WO2023063187A1 - Procédé de production d'organoïde - Google Patents

Procédé de production d'organoïde Download PDF

Info

Publication number
WO2023063187A1
WO2023063187A1 PCT/JP2022/037274 JP2022037274W WO2023063187A1 WO 2023063187 A1 WO2023063187 A1 WO 2023063187A1 JP 2022037274 W JP2022037274 W JP 2022037274W WO 2023063187 A1 WO2023063187 A1 WO 2023063187A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
muscle
culture
organoid
medium
Prior art date
Application number
PCT/JP2022/037274
Other languages
English (en)
Japanese (ja)
Inventor
一憲 清水
裕之 本多
一貴 山本
Original Assignee
国立大学法人東海国立大学機構
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 国立大学法人東海国立大学機構 filed Critical 国立大学法人東海国立大学機構
Publication of WO2023063187A1 publication Critical patent/WO2023063187A1/fr

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M3/00Tissue, human, animal or plant cell, or virus culture apparatus
    • 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
    • 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

Definitions

  • the present invention relates to an organoid production method and the like.
  • NMJ neuromuscular junction
  • ALS amyotrophic lateral sclerosis
  • SBMA spinal and bulbar muscular atrophy
  • Patent Document 1 An evaluation model for in vitro research on the onset mechanism of neuromuscular diseases and screening of therapeutic agents is known. For example, a technique is known in which iPS cells are induced to differentiate into muscle cells, nerve cells, Schwann cells, and the like to form neuromuscular junctions (see Patent Document 1). However, what is obtained by the method described in Patent Document 1 is planar tissue, which is different from neuromuscular tissue (three-dimensional) in a living body.
  • Patent Document 2 describes a technique in which muscle cells and nerve cells are cultured in separate compartments, and the nerve tissue is connected to the three-dimensional muscle tissue through holes that connect the two compartments.
  • this technique requires two types of cells to be prepared and cultured separately.
  • An object of the present invention is to provide a technique for producing neuromuscular organoids more simply and efficiently.
  • the present inventors have found that (1a) MyoD is transiently applied to pluripotent stem cells in a culture device having at least two pillars extending from the culture bottom to the culture tank side. and an organoid-forming composition containing at least one type of cell selected from the group consisting of pluripotent stem cells and early-induced cells expressed in the pillars to induce muscle differentiation while covering the periphery of the pillars, and (2) culturing the muscle organoids obtained in step (1a) in a medium containing neurotrophic factors to induce neuronal differentiation to obtain neuro-muscle organoids.
  • the above problems can be solved by a method for producing organoids. Based on this finding, the inventors have further studied and completed the present invention. That is, the present invention includes the following aspects.
  • Section 1 A group consisting of pluripotent stem cells and initial induced cells obtained by transiently expressing MyoD in pluripotent stem cells in a culture device having at least two pillars extending from the bottom of the culture to the culture tank side.
  • the organoid-forming composition containing at least one cell selected from the above is covered with the pillar to induce muscle differentiation to obtain a muscle organoid, and (2) obtained in step (1a) culturing the obtained muscle organoids in a medium containing neurotrophic factors to induce neuronal differentiation to obtain neuro-muscle organoids; Methods for producing neuro-muscular organoids.
  • Section 3 The production method according to item 1 or 2, wherein the muscle differentiation induction includes transient expression of MyoD in the cells by expression induction using a drug.
  • Item 5 wherein the concentration of doxycycline in the medium is 0.05 ⁇ g/ml or more.
  • Item 6. The production method according to any one of Items 1 to 5, wherein the cell concentration in the organoid-forming composition is 5 ⁇ 10 6 cells/ml or more at the start of muscle differentiation induction in step (1a).
  • Item 7 The production method according to any one of Items 1 to 6, wherein the culture is performed in a medium containing a lock inhibitor during part or all of the muscle differentiation induction culture period.
  • Item 8 The production method according to any one of items 1 to 7, wherein the amount of medium in step (1a) is 5 to 50 ⁇ l per 1 ⁇ 10 4 cells at the start of muscle differentiation induction in step (1a).
  • Item 9 Items 1 to 8, wherein the organoid-forming composition contains a cell scaffolding material.
  • Item 10 Items 1 to 8, wherein the cell scaffolding material contains at least one selected from the group consisting of fibrin, matrigel, and collagen.
  • Item 11 The production method according to any one of Items 1 to 10, wherein the neurotrophic factor includes at least one selected from the group consisting of glial cell line-derived Neurotrophic Factor (GDNF) and brain-derived Neurotrophic Factor (BDNF). .
  • GDNF glial cell line-derived Neurotrophic Factor
  • BDNF brain-derived Neurotrophic Factor
  • Item 12. The production method according to any one of Items 1 to 11, further comprising (3) moving the organoid-forming composition and/or its differentiation inducer to the upper part of the pillar by centrifugation.
  • Item 13 Item 13.
  • (1b) Initial induced cells, pluripotent stem cells, and myoblasts obtained by transiently expressing MyoD in pluripotent stem cells in a culture device having at least two pillars extending from the bottom of the culture to the culture tank side a step of inducing muscle differentiation with an organoid-forming composition containing at least one cell selected from the group consisting of cells covering the pillars, Furthermore, (3) moving the organoid-forming composition and/or its differentiation inducer to the top of the pillar by centrifugation, Methods of producing organoids.
  • Item 14 Initial induced cells, pluripotent stem cells, and myoblasts obtained by transiently expressing MyoD in pluripotent stem cells in a culture device having at least two pillars extending from the bottom of the culture to the culture tank side a step of inducing muscle differentiation with an organoid-forming composition containing at least one cell selected from the group consisting of cells covering the pillars, The cells are at least one type selected from the group consisting of initial induction cells obtained by transiently expressing MyoD in pluripotent stem cells, and pluripotent stem cells, and one of the culture periods for the muscle differentiation induction.
  • Item 15. The production method according to item 14, wherein the amount of medium in step (1b) is 5 to 50 ⁇ l per 1 ⁇ 10 4 cells at the start of muscle differentiation induction in step (1b).
  • Item 16 An organoid obtained by the production method according to any one of Items 1 to 15.
  • a kit for producing nerve-muscle organoids comprising a culture device with at least two pillars extending from the bottom of the culture to the side of the culture tank, and wherein the culture device has one culture tank.
  • a nerve-muscle organoid muscle contraction test kit comprising a culture device having at least two pillars extending from the culture bottom toward the culture tank and nerve-muscle organoids formed around the pillars.
  • Item 19 (Aa) performing some or all of the steps of the production method according to any one of items 1 to 15 in the presence of a test substance to obtain an organoid, or (Ab) the nerve-muscle organoid according to item 16; contacting with a test substance; and (B) assessing the function and/or condition of the organoids;
  • a method of screening for therapeutic agents for neuromuscular diseases comprising:
  • FIG. 1 shows a three-dimensional view of an example of a culture device of the present invention
  • FIG. 1 shows a plan view of an example of a culture device of the present invention
  • FIG. 1 shows a cross-sectional view of an example of a culture device of the present invention
  • FIG. This figure is a cross-sectional view taken along line AA in FIG. 1 shows a three-dimensional view of an example of a culture device of the present invention
  • FIG. Figure 2 shows the size of the device of the invention obtained in the example. All numerical values are in mm.
  • FIG. 4 shows a schematic diagram of a well of a culture device-mounted microwell plate. An outline of the nerve-muscle organoid production process of Test Example 1 is shown.
  • FIG. 1 shows a schematic diagram of a neuromuscular organoid obtained in Test Example 1.
  • FIG. The ribbon-shaped tissue above the pillars is the neuromuscular organoid.
  • 2 shows the measurement results of Test Example 2-1.
  • the vertical axis indicates the amount of displacement of the upper part of the pillar, and the horizontal axis indicates the elapsed time.
