WO2014097875A1 - Développement de cellules souches pluripotentes à l'aide d'un nouveau procédé d'induction d'une dédifférenciation - Google Patents

Développement de cellules souches pluripotentes à l'aide d'un nouveau procédé d'induction d'une dédifférenciation Download PDF

Info

Publication number
WO2014097875A1
WO2014097875A1 PCT/JP2013/082399 JP2013082399W WO2014097875A1 WO 2014097875 A1 WO2014097875 A1 WO 2014097875A1 JP 2013082399 W JP2013082399 W JP 2013082399W WO 2014097875 A1 WO2014097875 A1 WO 2014097875A1
Authority
WO
WIPO (PCT)
Prior art keywords
mrna
pluripotent stem
cells
mir
gene
Prior art date
Application number
PCT/JP2013/082399
Other languages
English (en)
Japanese (ja)
Inventor
典正 三浦
Original Assignee
国立大学法人鳥取大学
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 国立大学法人鳥取大学 filed Critical 国立大学法人鳥取大学
Priority to JP2014553063A priority Critical patent/JPWO2014097875A1/ja
Publication of WO2014097875A1 publication Critical patent/WO2014097875A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7105Natural ribonucleic acids, i.e. containing only riboses attached to adenine, guanine, cytosine or uracil and having 3'-5' phosphodiester links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/54Ovaries; Ova; Ovules; Embryos; Foetal cells; Germ cells
    • A61K35/545Embryonic stem cells; Pluripotent stem cells; Induced pluripotent stem cells; Uncharacterised stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/65MicroRNA
    • 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
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to a method for inducing cell dedifferentiation, a method for producing pluripotent stem cells, a therapeutic agent for malignant tumors, and the like.
  • Pluripotent stem cell production technology is a field that has received particular attention in the medical industry in recent years.
  • the method described in Patent Document 1 can be mentioned.
  • This document describes that pluripotent stem cells were prepared by introducing four genes (Oct3 / 4, Klf4, Sox2, c-Myc) into cells. These cells are called iPS cells, and since the development of this technology, the number of reports on research results related to pluripotent stem cells has been increasing rapidly.
  • Patent Document 2 describes that pluripotent stem cells were prepared by introducing three genes (Oct3 / 4, Klf4, Sox2) and one miRNA (hsa-miR-372 etc.) into the cells. Has been.
  • Non-Patent Document 1 describes that the efficiency of producing pluripotent stem cells increased when the p53 gene of cells to be pluripotent stem cells was deleted when the above four or three genes were introduced.
  • Patent Documents 3 and 4 describe that pluripotent stem cells were prepared by introducing specific RNA strands (such as miR-520d-5p) into cells.
  • Non-Patent Document 2 that hTERTTERmRNA can be applied as a biomarker of cancer.
  • Patent Documents 3 and 4 report that specific RNA strands can be used for the treatment of malignant tumors.
  • pluripotent stem cells there are examples of successful production of pluripotent stem cells, but there are no products that have been approved by the authorities for therapeutic use. This is due to the fact that research does not progress easily due to limited methods for producing pluripotent stem cells. Except for a few of the few methods that have confirmed obvious side effects (for example, canceration in the early stage of culture by c-Myc), the effective methods are further limited.
  • malignant tumors are considered to be the top cause of human death in the future. Therefore, it is necessary to identify new anti-malignant tumor substances and accumulate information on malignant tumors in order to develop new drugs or treatment strategies for malignant tumors.
  • the present invention has been made in view of the above circumstances, and an object thereof is to provide a novel dedifferentiation induction method, a pluripotent stem cell production method, a malignant tumor therapeutic agent, and the like.
  • the present inventor has found that inhibition of ELAVL2 expression in normal cells or malignant tumor cells causes the cells to dedifferentiate and become pluripotent stem cells. Furthermore, when the obtained pluripotent stem cells were transplanted subcutaneously into mice, no malignant tumor formation was observed over a long period of 4 months or longer. From this, it became clear that the technique found by the present inventor is very excellent in safety.
  • the present inventors also tried to inhibit the expression of TEAD1 and GATAD2B in normal cells or malignant tumor cells.
  • the cells were dedifferentiated to become pluripotent stem cells. All of these genes were genes whose expression could be controlled by miR-520d-5p. Similar to these genes, other genes whose expression can be controlled by miR-520d-5p include SBF2.
  • a method for inducing cell dedifferentiation which comprises the step of inhibiting the expression or function of a miR-520d-5p regulated gene.
