtained by replacing the nucleus of an unfertilized egg with the nucleus of a somatic cell are ntES (nuclear transfer ES) cells.
  • ntES nuclear transfer ES
  • a combination of nuclear transplantation technology JB Cibelli et al. (1998), Nature Biotechnol., 16: 642-646) and ES cell production technology (above) is used (Seika Wakayama).
  • somatic cell nuclei can be injected into unenucleated unfertilized eggs of mammals and cultured for several hours to initialize them.
  • Muse cells are pluripotent stem cells produced by the method described in WO2011 / 007900, specifically fibroblasts or bone marrow stromal cells treated with long-term trypsin, preferably 8 hours or 16 hours. Pluripotent cells obtained by suspension culture after treatment and positive for SSEA-3 and CD105.
  • Skeletal muscle stem cells mean cells that have the ability to selectively differentiate into skeletal muscle cells and have both self-renewal ability and proliferative differentiation ability. Skeletal muscle stem cells in vivo are called satellite cells and usually exist between the basement membrane and plasma membrane of muscle fibers. As used herein, skeletal muscle stem cells include satellite cells.
  • Pax7, Hesr1, Hesr3, Myf5, CalcR, NGFR, ERBB3, CD56, CD82, etc. are known as marker genes for identifying skeletal muscle stem cells. It was found that FGFR4 (fibroblast growth factor receptor 4) was expressed in FGFR4 (fibroblast growth factor receptor 4), and it was found that a high-purity skeletal muscle stem cell population could be obtained by using the cell surface marker FGFR4. Therefore, the method for improving the purity of skeletal muscle stem cells of the present invention is characterized by using the cell surface marker FGFR4. FGFR4 is also referred to as CD334.
  • the method for improving the purity of skeletal muscle stem cells of the present invention may be any method including a step of recovering cell surface marker FGFR4-positive cells from a cell population.
  • the cell population is not particularly limited as long as it is a cell population that may contain skeletal muscle stem cells.
  • the cell population used in the method for improving the purity of skeletal muscle lineage cells of the present invention can also be used in the method for improving the purity of skeletal muscle stem cells of the present invention.
  • a cell suspension prepared from a surgical specimen containing skeletal muscle or a cell population in the process of inducing differentiation of pluripotent stem cells into skeletal muscle cells can be used.
  • the cell population may be a cell population with improved purity of skeletal muscle lineage cells obtained by the method for improving purity of skeletal muscle lineage cells of the present invention.
  • the cell population with improved purity of skeletal muscle lineage cells is preferably a cell population with improved purity from a cell population in the process of inducing differentiation from pluripotent stem cells to skeletal muscle cells.
  • a cell population containing high-purity CDH13-positive and FGFR4-positive skeletal muscle stem cells is useful as skeletal muscle stem cells for living body transplantation.
  • the method for recovering FGFR4-positive cells is not particularly limited, and examples thereof include a method using a cell sorter and a method using affinity chromatography.
  • a cell sorter for example, a cell suspension of a cell population is prepared, an anti-FGFR4 antibody is added to the cell suspension, the anti-FGFR4 antibody is bound to the FGFR4-positive cells, and the cells are subjected to the cell sorter to obtain FGFR4-positive cells. Can be sorted and collected.
  • affinity chromatography for example, a cell suspension of a cell population is prepared, and the FGFR4 positive cells are bound to the anti-FGFR4 antibody by passing the cell suspension through a column filled with a carrier to which the anti-FGFR4 antibody is bound.
  • FGFR4-positive cells can be recovered by dissociating the binding with the anti-FGFR4 antibody.
  • the proportion of FGFR4 negative cells in the original cell population decreases and the proportion of FGFR4 positive cells increases. It is a group.
  • the present invention provides a method for producing skeletal muscle lineage cells.