  • 2 shows the measurement results of Test Example 2-2.
  • the vertical axis indicates the relative value of the MNX1 gene expression level.
  • the horizontal axis indicates the number of days elapsed since the organization (FIG. 7: D0).
  • FIG. 2 shows an immunostained image obtained in Test Example 2-3.
  • FIG. 4 shows the measurement results of Test Example 2-4.
  • the vertical axis indicates the amount of displacement of the upper part of the pillar, and the horizontal axis indicates the elapsed time from the addition of the chemical. Curare indicates vecuronium bromide and TTX indicates tetrodotoxin.
  • the measurement results of Test Example 3 are shown.
  • the vertical axis indicates the contractile force of the tissue, and the horizontal axis indicates the concentration of Doxycycline added to the muscle induction medium.
  • the measurement results of Test Example 4 are shown.
  • the vertical axis indicates the contractile force of the tissue, and the horizontal axis indicates the presence or absence of the ROCK inhibitor (yes: Y+, no: Y-). 4 shows the measurement results of Test Example 5.
  • the vertical axis indicates the contractile force of the tissue
  • the horizontal axis indicates the cell concentration in the fibrin gel at the start of muscle differentiation induction. 4 shows the measurement results of Test Example 5.
  • FIG. The vertical axis indicates the contractile force of the tissue, and the horizontal axis indicates the number of days elapsed since the organization (FIG. 7: D0).
  • "96MNM” indicates the results of culture in a 96-well plate (200 ⁇ l of medium)
  • “48MNM-S” indicates the results of culture in a 48-well plate (700 ⁇ l of medium).
  • the measurement results of Test Example 6 are shown.
  • the vertical axis indicates the contractile force of the tissue
  • the horizontal axis indicates the number of days elapsed since the organization (FIG. 7: D0).
  • "96MNM” indicates the results of culturing in a medium containing neurotrophic factors in a 96-well plate (200 ⁇ l of medium)
  • "96HS” indicates a muscle differentiation medium (2% horse serum, 1% ITS supplement I3146 , and DMEM containing 100 units penicillin).
  • 2 shows the measurement results of contractile force in Test Example 7.
  • FIG. 2 shows the coefficient of variation of contractile force measured in Test Example 7.
  • FIG. The horizontal axis indicates the number of days elapsed after the medium was replaced with the muscle differentiation medium.
  • the organoid-forming composition containing at least one cell selected from the above is covered with the pillar to induce muscle differentiation to obtain a muscle organoid, and (2) obtained in step (1a) culturing the obtained muscle organoids in a medium containing neurotrophic factors to induce neuronal differentiation to obtain neuro-muscle organoids; and methods for producing neuro-muscular organoids.
  • (1b) Initial induced cells, pluripotent stem cells, and myoblasts obtained by transiently expressing MyoD in pluripotent stem cells in a culture device having at least two pillars extending from the bottom of the culture to the culture tank side A step of inducing muscle differentiation with an organoid-forming composition containing at least one cell selected from the group consisting of cells covering the pillars, and a method for producing organoids.
  • Process (1a) and process (1b) are collectively referred to as “process (1)” in some cases.
  • the above manufacturing methods may be collectively referred to as “the manufacturing method of the present invention”.
  • the culture device will be explained with reference to Figures 1 to 4.
  • the culture device comprises a culture bottom 11 and at least two pillars.
  • culture device 1 includes two pillars, pillar 12a and pillar 12b.
  • the pillars 12a and 12b extend from the culture bottom surface 11 toward the culture tank 14 side.
  • the pillars 12a, 12b are preferably substantially perpendicular to the bottom surface 11 of culture.
  • the smallest acute angle formed by the pillars 12a, 12b and the culture bottom surface 11 is, for example, 75-90°, preferably 80-90°, more preferably 85-90°.
  • the shape of the pillars 12a and 12b is not particularly limited, and may be, for example, a cylindrical shape, a prismatic shape, a conical shape, a pyramidal shape, or a combination thereof.
  • the maximum cross-sectional diameter of the pillars 12a and 12b is not particularly limited, but is, for example, 0.1-2 mm, preferably 0.2-1 mm, and more preferably 0.3-0.7 mm.
  • the height of the pillars 12a and 12b is not particularly limited, it is, for example, 0.5-15 mm, preferably 1-10 mm, and more preferably 2-6 mm.
  • the distance between the fulcrum 16a of the pillar 12a and the fulcrum 16b of the pillar 12b is not particularly limited as long as a muscle organoid can be formed between the pillars, and is, for example, 0.5-5 mm, preferably 1-4 mm, more preferably 2-3 mm.
  • the pillars 12a, 12b are preferably provided with barrier structures 13a, 13b at their upper ends.
  • the barrier structures 13a, 13b can more reliably prevent the organoids from slipping out of the upper ends of the pillars 12a, 12b.
  • the pillars 12a, 12b, if not provided with barrier structures 13a, 13b, are preferably joined at the top to form a pillar link 12 as shown in FIG.
  • the shape of the barrier structures 13a and 13b is not particularly limited.
  • the barrier structures 13a, 13b can be plate-shaped, for example.
  • the barrier structures 13a, 13b preferably have a side or diameter that is longer than the maximum cross-sectional diameter of the pillars 12a, 12b.
  • Culture device 1 preferably comprises a wall 15 .
  • the wall 15 allows the shape and area of the culture bottom surface 11 to be adjusted.
  • the shape of the culture bottom surface 11 is not particularly limited, but in a preferred embodiment, it can be dumbbell-shaped.
  • the area of the culture bottom surface 11 is not particularly limited, but is, for example, 1-30 mm 2 , preferably 2-20 mm 2 , more preferably 3-10 mm 2 .
  • the culture device 1 may include only the culture tank 14 as a culture tank, or may include culture tanks other than the culture tank 14. According to the present invention, the culture device 1 preferably has only one fermenter, as neuro-muscular organoids can be formed in only one fermenter.
  • the culture device may be formed by cutting, formed by a 3D printer, or the like.
  • a template may be produced first using a photolithographic technique and then transferred.
  • a plurality of templates may be used as necessary.
  • the material of the culture device there are no particular restrictions on the material of the culture device as long as it can be cut or the template can be transferred.
  • the material of the culture device for example, polyethylene, polypropylene, polyvinyl chloride, polyvinylidene chloride, polystyrene, polyvinyl acetate, polytetrafluoroethylene, ABS (acrylonitrile butadiene styrene) resin, AS (acrylonitrile styrene) resin, fluorine resin such as Teflon (registered trademark), Thermoplastic resins such as acrylic resins (PMMA); Thermosetting resins such as phenolic resins, epoxy resins, melamine resins, urea resins, unsaturated polyester resins, alkyd resins, polyurethanes, thermosetting polyimides, and silicone rubbers.
  • a resin having optical transparency is desirable.
  • the resin in that case include cycloolefin polymer (COP), polydimethylsiloxane (PDMS), polymethylmethacrylate (PMMA), polycarbonate (PC), plastics such as rigid polyethylene, and silicon.
  • COP cycloolefin polymer
  • PDMS polydimethylsiloxane
  • PMMA polymethylmethacrylate
  • PC polycarbonate
  • plastics such as rigid polyethylene, and silicon.
  • metal may be used instead of resin.
  • the culture device is usually used by installing it on the culture dish or the bottom surface of the well of the microwell plate (preferably the bottom surface of the well) as shown in FIG.
  • Pluripotent stem cells are stem cells that have pluripotency capable of differentiating into many cells existing in the body and also have proliferative ability, and are induced to intermediate mesoderm cells used in the present invention. Any cell that is Examples of pluripotent stem cells include, but are not limited to, embryonic stem (ES) cells, cloned embryo-derived embryonic stem (ntES) cells obtained by nuclear transfer, spermatogonial stem cells (“GS cells”), embryonic These include germ cells (“EG cells”), induced pluripotent stem (iPS) cells, cultured fibroblasts and pluripotent cells derived from bone marrow stem cells (Muse cells).
  • ES embryonic stem
  • ntES cloned embryo-derived embryonic stem
  • GS cells spermatogonial stem cells
  • EG cells germ cells
  • iPS induced pluripotent stem
  • Muse cells pluripotent stem cells derived from bone marrow stem cells
  • a method for producing iPS cells is known in the art, and can be produced by introducing a reprogramming factor into any somatic cell.