  • a method for producing pluripotent stem cells comprising a step of inducing cell dedifferentiation by the above method.
  • the pluripotent stem cell or population of a pluripotent stem cell obtained through said production method is provided.
  • cultivating the said pluripotent stem cell is provided.
  • a therapeutic agent for malignant tumor comprising an expression or function inhibitor for miR-520d-5p controlled gene.
  • a dedifferentiation inducing agent comprising an expression or function inhibitor for miR-520d-5p controlled gene.
  • a pluripotent stem cell inducer comprising an expression or function inhibitor for miR-520d-5p regulated gene.
  • the miR-520d-5p regulated gene is ELAVL2, TEAD1, GATAD2B, SBF2, PUM2, NBEA, UBE2E3, CPEB3, GNAO1, WNK1, MEF2A, SEH1L, HOOK3, KLHL23, PAPOLG Or one or more genes selected from the group consisting of JAZF1, JAG1, CPEB2, ZCRB1, ZFC3H1, ZEB2, EPHA7, UBN2, PELI2, PRKD3, and FCHO2.
  • the inhibition may be inhibition by an RNAi molecule against mRNA of a miR-520d-5p controlled gene.
  • the inhibitor may be an RNAi molecule against mRNA of a miR-520d-5p-controlled gene, or a polynucleotide encoding the RNAi molecule.
  • the present invention it is possible to induce dedifferentiation, produce pluripotent stem cells, or treat malignant tumors by a novel method.
  • FIG. 1 shows the result of observing the pluripotent stem cells according to the example with a microscope.
  • FIG. 2 shows the results of evaluating the gene expression profile of pluripotent stem cells according to the Example at the transcription level.
  • FIG. 3 shows the results of examining the ELAVL2 expression suppression rate for the ELAVL2 siRNA-introduced HLF cells according to the example.
  • FIG. 4 shows the results of evaluating the gene expression profile of pluripotent stem cells according to the example at the translation level.
  • FIG. 5 shows the results of observing the protein level of pluripotent stem cells according to Example by cell immunostaining.
  • FIG. 6 shows the results of observing the proliferation of pluripotent stem cells according to the example.
  • FIG. 1 shows the result of observing the pluripotent stem cells according to the example with a microscope.
  • FIG. 2 shows the results of evaluating the gene expression profile of pluripotent stem cells according to the Example at the transcription level.
  • FIG. 3 shows the results of examining
  • FIG. 7 shows the results of AP staining of pluripotent stem cells according to the example.
  • FIG. 8 shows the results of transplanting pluripotent stem cells according to the Example into mice.
  • FIG. 9 shows the results of reporter assay performed on ELAVL2 according to the example.
  • FIG. 10 shows the results of examining the ELAVL2 expression suppression rate for the HLF cells into which the ELAVL2 siRNA according to the example was introduced.
  • FIG. 11 shows the results of examining the suppression rate of TEAD1 expression in HLF cells into which TEAD1 siRNA according to the example was introduced.
  • FIG. 12 shows the results of examining the GATAD2B expression suppression rate for HLF cells into which the GATAD2B siRNA according to the example was introduced.
  • FIG. 13 shows the results of examining the DNA methylation level of HLF cells into which ELAVL2 siRNA, TEAD1 siRNA, or GATAD2B siRNA according to the example was introduced.
  • FIG. 14 shows the results of examining the DNA methylation level of Huh7 cells into which ELAVL2 siRNA, TEAD1 siRNA, or GATAD2B siRNA according to the example was introduced.
  • One embodiment of the present invention is a novel dedifferentiation induction method.
  • This method is, for example, a method for inducing cell dedifferentiation, which includes a step of inhibiting the expression or function of a miR-520d-5p-controlled gene.
  • pluripotent stem cells can be produced by inducing cell dedifferentiation, as demonstrated in the examples described later.
  • the inhibition can be performed by, for example, RNAi against mRNA of miR-520d-5p controlled gene.
  • the “miR-520d-5p regulated gene” is a gene whose expression is controlled by miR-520d-5p.
  • the miR-520d-5p controlled gene may have, for example, a miR-520d-5p binding site on the 3 ′ UTR.
  • the miR-520d-5p regulated gene is ELAVL2, TEAD1, GATAD2B, SBF2, PUM2, NBEA, UBE2E3, CPEB3, GNAO1, WNK1, MEF2A, SEH1L, HOOK3 from the viewpoint of more stably causing cell dedifferentiation.