  • the method for producing skeletal muscle lineage cells of the present invention may include the following steps (1) and (2). (1) Step of initiating differentiation induction from pluripotent stem cells to skeletal muscle cells (2) Step of recovering cell surface marker CDH13-positive cells from cell population in the process of differentiation induction
  • the method for producing skeletal muscle lineage cells of the present invention can be carried out in the same manner as in the embodiment of the method for producing skeletal muscle stem cells described below.
  • the time to recover the cell surface marker CDH13-positive cells is not limited to the same time as the method for producing skeletal muscle stem cells described below, and the induction of differentiation from pluripotent stem cells to skeletal muscle cells is not limited to the same time. It may be the time when CDH13-positive cells appear in the developing cell population.
  • the differentiation induction method from pluripotent stem cells to skeletal muscle cells is induced using the differentiation induction method described in "Experimental Materials and Methods" (2) of the Examples of the present application, 2 weeks to 24 from the start of differentiation induction.
  • CDH13-positive cells can be recovered weekly.
  • preliminary studies can be performed as appropriate to determine the recovery time of CDH13-positive cells.
  • the present invention provides a method for producing skeletal muscle stem cells.
  • the method for producing skeletal muscle stem cells of the present invention may include the following steps (1) and (2). (1) Step of initiating differentiation induction from pluripotent stem cells to skeletal muscle cells (2) Step of recovering cell surface marker FGFR4-positive cells, CDH13-positive cells or FGFR4-positive and CDH13-positive cells from a cell population in the process of inducing differentiation
  • Step of initiating differentiation induction from pluripotent stem cells to skeletal muscle cells (2) Step of recovering cell surface marker FGFR4-positive cells, CDH13-positive cells or FGFR4-positive and CDH13-positive cells from a cell population in the process of inducing differentiation
  • FGFR4-positive cells cell surface marker FGFR4-positive cells, CDH13-positive cells or FGFR4-positive and CDH13-positive cells from a cell population in the process of inducing differentiation
  • step (1) the induction of differentiation from pluripotent stem cells to skeletal muscle cells is started.
  • pluripotent stem cells include embryonic stem (ES) cells, embryonic stem (ntES) cells derived from cloned embryos obtained by nuclear transplantation, sperm stem (GS) cells, embryonic reproduction (EG) cells, and induced pluripotent cells.
  • Examples include pluripotent stem (iPS) cells, cultured fibroblasts and pluripotent cells (Muse cells) derived from bone marrow stem cells.
  • the pluripotent stem cells used in the present invention may be iPS cells, ES cells, ntES cells, or iPS cells.
  • the method for inducing differentiation of pluripotent stem cells into skeletal muscle cells is not particularly limited, and can be appropriately selected from known methods for inducing differentiation of pluripotent stem cells into skeletal muscle cells.
  • the method for inducing differentiation of pluripotent stem cells into skeletal muscle cells is a differentiation medium to which various compounds and physiologically active substances are added without introducing an exogenous skeletal muscle inducing gene into the pluripotent stem cells. It may be a method using.
  • the compounds contained in the differentiation medium include ROCK (Rho-associated coiled-coil forming kinase) inhibitor, GSK3 (Glycogen synthase kinase 3) inhibitor, TGF- ⁇ (Transforming Growth Factor- ⁇ ) inhibitor, and BMP (Bone morphogenetic). protein) Inhibitors and the like.
  • the physiologically active substance include IGF-1 (Insulin-like Growth Factor 1), bFGF (basic Fibroblast Growth Factor), HGF (Hepatocyte Growth Factor), and retinoic acid.
  • the method for inducing the differentiation of pluripotent stem cells into skeletal muscle cells is to use a differentiation medium supplemented with the above-mentioned various compounds and physiologically active substances to mature skeletal muscle cells in a manner that mimics the developmental process of the fetus. It may be a method of inducing differentiation of a skeletal muscle lineage cell population including skeletal muscle stem cells.
  • the form that mimics the developmental process of the fetus means a method of inducing differentiation of skeletal muscle lineage cells via paraxial mesoderm cells, somites cells, and cutaneous musculoskeletal cells.