  • the reprogramming factors are, 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 or Glis1 genes or gene products are exemplified, these reprogramming factors may be used alone or in combination Also good.
  • Combinations of initialization factors include WO2007/069666, WO2008/118820, WO2009/007852, WO2009/032194, WO2009/058413, WO2009/057831, WO2009/075119, WO2009/079007, WO2009/091659, WO2009/091659, and WO2009/091659.
  • 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. , passaged cells, and cell lines are all included.
  • somatic cells include, for example, (1) tissue stem cells (somatic stem cells) such as neural stem cells, hematopoietic stem cells, mesenchymal stem cells, and dental pulp stem cells, (2) tissue progenitor cells, and (3) blood cells (peripheral stem cells).
  • lymphocytes epithelial cells, endothelial cells, muscle cells, fibroblasts (skin cells, etc.), hair cells, hepatocytes, gastric mucosa cells, enterocytes, splenocytes, pancreatic cells (pancreatic exocrine cells) etc.), differentiated cells such as brain cells, lung cells, kidney cells and adipocytes.
  • Early-stage induced cells are cells at the early stage of muscle differentiation obtained by transiently expressing MyoD in pluripotent stem cells, and are not particularly limited in this respect. Early induced cells are cells at a stage of differentiation prior to differentiation into myoblasts.
  • MyoD used in the present invention includes human myogenic differentiation 1 (MYOD1) consisting of the amino acid sequence represented by SEQ ID NO: 2, its orthologs in other mammals, and transcriptional and splicing variants thereof. . Alternatively, it has 90% or more, preferably 95% or more, more preferably 97% or more amino acid identity with any of the above proteins, and has a function equivalent to the protein (e.g., activation of transcription of a muscle-specific promoter) etc.).
  • MYOD1 human myogenic differentiation 1
  • Nucleic acids encoding MyoD include human myogenic differentiation 1 (MYOD1) cDNA consisting of the nucleotide sequence represented by nucleotide numbers 213 to 1175 in SEQ ID NO: 1, its orthologues in other mammals, transcriptional variants thereof, splicing Examples include mutants. Alternatively, it has 90% or more, preferably 95% or more, more preferably 97% or more nucleotide identity with any of the above nucleic acids, and has a function equivalent to the protein encoded by the nucleic acid (e.g., muscle-specific It may be a nucleic acid encoding a protein having a transcriptional activation of a promoter, etc.).
  • MYOD1 human myogenic differentiation 1
  • stringent conditions are binding complexes or probes as taught by Berger and Kimmel (1987, Guide to Molecular Cloning Techniques Methods in Enzymology, Vol. 152, Academic Press, San Diego CA). It can be determined based on the melting temperature (Tm) of the nucleic acid. For example, washing conditions after hybridization include conditions of "0.1 ⁇ SSC, 0.1% SDS, 60° C.”, and it is preferable that the hybridized state is maintained even after washing under such conditions. .
  • the nucleic acid encoding MyoD may be DNA, RNA, or a DNA/RNA chimera. Also, the nucleic acid may be single-stranded, double-stranded DNA, double-stranded RNA, or a DNA:RNA hybrid. Double-stranded DNA or single-stranded RNA is preferred.
  • the RNA may be RNA incorporated with 5-methylcytidine and pseudouridine (TriLink Biotechnologies) to suppress degradation, or modified RNA by phosphatase treatment.
  • the method for transiently expressing MyoD in pluripotent stem cells is not particularly limited, for example, the following method can be used.
  • expression means that, in the case of a nucleic acid encoding MyoD, the MyoD protein is generated by transcription and translation from the nucleic acid in the cell, and in the case of the MyoD protein, It is synonymous with introducing the protein into the cell.
  • vectors such as viruses, plasmids, and artificial chromosomes can be introduced into pluripotent stem cells by methods such as lipofection, liposomes, and microinjection.
  • viral vectors include retroviral vectors, lentiviral vectors, adenoviral vectors, adeno-associated viral vectors, Sendai viral vectors and the like.
  • artificial chromosome vectors include human artificial chromosomes (HAC), yeast artificial chromosomes (YAC), bacterial artificial chromosomes (BAC, PAC), and the like.
  • plasmids include plasmids for mammalian cells.
  • 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 DNA encoding MyoD can be expressed.
  • regulatory sequences such as a promoter, an enhancer, a ribosome binding sequence, a terminator, and a polyadenylation site so that the DNA encoding MyoD can be expressed.
  • selectable marker sequences such as thymidine kinase gene, diphtheria toxin gene, etc.
  • reporter gene sequences such as fluorescent protein, ⁇ -glucuronidase (GUS), FLAG, etc.
  • Promoters include 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 A promoter and a TRE promoter (a CMV minimal promoter with a Tet response element consisting of 7 consecutive tetO sequences) are exemplified.
  • the MyoD gene may be introduced into pluripotent stem cells using a transient expression vector. It is preferred to use Examples of inducible expression systems include drug-induced expression systems using tetracycline and its derivatives (e.g., doxycycline). It is preferable to use pluripotent stem cells into which a gene construct capable of drug-induced expression of MyoD has been previously introduced. .
  • a fusion protein with tetR and VP16AD or a fusion protein with reverse tetR (rtetR) and VP16AD (rtTA) in the same cell it is desirable to simultaneously express a fusion protein with tetR and VP16AD or a fusion protein with reverse tetR (rtetR) and VP16AD (rtTA) in the same cell.
  • rtetR reverse tetR
  • rtTA reverse tetR
  • MyoD can be expressed transiently during the addition of
  • the above vector incorporates an expression cassette consisting of a DNA encoding a promoter and MyoD linked thereto into the chromosome of the pluripotent stem cell, and, if necessary, excises it.
  • an expression cassette consisting of a DNA encoding a promoter and MyoD linked thereto into the chromosome of the pluripotent stem cell, and, if necessary, excises it.
  • transposon sequences include, but are not limited to, piggyBac.
  • the expression cassette may be preceded and followed by LoxP sequences for the purpose of removing the expression cassette.
  • MyoD when it is in the form of RNA, it may be introduced into pluripotent stem cells by techniques such as electroporation, lipofection, and microinjection.
  • MyoD when MyoD is in protein form, it may be introduced into pluripotent stem cells by techniques such as lipofection, fusion with cell membrane penetrating peptides (eg, TAT and polyarginine from HIV), microinjection, and the like.
  • MyoD Early-induced cells directly control the expression of MyoD itself (for example, introducing a nucleic acid encoding MyoD, expressing it using an agent that controls the promoter activity of an artificially introduced MyoD expression cassette, etc.) , Indirectly controlling the expression of MyoD, such as controlling the expression of transcription factors (e.g., Pax7, etc.) upstream of MyoD, and finally expressing MyoD by controlling the concentration of compounds such as cytokines can be obtained by
  • the period during which MyoD is transiently expressed in pluripotent stem cells may be a period during which cells in the early stage of muscle differentiation can be obtained, and can be appropriately changed depending on the type and properties of the pluripotent stem cells used. 12-48 hours, preferably 18-30 hours. For example, when using the drug-inducible expression system described above, it is preferable to add the drug and culture for this period.
  • expression is stopped by introducing a transposase into the cell after the above-mentioned period of time has passed, and when a vector having a LoxP sequence is used, a desired period of time has passed. Later, a method of terminating expression by introducing Cre into cells is exemplified.
  • introduction may be performed multiple times so that MyoD is present in the cell during the above period.
  • Lock inhibitors are not particularly limited and include, for example, Y-27632, CloneR TM , RevitaCell, SMC4, CEPT and the like.
  • the culture conditions in the preparation of initial induced cells are preferably adherent culture conditions.
  • cell adhesion molecules such as extracellular matrices, specifically matrigel (BD), type I collagen, type IV collagen, gelatin, laminin, heparan sulfate proteoglycans, or entactin, and combinations thereof. It is preferable to culture in a culture dish with the addition of serum or a serum substitute, using a medium used for culturing animal cells as a basal medium.
  • GMEM Gasgow Minimum Essential Medium
  • IMDM Iscove's Modified Dulbecco's Medium
  • 199 medium Eagle's Minimum Essential Medium (EMEM)
  • ⁇ MEM medium Dulbecco's modified Eagle's Medium (DMEM)
  • Ham's F12 medium RPMI 1640 medium
  • Fischer's medium and mixed media thereof.