  • the miR-520d-5p controlled gene is preferably ELAVL2, TEAD1, or GATAD2B from the viewpoint of safety to living bodies.
  • the control includes suppression or inhibition.
  • HGNC IDs listed on HGNC are HGNC: 3313 for ELAVL2, HGNC: 11714 for TEAD1, HGNC: 30778 for GATAD2B, HGNC: 2135 for PBF2, HGNC: 14958 for PUM2, HGNC: 7648 for UBE2E3, HGNC for UBE2E3: 12479, CPEB3 is HGNC: 21746, GNAO1 is HGNC: 4389, WNK1 is HGNC: 14540, MEF2A is HGNC: 6993, SEH1L is HGNC: 30379, HOOK3 is HGNC: 23576, KLHL23 is HGNC: 27506, PAPOLG is HGNC: 14982, JAZF1 is HGNC: 14982, JAZF1 is HGNC: 14982, JAZF1 is HGNC: 14982, JAZF1 is HGNC: 14982, JAZF1 is HG
  • ELAVL2 mRNA may contain the base sequence of SEQ ID NO: 29.
  • TEAD1 mRNA may contain the nucleotide sequence of SEQ ID NO: 30.
  • GATAD2B mRNA may contain the base sequence of SEQ ID NO: 31.
  • Each gene may be called differently, and other names are listed on the HGNC website. Therefore, each gene may be called by another name.
  • ELAVL2 contains a gene called HEL-N1 or HuB.
  • miR-520d-5p includes, for example, hsa-miR-520d-5p whose miRBase accession number is MI0003164.
  • One embodiment of the present invention is a method for producing pluripotent stem cells, which comprises a step of inhibiting the expression or function of a miR-520d-5p regulated gene.
  • a pluripotent stem cell or a pluripotent stem cell population can be obtained.
  • a regenerative medical material for example, a regenerative medical organ
  • the pluripotent stem cells obtained by using this production method demonstrate that endogenous p53 is highly expressed in Examples described later. Since p53 is a gene classified as a malignant tumor suppressor gene, it can be said that the pluripotent stem cells that highly express this p53 have a low risk of malignant tumor formation.
  • One embodiment of the present invention is a dedifferentiation inducing agent comprising an expression or function inhibitor for miR-520d-5p regulated genes. If this dedifferentiation inducer is used, cell dedifferentiation can be induced. Moreover, as a result of dedifferentiating the cells, for example, pluripotent stem cells can be obtained. Or if this dedifferentiation inducer is applied to a malignant tumor cell, a malignant tumor can be treated. It can also be used as an additive for assisting animal growth in livestock through effects such as suppression of malignant tumors.
  • One embodiment of the present invention is a method for treating a malignant tumor, comprising a step of inhibiting the expression or function of a miR-520d-5p controlled gene.
  • a therapeutic agent for a malignant tumor comprising an expression or function inhibitor for a miR-520d-5p regulated gene.
  • Another embodiment is the use of an expression or function inhibitor against the miR-520d-5p regulated gene to produce a therapeutic agent for malignancy.
  • a conventional therapeutic agent comprising a low molecular weight compound is one that induces apoptosis to treat a malignant tumor, but the therapeutic method, therapeutic agent, or inducer of this embodiment induces a malignant tumor in the direction of dedifferentiation. By doing so, a therapeutic effect can be exhibited.
  • One embodiment of the present invention is a method for transforming a cell into a pluripotent stem cell, comprising a step of inhibiting the expression or function of a miR-520d-5p regulated gene.
  • a pluripotent stem cell inducer comprising an expression or function inhibitor for a miR-520d-5p regulated gene. If this method or a pluripotent stem cell inducer is used, pluripotent stem cells can be prepared. Or if this method or a pluripotent stem cell inducer is applied to a malignant tumor cell, a malignant tumor can be treated. It can also be used as an additive for assisting animal growth in livestock through effects such as suppression of malignant tumors.
  • One embodiment of the present invention is a method of increasing the proportion of pluripotent stem cells in a cell population, comprising a step of contacting an expression or function inhibitor for a miR-520d-5p regulated gene with the cell population.
  • Yet another embodiment is a method of producing a cell population with an increased proportion of pluripotent stem cells, comprising the step of contacting an expression or function inhibitor for a miR-520d-5p regulated gene with the cell population.
  • One embodiment of the present invention is a research or medical kit containing the inhibitor described above.