  • Examples of such a differentiation-inducing method include the method described in Non-Patent Document 1 (Zhao et al., Stem Cell Reports, Vol. 15 1-15 July 14, 2020.), And described in International Publication WO2016 / 108288 A1. , And the method described in Hicks et al. (Nat Cell Biol. 2018 Jan; 20 (1): 46-57.).
  • step (2) cell surface marker FGFR4-positive cells, CDH13-positive cells or FGFR4-positive and CDH13-positive cells are collected from the cell population in the process of inducing differentiation.
  • the method for recovering each cell surface marker-positive cell is not particularly limited, and examples thereof include a method using a cell sorter and a method using affinity chromatography.
  • FGFR4 positive cells using a cell sorter prepare a cell suspension of the cell population, add the anti-FGFR4 antibody to the cell suspension, bind the anti-FGFR4 antibody to the FGFR4 positive cells, and supply the cells to the cell sorter.
  • FGFR4 positive cells can be selected and recovered.
  • CDH13-positive cells When collecting CDH13-positive cells using a cell sorter, prepare a cell suspension of the cell population, add the anti-CDH13 antibody to the cell suspension, bind the anti-CDH13 antibody to the CDH13-positive cells, and supply the cells to the cell sorter. CDH13-positive cells can be selected and recovered.
  • CDH13-positive cells When recovering FGFR4-positive and CDH13-positive cells using a cell sorter, prepare a cell suspension of the cell population, add anti-FGFR4 antibody and anti-CDH13 antibody to the cell suspension, and add anti-FGFR4 antibody to FGFR4-positive cells. By binding anti-CDH13 antibodies to CDH13-positive cells and subjecting them to a cell sorter, FGFR4-positive and CDH13-positive cells can be selected and recovered.
  • FGFR4 positive cells When recovering FGFR4 positive cells using affinity chromatography, prepare a cell suspension of the cell population and pass the cell suspension through a column filled with a carrier to which an anti-FGFR4 antibody is bound to turn the FGFR4 positive cells into an anti-FGFR4 antibody. FGFR4-positive cells can be recovered by binding and then dissociating the binding to the anti-FGFR4 antibody.
  • CDH13-positive cells When recovering CDH13-positive cells using affinity chromatography, prepare a cell suspension of the cell population and pass the cell suspension through a column filled with a carrier bound to the anti-CDH13 antibody to turn the CDH13-positive cells into anti-CDH13 antibody. CDH13-positive cells can be recovered by binding and then dissociating the binding to the anti-CDH13 antibody.
  • the CDH13-positive cells are recovered by passing the cell suspension of the cell population through a column filled with a carrier to which the anti-CDH13 antibody is bound by the above procedure, and then anti-CDH13.
  • FGFR4-positive and CDH13-positive cells can be recovered through the recovered CDH13-positive cells in a column filled with a carrier to which the FGFR4 antibody is bound.
  • FGFR4-positive cells are collected through a cell suspension of a cell population in a column previously filled with a carrier bound with an anti-FGFR4 antibody, and then the recovered FGFR4-positive cells are passed through a column filled with a carrier bound with an anti-CDH13 antibody.
  • FGFR4-positive and CDH13-positive cells may be recovered.
  • step (2) the time to recover the cell surface marker FGFR4-positive cells, CDH13-positive cells or FGFR4-positive and CDH13-positive cell skeletal muscle stem cells from the cell population in the process of inducing differentiation from pluripotent stem cells to skeletal muscle cells is determined. It may be after the time when a part of pluripotent stem cells differentiates into skeletal muscle stem cells. For example, when the differentiation induction method from pluripotent stem cells to skeletal muscle cells is induced using the differentiation induction method described in "Experimental Materials and Methods" (2) of the Examples of the present application, 3 weeks to 24 weeks from the start of differentiation induction.
  • Skeletal muscle stem cells can be recovered by recovering FGFR4-positive cells, CDH13-positive cells or FGFR4-positive and CDH13-positive cells per week.
  • the recovery time may be 10 to 16 weeks, 11 to 13 weeks, or 12 weeks from the start of differentiation induction.