  • serum substitutes albumin, transferrin, fatty acids, insulin, collagen precursors, trace elements
  • KSR Knockout Serum Replacement
  • the culture temperature is not particularly limited, but is about 30 to 40°C, preferably about 37°C, culture is performed in an atmosphere of air containing CO 2 , and the CO 2 concentration is preferably about 2 to 5%. .
  • Myoblasts are preferably skeletal myoblasts and are characterized by the expression of marker genes such as CD56, MyoD or myogenin.
  • Myoblasts may be, for example, isolated cells or established cell lines obtained by subculturing them.
  • Myoblasts are preferably of mammalian origin, more preferably of human origin.
  • the organoid-forming composition contains at least one type of cell selected from the group consisting of early induced cells, pluripotent stem cells, and myoblasts.
  • the organoid-forming composition preferably comprises early-derived cells and may also comprise pluripotent stem cells.
  • the organoid-forming composition preferably contains a cell scaffolding material.
  • the cell scaffold material is not particularly limited as long as it is a material that serves as a scaffold for cells.
  • cell scaffold materials include fibrin, collagen, matrigel, gelatin, elastin, keratin, laminin, entactin, fibronectin, heparan sulfate proteoglycan, hyaluronic acid, chitosan, alginic acid, agarose, polyethylene glycol hydrogel, peptide gel, DNA gel, ultra Molecular gel, polylactic acid, polyethylene glycol, silica gel, cellulose and the like.
  • fibrin, Matrigel, collagen and the like are particularly preferred.
  • the organoid-forming composition is gel-like or sol-like, preferably gel-like.
  • the organoid-forming composition can be obtained by mixing cells with a cell scaffold material.
  • a cell scaffold material For example, when fibrin is employed as a cell scaffolding material, cells, fibrinogen, and thrombin are mixed in a liquid so that thrombin acts on fibrinogen to form fibrin, thereby forming a gel-like organoid-forming composition.
  • the organoid-forming composition can preferably be a fibrin gel containing cells.
  • the cell concentration in the organoid-forming composition is not particularly limited as long as muscle organoids can be formed from the organoid-forming composition.
  • the cell concentration is, for example, 1 ⁇ 10 6 cells/ml or more, preferably 2 ⁇ 10 6 cells/ml or more, more preferably 3 ⁇ 10 6 cells/ml or more, still more preferably 4 ⁇ 10 6 cells/ml or more, More preferably, it is 5 ⁇ 10 6 cells/ml or more.
  • the cell concentration is preferably 6 ⁇ 10 6 cells/ml or higher, more preferably 7 ⁇ 10 6 cells/ml or higher, and even more preferably 7.5 ⁇ 10 6 cells /ml or higher, within the above concentration range.
  • the upper limits of the cell concentration are, for example, 40 ⁇ 10 6 cells/ml, 20 ⁇ 10 6 cells/ml, 15 ⁇ 10 6 cells/ml and 10 ⁇ 10 6 cells/ml.
  • step (1) muscle differentiation is induced in a culture device with the organoid-forming composition covering the periphery of the pillars.
  • the method for covering the perimeter of the pillars with the organoid-forming composition is not particularly limited, but for example, the precursor solution of the organoid-forming composition is placed on the culture bottom surface of the culture device and spread over the entire culture bottom surface. A method of gelling or solifying the precursor solution in a spread state may be mentioned.
  • the method for inducing muscle differentiation in step (1) is not particularly limited as long as it is a method capable of forming muscle organoids from the organoid-forming composition.
  • Induction of muscle differentiation is preferably performed by transiently expressing MyoD in cells, and/or adding the organoid-forming composition to a low serum medium (serum concentration is, for example, 8% or less, preferably 5% or less, more preferably 3% or less medium) or serum-free medium.
  • step (1) the method for transiently expressing MyoD is the same as the method for producing the initial induced cells described above, except for the following.
  • the period during which MyoD is transiently expressed in cells to induce muscle cells should be sufficient for muscle differentiation, and can be changed as appropriate depending on the type and properties of the cells used.
  • the period is, for example, about 3 to 12 days, preferably about 4 to 10 days, more preferably about 5 to 8 days.
  • expression is stopped by introducing a transposase into the cell after the above-mentioned period of time has passed, and when a vector having a LoxP sequence is used, a desired period of time has passed. Later, a method of terminating expression by introducing Cre into cells is exemplified.
  • the induction of muscle differentiation preferably includes transient expression of MyoD in the cells by induction of expression using a drug.
  • the drug preferably includes doxycycline.
  • culture is performed in a medium containing doxycycline to transiently express MyoD.
  • concentration of doxycycline in the medium is, for example, 0.01 ⁇ g/ml or more, preferably 0.02 ⁇ g/ml or more, more preferably 0.05 ⁇ g/ml or more, still more preferably 0.07 ⁇ g/ml or more, and even more preferably 0.09 ⁇ g/ml or more. , particularly preferably 0.1 ⁇ g/ml or more.
  • the upper limit of the concentration is, for example, 5 ⁇ g/ml, 4 ⁇ g/ml, 3 ⁇ g/ml, or 2 ⁇ g/ml.
  • a lock inhibitor-containing medium it is preferable to culture in a lock inhibitor-containing medium during part or all of the culture period for inducing muscle differentiation in step (1). This can lead to higher muscle contractility of the resulting muscle organoids, and even neuro-muscle organoids, making these organoids more suitable for drug screening.
  • lock inhibitors those described above can be used.
  • the concentration of the lock inhibitor in the medium is, for example, 1-50 ⁇ M, preferably 2-40 ⁇ M, more preferably 5-20 ⁇ M.
  • the period of culture in the lock inhibitor-containing medium is, for example, 1 to 4 days, preferably 1 to 3 days, more preferably 1 to 2 days.
  • the medium amount in step (1) is not particularly limited.
  • the amount of medium in step (1) is, for example, 5 to 200 ⁇ l per 1 ⁇ 10 4 of the cells (cells in the organoid-forming composition) at the start of muscle differentiation induction in step (1a).
  • the volume of the medium is preferably 5-100 ⁇ l, more preferably 5-50 ⁇ l, even more preferably 10-40 ⁇ l, still more preferably 10-30 ⁇ l.
  • Muscle organoids are obtained from the organoid-forming composition in step (1).
  • the availability of muscle organoids can be confirmed by the presence of muscle markers such as MHC and MEF2c. It should be noted that the muscle organoids produced in this manner may be a cell population containing several types of cells rather than a single cell population.
  • Muscle organoids can be contracted by electrical stimulation. The contractile force can be measured according to or according to the method described in Test Example 3 below.
  • step (2) the muscle organoids obtained in step (1) are cultured in a medium containing neurotrophic factors to induce neuronal differentiation to obtain nerve-muscle organoids.
  • Neurotrophic factors are ligands to membrane receptors that play an important role in the survival and maintenance of motor neuron function.
  • NT-3 Neurotrophin 4/5 (NT-4/5), Neurotrophin 6 (NT-6), basic FGF, acidic FGF, FGF-5, Epidermal Growth Factor (EGF), Hepatocyte Growth Factor (HGF), Insulin , Insulin Like Growth Factor 1 (IGF 1), Insulin Like Growth Factor 2 (IGF 2), Glia cell line-derived Neurotrophic Factor (GDNF), TGF-b2, TGF-b3, Interleukin 6 (IL-6), Ciliary Neurotrophic Factor (CNTF) and LIF.
  • Preferred neurotrophic factors in the present invention are GDNF and BDNF.
  • Neurotrophic factors are commercially available from, for example, Wako, R&D systems, etc. and can be easily used, but may be obtained by forced expression in cells by methods known to those skilled in the art.
  • the concentration of GDNF in the culture medium can be, for example, 0.1 ng/mL to 100 ng/mL, preferably 1 ng/mL to 50 ng/mL, more preferably 5 ng/mL to 20 ng/mL.
  • the concentration of BDNF in the culture medium can be, for example, 0.1 ng/mL to 100 ng/mL, preferably 1 ng/mL to 50 ng/mL, more preferably 5 ng/mL to 20 ng/mL.
  • the culture medium used in step (2) can be prepared using a medium used for culturing animal cells as a basal medium.
  • basal media include Glasgow's Minimal Essential Medium (GMEM) 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, and RPMI 1640. Medium, Fischer's medium, Neurobasal Medium (Life Technologies) and mixed medium of these are included.
  • the medium may contain serum or may be serum-free.
  • the medium contains, for example, albumin, transferrin, Knockout Serum Replacement (KSR) (serum replacement for FBS during ES cell culture), N2 supplement (Invitrogen), B27 supplement (Invitrogen), fatty acids, insulin, collagen May contain one or more serum replacements such as precursors, trace elements, 2-mercaptoethanol, 3'-thiolglycerol, lipids, amino acids, L-glutamine, Glutamax (Invitrogen), non-essential amino acids, vitamins, It may also contain one or more substances such as growth factors, small molecules, antibiotics, antioxidants, pyruvate, buffers, inorganic salts, and the like.