  • this kit include dedifferentiation induction, pluripotent stem cell preparation, artificial organ preparation, malignant tumor treatment, undifferentiation marker expression regulation, or p53-expressing cell preparation.
  • the kit may further include, for example, a buffer solution, a package insert describing information on active ingredients, a container for containing the active ingredients, or a package.
  • One embodiment of the present invention comprises a step of selecting a test substance that decreases the expression or functional amount of a miR-520d-5p regulated gene, a dedifferentiation inducer, a pluripotent stem cell inducer, or a malignant tumor
  • a dedifferentiation inducer, a pluripotent stem cell inducer, or a malignant tumor therapeutic agent can be obtained.
  • This method may include a step of introducing a test substance into a cell and a step of measuring the expression or functional amount of a miR-520d-5p controlled gene.
  • Each method according to the above embodiment further includes (a) a step of introducing an expression or function inhibitor for the miR-520d-5p controlled gene into the cell, or (b) a cell into which the expression or function inhibitor has been introduced.
  • a step of culturing or proliferating may be included.
  • “dedifferentiation” includes changing a cell into a cell with a lower differentiation state. For example, if a cell that has undergone differentiation has changed to a pluripotent stem cell, it can be said that it has been dedifferentiated.
  • the “dedifferentiation inducer” is a substance having an action of bringing a cell closer to a cell having a lower differentiation state.
  • the “pluripotent stem cell inducer” is a substance having an action of bringing a cell closer to a pluripotent cell such as a pluripotent stem cell.
  • “reprogramming” refers to the action of bringing a cell closer to a pluripotent cell such as a pluripotent stem cell.
  • a “pluripotent stem cell” is a pluripotent stem cell.
  • the pluripotent stem cell includes, for example, a cell expressing any undifferentiated marker at the same level or higher than hiPSC (HPS0002S253G1) which is a human induced pluripotent stem cell. Artificially produced pluripotent stem cells are sometimes referred to as iPS cells.
  • the pluripotent stem cell obtained by the production method of the above-mentioned embodiment may highly express p53.
  • This p53 expression level is significantly higher than the p53 expression level of, for example, control samples (eg, hiPSC (HPS0002 253G1), p53 knockout cells, normal cells, or samples derived therefrom). It is preferable.
  • the expression level of p53 is, for example, 1.05, 1.1, 1.2, 1.4, 1.6, 1.8, 2.0, 3.0, 4.0, 5.0, 10, 100, or 1000 times or more that of the control sample. May be.
  • the method for measuring the expression level of p53 is preferably real-time PCR in terms of measurement accuracy and simplicity.
  • a protruding structure may be generated around the cell.
  • the “cell” used for dedifferentiation, pluripotent stem cell formation and the like may be a somatic cell.
  • somatic cells are other cells excluding germ cells, and include skin-derived cells or fibroblasts. Somatic cells usually have limited or disappeared pluripotency.
  • the cell may be a malignant tumor cell.
  • Malignant tumors include, for example, tumors that arise when normal cells are mutated. Malignant tumors can arise from all organs and tissues throughout the body, and when malignant tumor cells proliferate, they are known to clump together and invade and destroy surrounding normal tissues.
  • This malignant tumor is, for example, carcinoma, sarcoma, hematological malignancy, lung cancer, esophageal cancer, stomach cancer, liver cancer, pancreatic cancer, kidney cancer, adrenal cancer, biliary tract cancer, breast cancer, colon cancer, small intestine cancer, cervical cancer, uterus Body cancer, ovarian cancer, bladder cancer, prostate cancer, ureteral cancer, renal pelvic cancer, ureteral cancer, penile cancer, testicular cancer, brain tumor, cancer of central nervous system, cancer of peripheral nervous system, head and neck cancer (oral cancer, Pharyngeal cancer, laryngeal cancer, nasal cavity / sinus cancer, salivary gland cancer, thyroid cancer, etc.), glioma, glioblastoma multiforme, skin cancer, melanoma, thyroid cancer, salivary gland cancer, or malignant lymphoma.
  • the “undifferentiation marker” is a general term for compounds such as DNA strands, RNA strands or proteins that are specifically expressed in undifferentiated cells. Examples include Klf4, c-Myc, Oct4, Sox2, PROM1, Nanog, SSEA-1, ALP, eRas, Esg1, Ecat1, Fgf4, Gdf3, or REX-1. Sometimes referred to as a pluripotent stem cell marker.
  • endogenous means that the test substance is derived from the internal mechanism of the cell.
  • a protein that is constitutively expressed in a cell is endogenously contained only in the expressed protein.