  • the inventors of the present application have confirmed that the skeletal muscle stem cells recovered 12 weeks after the start of the induction of differentiation have a high engraftment rate after transplantation.
  • preliminary studies can be appropriately performed to determine the recovery time of skeletal muscle stem cells.
  • the skeletal muscle stem cells produced by the method for producing skeletal muscle stem cells of the present invention can be used for living body transplantation.
  • the present inventors transplanted skeletal muscle stem cells (CDH13-positive cells and FGFR4-positive cells) produced by the method for producing skeletal muscle stem cells of the present invention into a low-temperature injured tibialis anterior muscle of a muscular dystrophy model, and as a result, transplanted them into a living body. It has been confirmed that it efficiently differentiates into skeletal muscle cells and engrafts (see Examples).
  • transplanted cells were positive for PAX7, which is a marker specific to satellite cells, and engrafted in the place where satellite cells exist, inside LAMININ, and it was confirmed that they exist as satellite cells in vivo. ing. This means that the ability to regenerate skeletal muscle is sustained for a long period of time after transplantation, and that the effect of cell therapy may be sustained.
  • PAX7 which is a marker specific to satellite cells
  • the present invention includes a cell surface marker for purifying or detecting skeletal muscle lineage cells consisting of CDH13.
  • the present invention also includes cell surface markers for purification or detection of skeletal muscle stem cells consisting of FGFR4.
  • Pluripotent stem cells Human iPS cell line 201B7 (hereinafter referred to as "201B7") was established by transduction with a retrovirus from commercially available healthy human fibroblasts (Takahashi et al., Cell, 131; 861-872, 2007).
  • the DMD (Duchenne muscular dystrophy) iPS cell line (clone ID: CiRA00111, hereinafter referred to as "DMD”) was established by the episomal vector system from the skin fibroblasts of DMD patients lacking the dystrophin gene exxon 44. (Okita et al., Stem Cells, 31; 458-466, 2012).
  • the DMD repair iPS cell line (hereinafter referred to as "CKI") was established by knocking in exon 44 of the dystrophin gene into DMD (Li et al., Stem Cell Reports, 4; 143-154, 2015).
  • the DMD was established with the approval of the Kyoto University graduate School and the Institutional Review Board of the Faculty of Medicine (approval numbers # R0091 and # G259) and with written consent.
  • the stock iPS cell line Ff-WJ14s01 (hereinafter referred to as "S01”) for regenerative medicine was established from the cord blood of a healthy donor with a homozygous HLA refrequency allele in Japan.
  • CDMi medium is a 1: 1 mixture of IMDM (Iscove's Modified Dulbecco's Medium, Invitrogen, 12440053) containing L-glutamine, 25 mM HEPES and Ham's F-12 Nutrient Mix (Invitrogen, 11765054) at 1% BSA. (Sigma), 1% penicillin streptomycin mixed solution (Nacalai), 1% CD lipid concentrate (Invitrogen), 1% insulin-transferase-selenium (Invitrogen) and 450 ⁇ M 1-thioglycerol (Sigma).
  • BSA bovine serum albumin
  • 2-ME 2-mercaptoethanol
  • 10 ng / ml recombinant human IGF-1 PeproTech
  • 10 ng / ml recombinant human bFGF Oriental yeast
  • 10 ng / ml recombinant human HGF PeproTech
  • medium was 0.5% penicillin streptomycin mixed solution (Nacalai), 2 mM L-glutamine (Nacalai), 0.1 mM 2-ME, 2% horse serum (HS, Sigma), 5 ⁇ M SB431542 and 10 ng / ml. It was changed to DMEM (Invitrogen, 11960069) to which IGF-1 was added. After that, the medium was changed every 2 to 3 days. Cells were cultured in this medium until used in the experiment. Cells on day 84 of differentiation were subjected to analysis.
  • the cells were then shaded and incubated with the corresponding antibody for 20 minutes on ice. After incubation, cells were washed twice again with HBSS buffer (1000 rpm, 4 ° C., centrifuge for 5 minutes). Finally, cells were resuspended in HBSS buffer containing 1% Hoechst and filtered through a 40 nm mesh. Cells were stored on ice until FACS sorting. FACS sorting and analysis of living cells was performed by Aria II (BD Biosciences).