  • Preferred media are media containing non-essential amino acids, B27 supplement, cAMP, GDNF, BDNF, IGF, retinoic acid, purmorphamine, and ascorbic acid.
  • neural differentiation can be induced by stopping MyoD expression and changing the medium to a medium for neural differentiation.
  • the culture temperature is not particularly limited, but is about 30 to 40°C, preferably about 37°C . 5%.
  • the culture period is not particularly limited as long as motor neurons and Schwann cells appear, but step (2) is preferably performed for at least 3 days. It is more preferably 4 days or more, still more preferably 5 days or more.
  • the upper limit of the culture period is not particularly limited, and is, for example, 30 days, 50 days, 100 days, or 200 days. Longer periods of culture can yield more mature organoids. However, from the viewpoint of cost and convenience, the upper limit of the culture period is preferably 20 days, more preferably 12 days, and even more preferably 10 days.
  • neuromuscular junctions that is, neuromuscular organoids containing motor neurons, muscle cells, Schwann cells, etc., can be obtained.
  • an organoid-forming composition or a differentiation inducer thereof (a differentiation inducer in the middle of differentiation from an organoid-forming composition to a muscle organoid, a muscle organoid, a muscle organoid to a nerve-muscle Differentiation inducers in the middle stages of differentiation into organoids, or neuro-muscle organoids) tend to migrate toward the top of the pillar as the contractile force increases.
  • this lifting operation is not particularly limited, and for example, a centrifugal operation, a suction operation, or the like can be employed. In order to reduce sample-to-sample variability in muscle contractility, this lifting operation is preferably performed by centrifugation.
  • the production method of the present invention preferably further includes a step (3) of moving the organoid-forming composition and/or its differentiation inducer to the upper part of the pillar by centrifugation.
  • the centrifugation operation is not particularly limited as long as the composition for forming an organoid and/or its differentiation inducer can move to the upper part of the pillar.
  • a culture dish or microwell plate equipped with a culture device is placed in a centrifuge with a lid so that the medium does not spill out, and the pillar top faces in the direction opposite to the center of centrifugation, By centrifuging, the organoid-forming composition and/or its differentiation inducer can be moved to the top of the pillars.
  • the centrifugal force can be appropriately set according to the length of the pillar, the shape of the pillar, the structure of the upper end of the pillar, etc.
  • the centrifugal force is, for example, 50-500 ⁇ g, preferably 80-300 ⁇ g, more preferably 100-250 ⁇ g.
  • the centrifugation time can be set as appropriate according to the length of the pillar, etc.
  • the centrifugation time is, for example, 1 second to 1 minute, preferably 3 to 30 seconds, more preferably 5 to 15 seconds.
  • the number of times of step (3) is not particularly limited.
  • the number of times can be, for example, 1 to 10 times or 1 to 5 times from the viewpoint of cost and convenience.
  • kit 1 In one aspect of the present invention, a kit for producing nerve-muscle organoids, comprising a culture device having at least two pillars extending from the bottom surface of the culture to the side of the culture tank, and the culture device has one culture tank, Regarding. This will be explained below.
  • the culture device is as explained above.
  • the kit of the present invention is preferably a kit for use in the production method of the present invention.
  • the kit of the present invention preferably contains the cells, culture medium, additives, etc. used in steps (1) and (2) described above.
  • one or more reagents selected from the group consisting of pluripotent stem cells introduced in a MyoD-inducible state, drugs such as tetracycline or derivatives thereof, neurotrophic factors, culture dishes, microwell plates, and basal medium
  • a kit containing The kit of the present invention may further include a document or an instruction describing the procedure of the manufacturing process.
  • Screening method In one aspect of the present invention, (Aa) a step of performing some or all of the steps of the production method of the present invention in the presence of a test substance to obtain an organoid, or (Ab) a neuro-muscle organoid obtained by the production method of the present invention and a test substance contacting; and (B) assessing the function and/or condition of the organoids; and screening methods for therapeutic agents for neuromuscular diseases, including This will be explained below.
  • SBMA spinobulbar muscular atrophy
  • Test substances are, for example, cell extracts, cell culture supernatants, microbial fermentation products, extracts from marine organisms, plant extracts, purified or crude proteins, peptides, non-peptide compounds, synthetic low-molecular-weight compounds, and natural compounds. are exemplified.
  • Test substances can also be used in (1) biological libraries, (2) synthetic library methods using deconvolution, (3) "one-bead one-compound” library methods, and (4) can be obtained using any of the many approaches in combinatorial library methods known in the art, including synthetic library methods using affinity chromatography selection; Biological library methods using affinity chromatography sorting are limited to peptide libraries, whereas the other four approaches can be applied to small compound libraries of peptides, non-peptide oligomers, or compounds (Lam (1997 ) Anticancer Drug Des. 12: 145-67). Examples of methods for synthesizing molecular libraries can be found in the art (DeWitt et al. (1993) Proc. Natl. Acad. Sci.
  • Step (Aa) can be carried out, for example, by adding the test substance to the culture medium of step (1) and/or step (2).
  • Step (Ab) can be carried out, for example, by adding the test substance to the medium containing the neuro-muscle organoids.
  • the concentration of the test substance in the medium can be appropriately set according to the type of the test substance.
  • steps (Aa) and (Ab) a substance that induces or exacerbates neuromuscular disease can be used together with the test substance. Further, in step (Aa) and step (Ab), as cells in the organoid-forming composition, cells having gene mutations that induce or exacerbate neuromuscular diseases are used, or obtained using the cells Organoids can also be used.
  • the period of contact with the test substance is not particularly limited, but can be, for example, 1 hour to 14 days.
  • the evaluation target in step (B) is the function and/or state of the organoid.
  • Organoid functions include, but are not limited to, contractile force.
  • the contractile force can be measured, for example, according to or according to the methods described in Test Examples 2-4 and 3 below.
  • the state of the organoid is not particularly limited, but includes, for example, the shape and size of the organoid, and the expression level and location of genes such as muscle cell markers and nerve cell markers in the organoid. These can be measured by, for example, PCR, Western blotting, immunostaining, etc., according to or in accordance with conventionally known methods.
  • the test substance can be selected as a therapeutic drug (or a candidate substance thereof) for neuromuscular diseases.
  • a nerve-muscle organoid muscle contraction test kit comprising a culture device having at least two pillars extending from the bottom surface of the culture to the culture tank side and a nerve-muscle organoid formed around the pillars , concerning.
  • the culture device, etc. are as explained above.
  • the kit of the present invention is preferably a kit for use in the screening method of the present invention.
  • the kit of the present invention preferably contains the cells, culture medium, additives, etc. used in the screening method of the present invention.
  • Examples include kits containing one or more reagents selected from the group consisting of test substances, culture dishes, microwell plates, and basal media.
  • the kit of the present invention may further include written documents or instructions describing procedures such as a muscle contraction test, screening steps, and the like.
  • the culture device 1 includes a culture bottom surface 11 and two pillars (pillar 12a, pillar 12b). The two pillars extend from the culture bottom surface 11 toward the culture tank 14 side.
  • the culture device 1 has a wall 15 so that the culture bottom surface 11 has a dumbbell shape.
  • the pillars 12a, 12b are provided with semicircular barrier structures 13a, 13b at their upper ends (ends opposite to the culture bottom surface 11).
  • the manufacturing method of the culture device is as follows.
  • the culture device was fabricated using PDMS (Polydimethylsiloxane) (SILPOT 184, DuPont Toray Spacialty Materials, Tokyo, Japan).
  • PDMS Polydimethylsiloxane
  • SILPOT 184 DuPont Toray Spacialty Materials, Tokyo, Japan.
  • PDMS was poured into a mold made of Teflon and solidified by heating at 70°C for 1 hour. After removing from the mold, a disk made of thin film PDMS was placed on top of the pillar.
  • a thin film disc was prepared as follows: a few drops of PDMS were dropped on a Teflon sheet, the rotation speed was increased to 1000 rpm for 30 seconds, spin-coated for 30 seconds, and then heated at 70°C for 1 hour.
  • the thin film disk was placed on the top of the pillars of the culture device and fixed to the top of the pillars by heating at 70°C for 1 hour. The thin film disk was then cut in half to give a semicircular shape.
  • FIG. 6 shows a schematic diagram of the wells of the culture device-mounted microwell plate. The bottom of FIG. 6 is the bottom of the well.
  • the prepared plates were sterilized by UV light with the lid open for at least 30 minutes.
  • the culture device was filled with a 2% Pluronic (registered trademark) solution the day before the start of culture and allowed to stand at 4°C. Pluronic solution was removed immediately before use.
  • Test example 1 Production of nerve-muscle organoids Induced cells were prepared by transiently expressing MyoD in pluripotent stem cells (Test Examples 1-1 and 1-2) and cultured (Test Example 1-3). After inducing muscle differentiation to obtain a muscle organoid while covering the two pillars of the device (Test Example 1-4), the obtained muscle organoid was kept in contact with the two pillars. The cells were cultured in a medium containing neurotrophic factors to induce neuronal differentiation to obtain nerve-muscle organoids (Test Example 1-5). An outline of the manufacturing process is shown in FIG. Details of the materials and methods used are as follows.
  • Example 1-1 Drug-dependent MyoD overexpressing human iPS cell line 409B2 MyoD > Undifferentiated human iPS cell line 409B2 MyoD (Uchimura, T., et al., A human iPS cell myogenic differentiation system permitting high-throughput drug screening, Stem Cell res. 25: 98-106 (2017)) is Kyoto University iPS cell Provided by the laboratory. 409B2 MyoD cells are induced to overexpress exogenous MyoD by Doxycycline, and iPS cells can be directly induced to become skeletal muscle cells. In addition, 409B2 MyoD cells have been introduced with puromycin resistance, and by adding puromycin to the medium, non-transfected cells can be removed.
  • ⁇ Test example 1-2 Undifferentiated culture of iPS cells> 409B2 MyoD, human iPS cells, were cultured according to the following procedure. Media was removed from the growing T25 flask and washed once with PBS, followed by TrypLE TM Select CTS TM (A12859-01, Life Technologies, California, USA) and UltraPure TM 0.5 M diluted 1:1000 in PBS. 350 ⁇ l of a trypsin solution mixed with an equal volume of EDTA pH 8.0 (15575-038, Life Technologies, California, USA) was added and incubated in a 37°C incubator for 4 minutes.
  • the lock inhibitor Y-27632 (08945-71, Nacalai Tesque, Japan) was added to 10 ⁇ M StemFit TM (AK02N, Ajinomoto, Tokyo, Japan). was added, and the cells were harvested with a cell scraper (99002, Techno Plastic Products, Trasadingen, Switzerland). After suspending the recovered cells 10 times with a 1 ml pipetman, the cell count was calculated using a fully automatic cell counter (TC20 TM , Bio-Rad Laboratories, California, USA), and the cells were added to a T25 flask at 25,000 cells/ml.
  • TC20 TM fully automatic cell counter
  • AK02N containing 10 ⁇ M of Y-27632 and 10 ⁇ l of iMatrix-511 silk were cultured together in an incubator at 37° C. and 5% CO 2 . After 48 hours, the medium was replaced with AK02N without Y-27632 and iMatrix-511 Silk, and then after 72 hours, the medium was replaced. After an additional 48 hours, subconfluent cells were passaged again.
  • ⁇ Test example 1-3 Early differentiation induction of human iPS cells into skeletal muscle cells> Corning (registered trademark) Matrigel (registered trademark) Basement Membrane Matrix (354234, Corning. USA) was added to a 6-well plate (TR5000, Trueline, Japan) in serum-free DMEM/F-12 (Dulbecco's Modified Eagle Medium: Nutrient Mixture F- 12) Matrigel solution diluted 100-fold with (042-30555, Wako, Japan) was added at 1 ml/well and allowed to stand in a 37°C incubator for 1 hour.
  • DMEM/F-12 Dulbecco's Modified Eagle Medium: Nutrient Mixture F- 12
  • the matrigel solution is removed, and the human iPS cells obtained by the same procedure as at the time of passage are suspended in StemFit containing 10 ⁇ M Y-27632, and 1 ml per well is 1.5*10 5 cells/ml. and cultured in a 37°C, 5% CO 2 incubator.
  • Human ES cell medium (DMEM/F-12 + 20% Knockout Serum Replacement (KSR, 10828028, Sigma-Aldrich), 1% MEM Non-Essential Amino Acids solution (NEAA, 11140050, Gibco TM , USA), 2 Replace the medium with 10 ⁇ M Y-27632 added to mM L-glutamine, 100 ⁇ M 2-Mercaptoethanol (2-ME, 21438-82, Wako, Japan) and incubate at 37°C in a 5% CO 2 incubator for 24 hours. cultured for hours.
  • the medium was replaced with a human ES cell medium supplemented with 1 ⁇ g/ml Doxycycline Hyclate (Dox, D4116, Tokyo Chemical Industry), and cultured at 37° C. in a 5% CO 2 incubator for 24 hours.
  • Doxycycline Hyclate Dox, D4116, Tokyo Chemical Industry
  • Example 1-4 Preparation and induction of human iPS cell skeletal muscle tissue> The medium was removed from the 6-well plate in which the human iPS cells were initially differentiated, and the plate was washed with PBS three times. 350 ⁇ l of Accutase was added, and the mixture was allowed to stand in a 37°C incubator for 4 minutes. Add 1 ml of muscle induction medium ( ⁇ MEM + 10% KSR, 2% UltroserG, 200 ⁇ M 2-ME, 0.5% penicillin/streptomycin) supplemented with Y-27632 to 10 ⁇ M, pipette 10 times, Collected in a centrifuge tube. It was centrifuged at 800 rpm for 5 minutes and the supernatant was removed.
  • muscle induction medium ⁇ MEM + 10% KSR, 2% UltroserG, 200 ⁇ M 2-ME, 0.5% penicillin/streptomycin
  • the cells were counted and suspended in a growth medium to a concentration of 16.67*10 6 cells/ml. 48.4% cell suspension, 20% DMEM (2X), 20% Fibrinogen from bovine plasma (10 mg/ml) (F8630, Sigma-Aldrich, Massachusetts, USA), 10% Matrigel® Basement Membrane Matrix (354234) , Corning. USA) and 1.6% Thrombin (2%) (T4648, Sigma-Aldrich, Massachusetts, USA). Solidified by incubating. Then, 200 ⁇ l/well of muscle induction medium containing 10 ⁇ M Y-27632 and 1 ⁇ g/ml Dox plus 1 mg/ml TA to prevent gel degradation was added at 37°C, 5% CO. 2 cultured in an incubator. Every 24 hours thereafter, the medium was changed to myogenic induction medium without Y-27632 and supplemented with Dox, TA.
  • Example 1-5 Induction of human iPS cell neuromuscular organoids> After fibrin gel preparation and muscle induction for 6 days (Fig. 7: D6), the tissue was lifted to the top of the pillars, and the medium was changed to neural induction medium (KBM Neural stem cell (16050100, Kohjin Bio)) containing 1.0 mg/ml TA.
  • KBM Neural stem cell 16050100, Kohjin Bio
  • the cells
  • Test example 2 Evaluation of nerve-muscle organoids ⁇ Test Example 2-1. Acquisition of Shrinkage Characteristics> On the 14th day after organization (Fig. 7: D0), electrical stimulation with a voltage of 4 V/mm, a frequency of 30 Hz, and a pulse duration of 2 ms was applied to the neuromuscular organoids for 3 seconds. A digital camera for microscopes (ORCA-Spark, Hamamatsu Photonics, Hamamatsu, Japan) incorporated in Japan) was used to shoot movies at a frame rate of 60 Hz. Motion tracking was performed on the captured video using PV Studio 2D (OA Science, Miyazaki, Japan), and the state of contraction was quantified.
  • PV Studio 2D OA Science, Miyazaki, Japan
  • RNA extraction was performed using Nucleo Spin® RNA XS (740902.50, MACHEREY-NAGEL, Germany). The tissue on the device was washed with PBS three times, removed from the culture device, the PBS was wiped off, and the three samples were collectively collected in a microtube. After that, RNA was extracted according to the protocol of the kit. RNA quantification was performed using Qubit 3.0 Fluorometer (Invitrogen, USA) and Qubit RNA HS assay (Q32852, Invirogen).
  • Reverse transcription to cDNA was performed using ReverTra Ace qPCR RT Master Mix with gDNA Remover (FSQ-301, TOYOBO, Japan). Samples were diluted with Nuclease-free Water so that the RNA concentration was 16.7 ng/ ⁇ l, and the protocol was followed. The generated cDNA was stored at -80°C. RT-PCR was detected with StepOne real-time PCR System (Applied Biosystems, USA) using THUNDERBIRD SYBR qPCR Mix (QPS201, TOYOBO). First, the cDNA was denatured by heating at 95°C for 1 minute, followed by 40 cycles of annealing and extension at 60°C for 1 minute. The sequences of the primers used are shown below. Relative changes in gene expression were calculated by the comparative Ct ( ⁇ Ct) method and normalized to GAPDH as an endogenous control gene.