  • the cells were fixed with 2% PFA (paraformaldehyde) for 10 minutes and washed 3 times at 4 ° C for 10 minutes using PBS. The cells were then blocked with Blocking one (Nacalai) for 1 hour. Following blocking, cells were incubated overnight with primary antibody diluted in PBS containing 10% Blocking one. The cells were subsequently washed 3 times with PBS (PBS-T) containing 0.2% Triton X100 (Sigma-Aldrich) and the corresponding secondary antibody and DAPI (1: 5000) and cells were incubated for 1 hour at room temperature. .. Cells stained with antibody were observed under a BZ-X700 microscope (Keyence) and analyzed with BZ-X analyzer software (Keyence). The antibodies used are shown in Table 2.
  • PFA paraformaldehyde
  • Example 1 Examination of specific cell surface markers in Myf5-positive cells obtained by inducing differentiation from iPS cells to skeletal muscle stem cells.
  • Differentiation into skeletal muscle stem cells was induced using 201B7 as human iPS cells, and the cells were collected on day 84 and Myf5 positive cells and Myf5 negative cells were sorted by FACS.
  • RNA-seqing analysis was performed on Myf5-positive cells and Myf5-negative cells, respectively, and CDH13 and FGFR4 were identified as surface markers specifically highly expressed in Myf5-positive cells.
  • FIG. 1 shows the results of analysis of the expression patterns of CDH13 and FGFR4 using the mouse single-cell RNA sequencing database "Tabula Muris". Both CDH13 and FGFR4 were confirmed to be strongly expressed in the Limb Muscle surrounded by the dotted line.
  • CDH13 is weak in the results of analysis of the expression patterns of CDH13 and FGFR4 using a database of single-cell RNA sequencing of differentiated skeletal muscle cells from human fetal muscle tissue and human iPS cells.
  • FGFR4 was strongly expressed from Embryonic 5 weeks to Fetal 6 weeks, and was also strongly expressed in the iPS cell differentiation system.
  • FIG. 2 shows the results of analysis of the expression patterns of CDH13 and FGFR4 using a gene expression database in human fetal-derived tissues.
  • A is the result of CDH13
  • B is the result of FGFR4. Both CDH13 and FGFR4 were found to be highly expressed in the tissues of the skeletal muscle system shown in black.
  • FIG. 3 shows the results of analysis of the expression patterns of CDH13 and FGFR4 using a gene expression database in adult human tissues.
  • A) is the result of CDH13
  • (B) is the result of FGFR4. Both CDH13 and FGFR4 were confirmed to be highly expressed in muscle.
  • Example 2 Expression analysis of CDH13 and FGFR4 in cells induced to differentiate from iPS cells to skeletal muscle stem cells.
  • Differentiation into skeletal muscle stem cells was induced using 201B7 and S01 as human iPS cells, and on day 84, the cells were collected and Myf5-positive cells and Myf5-negative cells were sorted by FACS. Was measured by quantitative real-time PCR.
  • the results are shown in FIG. (A) is the result of CDH13, and (B) is the result of FGFR4.
  • the expression level was expressed as a relative expression level when the expression level of Myf5-negative cells was 1.
  • Both CDH13 and FGFR4 showed specifically high expression in Myf5-positive cells (*; p ⁇ 0.05, **; p ⁇ 0.01, ***; p ⁇ 0.001).
  • FIG. (A) is the result of CDH13
  • (B) is the result of FGFR4. It was shown that CDH13-positive cells and FGFR4-positive cells were present in a certain proportion in the cells induced to differentiate from CKI to skeletal muscle stem cells.
  • Example 3 Expression analysis of skeletal muscle stem cell-related genes in CDH13-positive cells and FGFR4-positive cells obtained by inducing differentiation from iPS cells to skeletal muscle stem cells.
  • Differentiation into skeletal muscle stem cells was induced using 201B7, CKI and DMD as human iPS cells, the cells were recovered on day 84, and CDH13-positive cells and FGFR4-positive cells were sorted by FACS.
  • the expression levels of skeletal muscle stem cell-related genes MYF5 and PAX7 and myoblast-related gene MYOD1 were measured by quantitative real-time PCR in CDH13-positive cells, FGFR4-positive cells and non-sorting cells (controls).
  • Fig. 7 shows the result of 201B7
  • Fig. 8 shows the result of CKI
  • Fig. 9 shows the result of DMD.
  • the expression level was expressed as the relative expression level when the expression level of the non-sorting cells (control) was 1.
  • FIGS. 7, 8 and 9 FGFR4-positive cells highly expressed the skeletal muscle stem cell-related genes MYF5 and PAX7.
  • CDH13-positive cells also had higher expression of MYF5 and PAX7 compared to controls, but lower expression levels than FGFR4-positive cells.
  • CDH13-positive cells highly expressed the myoblast-related gene MYOD1 (*; p ⁇ 0.05, **; p ⁇ 0.01, ***; p ⁇ 0.001, ****; p ⁇ 0.0001. ).
  • Example 4 Examination of in vitro differentiation potential of CDH13-positive cells and FGFR4-positive cells into skeletal muscle cells obtained by inducing differentiation from iPS cells to skeletal muscle stem cells]
  • (1) In vitro differentiation ability of CDH13-positive cells into skeletal muscle cells Induction of differentiation into skeletal muscle stem cells using 201B7, CKI and DMD as human iPS cells was performed, and on the 84th day, the cells were recovered and used with anti-CDH13 antibody. Staining was performed and CDH13-positive cells and CDH13-negative cells were sorted by FACS, respectively.
  • CDH13-positive cells and CDH13-negative cells were further induced to differentiate into skeletal muscle cells, and the ability to differentiate into skeletal muscle cells was examined using the expression of myosin heavy chain (hereinafter referred to as "MHC") as an index.
  • MHC myosin heavy chain
  • FIG. (A) is the result of immunostaining MHC
  • (B) is the result of calculating the differentiation index (MHC positive cell rate) by image analysis.
  • differentiation index MHC positive cell rate
  • FIG. (A) is the result of immunostaining MHC
  • (B) is the result of calculating the differentiation index (MHC positive cell rate) by image analysis. Differentiation into MHC-positive skeletal muscle cells was observed with a high efficiency of 80% or more in FGFR4-positive cells, but differentiation of MHC-positive skeletal muscle cells was also observed from FGFR4-negative cells. (**; p ⁇ 0.01, ***; p ⁇ 0.001).
  • Example 5 Transplantation experiment of CDH13-positive cells and FGFR4-positive cells obtained by inducing differentiation from iPS cells to skeletal muscle stem cells into mice] (1) Confirmation of engraftment of transplanted cells CKI and DMD were used as human iPS cells to induce differentiation into skeletal muscle stem cells, cells were collected on day 84, and CDH13-positive cells and CDH13-negative cells were used using FACS. , FGFR4 positive cells and FGFR4 negative cells were sorted respectively.
  • Each sorted cell was transplanted into NOG-mdx mice by the method described in (4) of the above [Experimental Materials and Methods], and after 4 weeks, the transplanted site was collected for histochemical analysis and measurement of the number of engrafted cells. Was done.
  • the transplanted tissue 4 weeks after transplantation was stained with anti-PAX7 antibody, anti-LAMININ ⁇ 2 antibody, anti-h-LAMIN A / C antibody and DAPI, and co-focused. Observed with a microscope. The results are shown in FIG. Human nucleus-positive and PAX7-positive cells located inside laminin were observed from both CDH13-positive cells and FGFR4-positive cells (arrowhead), confirming that the transplanted cells differentiated into satellite cells in the post-transplant muscle tissue. rice field.
  • Example 6 Comparison with known skeletal muscle lineage cell surface markers
  • FACS analysis Inducing differentiation into skeletal muscle stem cells using DMD and CKI as human iPS cells, and collecting the cells on the 84th day, CD56-positive cell rate, CD82-positive cell rate, and NGFR-positive cell by FACS. The rate, ERBB3-positive cell rate, CDH13-positive cell rate and FGFR4-positive cell rate were confirmed.
  • the results are shown in FIG. (A) is the result of DMD, and (B) is the result of CKI. It was shown that the proportion of CD56-positive cells and NGFR-positive cells was higher than that of other marker-positive cells.
  • FIG. 17 The results are shown in Fig. 17.
  • (A) is the result of DMD
  • (B) is the result of CKI.
  • the expression level was expressed as the relative expression level when the expression level of unsorted cells (not shown) was 1.
  • Expression of MYF5 is high in FGFR4-positive cells.
  • the expression of PAX7 is high in FGFR4-positive cells, and in CKI-derived cells, CDH13-positive cells also have high PAX7 expression.
  • the expression of MYOD1 is high in ERBB3-positive cells and CDH13-positive cells in CKI, suggesting that the fraction contains a large amount of myoblasts.
  • the expression of MYOD1 is low in FGFR4-positive cells, suggesting that the fraction is closer to that of skeletal muscle stem cells (*; p ⁇ 0.05, **; p ⁇ 0.01, ***; p ⁇ 0.001).