  • Fig. 10 The results are shown in Fig. 10. In neuromuscular organoids (16 days after organization (FIG. 7: D0)), it was found that the expression of the neuronal cell marker gene (MNX1) is enhanced.
  • a base solution PBS + 10% Goat serum (S-1000, Vector Laboratories, USA), 0.01% Triton X-100) was added and allowed to react at room temperature for 1 hour for blocking.
  • a primary antibody was added to the base solution and allowed to react overnight at room temperature.
  • the primary antibodies used were anti- ⁇ -actinin mouse IgG antibody (A7811-2ML, Sigma-Aldrich) and anti-tubulin ⁇ -3 rabbit IgG antibody (802001, Biolegend, USA), respectively.
  • a secondary antibody and DAPI 340-07971, Dojindo
  • the secondary antibodies used were CF488A TM Goat anti rabbit IgG (H+L) (2 mg/ml, 20012, biotium) and CF555 TM Goat anti mouse IgG (H+L) (2 mg/ml, 20231, biotium).
  • CF488A Goat anti rabbit IgG
  • H+L 2 mg/ml, 20012, biotium
  • CF555 TM Goat anti mouse IgG H+L
  • the tissue was placed on a slide glass, and a few drops of mounting medium for fluorescent staining (H-1400, Vector Laboratories) were added. Osaka, Japan) was used to acquire fluorescence images.
  • Example 2-4 Functional Evaluation of Neuromuscular Junction>
  • Glutamic acid (070-00502, Wako), an agonist of NMDA receptors, was used to stimulate motor neurons.
  • Glutamate is known to be a neurotransmitter that can act on lower motor neurons to promote neuronal firing.
  • glutamic acid was added to a final concentration of 400 ⁇ M, and the appearance of the tissue was observed with a microscope digital camera (ORCA-Spark, Hamamatsu Photonics, Hamamatsu, Japan) incorporated into an upright microscope (BX53F, OLYMPUS, Japan).
  • the video was shot at a frame rate of 60Hz.
  • tetrodotoxin was added to a final concentration of 2 ⁇ M, and the state of the tissue was observed.
  • Tetrodotoxin can inhibit neuronal firing by selectively blocking voltage-gated sodium channels in the excitatory membrane of motor neurons.
  • nerve cell stimulation causes skeletal muscle contraction via the neuromuscular junction
  • glutamic acid was added to confirm that nerve cell stimulation causes skeletal muscle contraction via the neuromuscular junction
  • vecuronium bromide Curare , 223-01811, Wako
  • Test example 3 A skeletal muscle tissue was constructed in the same manner as the production of human iPS cell skeletal muscle tissue with optimized Doxycycline concentration (Test Examples 1-1 to 1-4).
  • concentration of Doxycycline added to the muscle induction medium during culture was set to 0, 0.01, 0.1, and 1 ⁇ g/ml, and culture was performed for 6 days (up to D6 in FIG. 7). The tissue was lifted and the muscle contraction force was measured.
  • the details of the method for measuring muscle contractility are as follows.
  • Electrodes were fabricated using a 3D printer and platinum electrodes in order to measure the contractile force by applying electrical stimulation to muscle tissue. Chemically stable platinum was used as the electrode, and it was designed as a cylindrical electrode with a diameter of 0.5 mm and a distance between the electrodes of 5 mm.
  • the 3D-printed electrode base has a window at the top of each well, allowing observation of the muscle tissue. Each electrode is controlled by a selector switch, enabling electrical stimulation per well.
  • C-PACE C-PACE 100, IonOptix, Massachusetts, USA
  • Electrical stimulation is generated by C-PACE and distributed to each well by the fabricated electrodes.
  • a voltage of 4 V/mm, a frequency of 30 Hz, and a pulse duration of 2 msec were used for the measurements, using a microscope digital camera (ORCA-Spark, Hamamatsu Photonics, Hamamatsu, Japan) incorporated in an upright microscope (BX53F, OLYMPUS, Japan).
  • the movement of muscle tissue was imaged by The micropost displacement was calculated using image analysis software ImageJ from photographs of muscle tissue before and after electrical stimulation.
  • the measured micropost displacement was converted to contractile force using the following equation.
  • E modulus of elasticity of PDMS
  • R radius of cylinder
  • L length of cylinder
  • the distance contracted by each muscle tissue.
  • F 3 ⁇ ER4 ⁇ / 4L3
  • Test example 4 Analysis of Effect of ROCK Inhibitor Skeletal muscle tissue was constructed in the same manner as in the preparation of human iPS cell skeletal muscle tissue (Test Examples 1-1 to 1-4).
  • the cells were cultured under the condition that Y-27632 (ROCK inhibitor) was added/not added (PBS only) to the muscle induction medium during the initial two days of culture (Fig. 7: D0 to 1). After culturing for 6 days (up to D6 in FIG. 7), lifting operation was performed, and muscle contraction force was measured in the same manner as in Test Example 3.
  • Y-27632 ROCK inhibitor
  • Test example 5 Optimization of Cell Density
  • the early differentiation-induced cells obtained in Test Example 1-3 were collected with Accutase, and the number of cells was counted.
  • cell suspensions were prepared at concentrations of 16.67, 12.5, 8.33 and 4.17*10 6 cells/ml, and fibrin gels were prepared as described in Test Example 1-4.
  • the cell densities in the fibrin gel are 8, 6, 4, 2*10 6 cells/ml respectively.
  • culture was performed as in Test Example 1-4, and after culturing for 6 days (up to D6 in FIG. 7), lifting operation was performed, and muscle contraction force was measured in the same manner as in Test Example 3.
  • Test example 6 Comparison of medium amount Culture devices were fixed to 96-well plates and 48-well plates. The fixing method is the same as in Reference Example 2. Using the obtained well plate, skeletal muscle tissue was constructed in the same manner as in the preparation of human iPS cell skeletal muscle tissue (Test Examples 1-1 to 1-4). Medium volume is 200 ⁇ l for 96-well plates and 700 ⁇ l for 48-well plates. After culturing for 6 days (up to D6 in FIG. 7), muscle contractility was measured over time in the same manner as in Test Example 3.
  • Test example 7 We investigated the method of lifting the tissue in the improvement of the culture method and the induction of muscle differentiation. Details of the materials and methods used are as follows.
  • Example 7-1 Subculture of skeletal muscle cells> Hu5/KD3 (transferred from the National Center for Geriatrics and Gerontology) was used as skeletal muscle cells and cultured according to the following procedure. Collagen coating of the flask was performed first. A collagen solution was prepared by dissolving collagen (Cellmatrix type I-C: Nitta Gelatin Co., Ltd., Osaka, Japan) in 0.02N acetic acid at a concentration of 50 ⁇ g/ml. 5 ml of the prepared collagen solution was added to a T75 flask and allowed to stand at room temperature for 1 hour. After that, the collagen solution was removed and washed twice with 5 ml of PBS.
  • collagen Cellmatrix type I-C: Nitta Gelatin Co., Ltd., Osaka, Japan
  • the culture medium was removed from the cultured T75 flask, washed twice with PBS, added with 1 ml of 0.05% trypsin, and incubated in a 37° C. incubator for 2 minutes.
  • muscle growth medium DMEM (20% FBS, 2mM L-glutamine (073-05391, Wako, Tokyo, Japan), 0.5% penicillin/streptomycin, 2% UltroserG (15950-017, Sartorius, Gottingen, Germany)
  • DMEM 20% FBS, 2mM L-glutamine (073-05391, Wako, Tokyo, Japan), 0.5% penicillin/streptomycin, 2% UltroserG (15950-017, Sartorius, Gottingen, Germany)
  • the resulting pellet was suspended in 2 ml of muscle growth medium, the number of cells was calculated using a fully automatic cell counter (TC20 TM , Bio-Rad Laboratories, California, USA), and 2.0*10 5 cells were added to a coated flask. Add to 10 ml of muscle growth medium. The cells were seeded on T75 and cultured in an incubator at 37°C, 10% CO 2 , 5% O 2 . After 72 hours, the subconfluent cells were subcultured again.
  • TC20 TM fully automatic cell counter
  • Example 7-2 Preparation of skeletal muscle tissue and lifting of muscle tissue by centrifugal operation> The cells cultured in the T75 flask were collected by the same method as the subculture operation, the number of cells was counted, and the cells were suspended in a growth medium to a concentration of 4.17*10 6 cells/ml. 48.4% cell suspension, 20% DMEM (2X), 20% Fibrinogen from bovine plasma (10 mg/ml) (F8630, Sigma-Aldrich, Massachusetts, USA), 10% Matrigel® Basement Membrane Matrix (354234) , Corning.
  • a conventional aspiration operation and a new centrifugation operation were used for lifting.
  • a gel loading tip Q-010, QSP
  • muscle tissue was grasped by suction and lifted to the top of the pillar.
  • the plate was placed upside down in a tabletop centrifuge (5010, KUBOTA, Japan) and 200 xg for 10 seconds to lift the muscle tissue upwards.
  • 200 ⁇ l/well of muscle differentiation medium DMEM (+ 2% FBS, 1% penicillin/streptomycin, 1% ITS supplement (I3146, Sigma-Aldrich, Massachusetts, USA)

Landscapes

  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Virology (AREA)
  • Sustainable Development (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

La présente invention concerne une technologie permettant de produire plus facilement et plus efficacement un organoïde neural-musculaire. Le procédé de production d'un organoïde neural-musculaire comprend les étapes suivantes : (1a) obtention d'un organoïde musculaire, dans un dispositif de culture muni d'au moins deux piliers s'étendant de la surface du fond de culture vers un récipient de culture, par induction de la différenciation musculaire, dans un état de recouvrement des périphéries des piliers, vers une composition formant un organoïde contenant au moins un type cellulaire choisi dans le groupe constitué par les cellules souches pluripotentes et les cellules à induction précoce obtenues par expression transitoire de MyoD dans des cellules souches pluripotentes ; et (2) obtention d'un organoïde neural-musculaire par culture de l'organoïde musculaire obtenu dans l'étape (1a) dans un milieu de culture contenant un facteur neurotrophique pour y induire une différenciation neurale.
PCT/JP2022/037274 2021-10-11 2022-10-05 Procédé de production d'organoïde WO2023063187A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2021166757A JP2023057310A (ja) 2021-10-11 2021-10-11 オルガノイドの製造方法
JP2021-166757 2021-10-11

Publications (1)

Publication Number Publication Date
WO2023063187A1 true WO2023063187A1 (fr) 2023-04-20

Family

ID=85988620

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2022/037274 WO2023063187A1 (fr) 2021-10-11 2022-10-05 Procédé de production d'organoïde

Country Status (2)

Country Link
JP (1) JP2023057310A (fr)
WO (1) WO2023063187A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116731859A (zh) * 2023-05-12 2023-09-12 武汉大学 一种环形大脑类器官模型及其构建方法与应用

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
LIN CHUANG-YU, YOSHIDA MICHIKO, LI LI-TZU, IKENAKA AKIHIRO, OSHIMA SHIORI, NAKAGAWA KAZUHIRO, SAKURAI HIDETOSHI, MATSUI ERIKO, NAK: "iPSC-derived functional human neuromuscular junctions model the pathophysiology of neuromuscular diseases", JCI INSIGHT, vol. 4, no. 18, 19 September 2019 (2019-09-19), XP055810523, DOI: 10.1172/jci.insight.124299 *
TATSUYA OSAK, UZEL SEBASTIEN G M, KAMM ROGER D: "Microphysiological 3D model of amyotrophic lateral sclerosis (ALS) from human iPS-derived muscle cells and optogenetic motor neurons", SCIENCE ADVANCES, vol. 4, 10 October 2018 (2018-10-10), pages 1 - 15, XP055618418, DOI: 10.1126/sciadv.aat5847 *
YOSHIOKA KANTARO; ITO AKIRA; ARIFUZZAMAN MD; YOSHIGAI TAICHI; FAN FANGMING; SATO KEI-ICHIRO; SHIMIZU KAZUNORI; KAWABE YOSHINORI; K: "Miniaturized skeletal muscle tissue fabrication for measuring contractile activity", JOURNAL OF BIOSCIENCE AND BIOENGINEERING, ELSEVIER, AMSTERDAM, NL, vol. 131, no. 4, 23 December 2020 (2020-12-23), NL , pages 434 - 441, XP086525912, ISSN: 1389-1723, DOI: 10.1016/j.jbiosc.2020.11.014 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116731859A (zh) * 2023-05-12 2023-09-12 武汉大学 一种环形大脑类器官模型及其构建方法与应用
CN116731859B (zh) * 2023-05-12 2024-04-30 武汉大学 一种环形大脑类器官模型及其构建方法与应用

Also Published As

Publication number Publication date
JP2023057310A (ja) 2023-04-21

Similar Documents

Publication Publication Date Title
KR102297584B1 (ko) 망막 색소 표피 세포의 제조 방법
KR102278978B1 (ko) 망막 조직 및 망막 관련 세포의 제조 방법
Gärtner et al. Nucleofection of primary neurons
JP7420334B2 (ja) 液体注入方法
JP6612736B2 (ja) 心筋細胞の選別方法
WO2019189545A1 (fr) Procédé de production de cellules
JP6883791B2 (ja) 骨格筋前駆細胞の選択方法
WO2023063187A1 (fr) Procédé de production d'organoïde
JP7236738B2 (ja) 神経前駆細胞の選別方法
EP3219808B1 (fr) Procédé de criblage utilisant des neurones induits
WO2021172542A1 (fr) Procédé de production de cardiomyocytes matures
JP6780197B2 (ja) 新規成熟心筋細胞マーカー
WO2020100481A1 (fr) Procédé de production d'organoïdes cérébraux
Sugimoto et al. Effect of NeuroD2 expression on neuronal differentiation in mouse embryonic stem cells
JP6990921B2 (ja) 上位運動ニューロンの誘導方法
JP7140400B2 (ja) 多能性幹細胞から人工神経筋接合部を得る方法
Buttermore Phenotyping of Human IPSC-derived Neurons: Patient-driven Research
WO2020090836A1 (fr) Procédé de production de cellules
JP2023086705A (ja) 筋疲労/筋損傷の細胞モデル、その製造方法及びその用途
WO2018043303A1 (fr) Procédé de production de neurone à sérotonine

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: 22880883

Country of ref document: EP

Kind code of ref document: A1