Abstract

La présente invention concerne les éléments suivants : un procédé destiné à améliorer la pureté des cellules de lignée de muscle squelettique et caractérisé par la récupération des cellules positives pour un marqueur de surface cellulaire CDH13 dans une population cellulaire ; un procédé destiné à produire des cellules de lignée de muscle squelettique et comprenant une étape consistant à induire la différenciation des cellules souches pluripotentes en cellules de muscle squelettique, et une étape consistant à récupérer les cellules positives pour le marqueur de surface cellulaire CDH13 à partir d'une population cellulaire dans le processus d'induction de la différenciation ; un procédé destiné à améliorer la pureté des cellules de la lignée des muscles squelettiques et caractérisé par la récupération des cellules positives pour un marqueur de surface cellulaire FGFR4 dans une population cellulaire ; et un procédé de production de cellules souches de muscle squelettique comprenant une étape consistant à induire la différenciation de cellules souches pluripotentes en cellules de muscle squelettique, et une étape consistant à récupérer, à partir d'une population de cellules dans le processus d'induction de la différenciation, des cellules positives pour le marqueur de surface cellulaire FGFR4, des cellules CDH13-positives, ou des cellules FGFR4-positives et CDH13-positives.
PCT/JP2021/041709 2020-11-16 2021-11-12 Marqueur de surface cellulaire pour une purification à haut rendement de cellules de lignée de muscle squelettique et de cellules souches de muscle squelettique, et son utilisation WO2022102742A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2022562200A JPWO2022102742A1 (fr) 2020-11-16 2021-11-12

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2020190063 2020-11-16
JP2020-190063 2020-11-16

Publications (1)

Publication Number Publication Date
WO2022102742A1 true WO2022102742A1 (fr) 2022-05-19

Family

ID=81602366

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2021/041709 WO2022102742A1 (fr) 2020-11-16 2021-11-12 Marqueur de surface cellulaire pour une purification à haut rendement de cellules de lignée de muscle squelettique et de cellules souches de muscle squelettique, et son utilisation

Country Status (2)

Country Link
JP (1) JPWO2022102742A1 (fr)
WO (1) WO2022102742A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008506366A (ja) * 2004-05-14 2008-03-06 レセプター バイオロジックス インコーポレイテッド 細胞表面受容体アイソフォームならびにその同定および使用方法
JP2013527746A (ja) * 2010-04-22 2013-07-04 国立大学法人京都大学 多能性幹細胞から骨格筋または骨格筋前駆細胞への分化誘導法
WO2014038890A1 (fr) * 2012-09-07 2014-03-13 주식회사 마크로젠 Protéine de fusion comprenant axl et composition pour le traitement du cancer la comprenant
CN104745700A (zh) * 2015-03-27 2015-07-01 南京医科大学 一种食管癌相关甲基化生物标志物及应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008506366A (ja) * 2004-05-14 2008-03-06 レセプター バイオロジックス インコーポレイテッド 細胞表面受容体アイソフォームならびにその同定および使用方法
JP2013527746A (ja) * 2010-04-22 2013-07-04 国立大学法人京都大学 多能性幹細胞から骨格筋または骨格筋前駆細胞への分化誘導法
WO2014038890A1 (fr) * 2012-09-07 2014-03-13 주식회사 마크로젠 Protéine de fusion comprenant axl et composition pour le traitement du cancer la comprenant
CN104745700A (zh) * 2015-03-27 2015-07-01 南京医科大学 一种食管癌相关甲基化生物标志物及应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NALBANDIAN MINAS, ZHAO MINGMING, SASAKI-HONDA MITSURU, JONOUCHI TATSUYA, LUCENA-CACACE ANTONIO, MIZUSAWA TAKUMA, YASUDA MASAHIKO, : "Characterization of hiPSC-Derived Muscle Progenitors Reveals Distinctive Markers for Myogenic Cell Purification Toward Cell Therapy", STEM CELL REPORTS, CELL PRESS, UNITED STATES, vol. 16, no. 4, 1 April 2021 (2021-04-01), United States , pages 883 - 898, XP055930092, ISSN: 2213-6711, DOI: 10.1016/j.stemcr.2021.03.004 *

Also Published As

Publication number Publication date
JPWO2022102742A1 (fr) 2022-05-19

Similar Documents

Publication Publication Date Title
JP7166631B2 (ja) 新規軟骨細胞誘導方法
US9121011B2 (en) Method for inducing differentiation of human pluripotent stem cell into intermediate mesoderm cell
US8883498B2 (en) Method for inducing differentiation of pluripotent stem cells into skeletal muscle or skeletal muscle progenitor cells
JP6429280B2 (ja) 効率的な心筋細胞の誘導方法
JP6612736B2 (ja) 心筋細胞の選別方法
JP6883791B2 (ja) 骨格筋前駆細胞の選択方法
JP6373253B2 (ja) 新規心筋細胞マーカー
WO2016108288A1 (fr) Procédé de production de cellules progénitrices de muscle squelettique
JP5995247B2 (ja) 多能性幹細胞から樹状細胞を製造する方法
JP6025067B2 (ja) 新規心筋細胞マーカー
JP6708559B2 (ja) 多能性細胞識別化合物
JP6780197B2 (ja) 新規成熟心筋細胞マーカー
JP5842289B2 (ja) 効率的な内皮細胞の誘導方法
WO2022102742A1 (fr) Marqueur de surface cellulaire pour une purification à haut rendement de cellules de lignée de muscle squelettique et de cellules souches de muscle squelettique, et son utilisation
WO2021015086A1 (fr) Procédé de fabrication de cellules de myotube mâtures à partir de cellules souches du muscle squelettique
EP4180516A1 (fr) Cellules précurseurs de muscle squelettique et procédé de purification de celles-ci, composition pour le traitement de maladies myogènes et procédé de production d'un groupe de cellules contenant des cellules précurseurs de muscle squelettique
WO2022138593A1 (fr) Procédé de production d'une cellule de muscle squelettique

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: 21891987

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022562200

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: 21891987

Country of ref document: EP

Kind code of ref document: A1