  • “inhibiting gene expression” includes, for example, inhibiting a transcription mechanism from a gene to mRNA, or inhibiting a translation mechanism from mRNA to protein or polypeptide. In addition, for example, it includes inhibiting by inducing the degradation of a gene, mRNA, or protein.
  • the role of a gene includes, for example, producing mRNA derived from the gene, producing a protein derived from the gene, and causing the protein derived from the gene to exhibit activity. Therefore, in one embodiment of the present invention, “inhibiting gene function” includes a decrease in the amount of mRNA or protein produced as a result of inhibiting gene expression.
  • “inhibiting the function of a gene” includes reducing the activity of mRNA derived from the gene or protein derived from the gene.
  • the “state in which expression is inhibited” includes a state in which the expression level is significantly reduced as compared with the normal state.
  • the above “significantly reduced” may be, for example, a state where the expression level is reduced to 0.35, 0.3, 0.2, 0.1, 0.05, 0.01, or 0 times or less, and the range of any two values thereof It may be in a state of decreasing to the inside. From the viewpoint of causing cell dedifferentiation more strongly or stably, it is preferably 0.2 times or less, more preferably 0.1 times or less.
  • the expression level may be determined using the mRNA level or protein level as an index.
  • “significantly” may include, for example, a case where statistical significance is evaluated using Student's t test (one-sided or two-sided) and p ⁇ 0.05. Or the state in which the difference has arisen substantially is included.
  • the inhibitory strength in the “state in which the function is inhibited” is the same as in the embodiment of the inhibitory strength of expression inhibition.
  • the “form of the inhibitor” is not particularly limited, and may be, for example, an RNA chain, a DNA chain, a polynucleotide, a low molecular organic compound, an antibody, or a polypeptide.
  • RNA strand for example, an RNAi molecule against mRNA of a miR-520d-5p controlled gene can be used.
  • DNA strand or polynucleotide a DNA strand or polynucleotide encoding the RNA strand can be used.
  • the form of this DNA strand or polynucleotide may be, for example, a vector.
  • the low molecular weight organic compound may be obtained by utilizing combinatorial chemistry or HTS (High Throughput Screening).
  • HTS High Throughput Screening
  • an automatic synthesizer L-COS series (Shoko Scientific Co., Ltd.) may be used.
  • an Octet system (ForteBio) may be used for HTS.
  • the antibody is, for example, an antibody against a miR-520d-5p controlled protein.
  • the above antibody may be produced by a known antibody production method (for example, the method described in Clackson et al., Nature. 1991 Aug 15; 352 (6336): 624-628.), Or a contract company (for example, EVEC, Inc.).
  • the polypeptide can be purchased from a trust company (for example, Wako Pure Chemical Industries, Ltd.).
  • the inhibitor includes a substance that inhibits the expression of the subject or a substance that inhibits the function.
  • the inhibitor is preferably a substance having low cytotoxicity or substantially no cytotoxicity.
  • side effects can be suppressed when the inhibitor is administered in vivo.
  • the inhibitor is preferably an RNAi molecule or a polynucleotide encoding the RNAi molecule.
  • RNAi molecules against ELAVL2651mRNA correspond to positions 631 to 651, 721 to 741, 1158 to 1178, or 1281 to 1299 of SEQ ID NO: 29.
  • RNAi molecules that target RNA sequences are preferred.
  • RNAi molecules against TEAD1699 mRNA correspond to positions 681 to 699, 1021 to 1038, 1349 to 1367, or 1429 to 1447 of SEQ ID NO: 30.
  • RNAi molecules that target RNA sequences are preferred.
  • the RNAi molecule for GATAD2B mRNA has an RNA sequence corresponding to positions 368 to 286, 854 to 872, 1009 to 1027, or 1166 to 1184 of SEQ ID NO: 31.
  • the target RNAi molecule is preferred.
  • the target includes being capable of being combined.
  • the “RNAi molecule” is an RNA strand having an RNAi action, and examples thereof include siRNA, shRNA, miRNA, and small RNA having an RNAi action.
  • RNAi is a function of a target gene, mRNA, or the like by one or more of siRNA, shRNA, miRNA, short or long one or multiple strand RNA, or a modification thereof. Including the phenomenon that is suppressed or silenced. In general, the suppression mechanism by RNAi is sequence-specific and exists in various biological species. The mechanism of RNAi in a typical mammal when siRNA or shRNA is used is as follows. First, a vector capable of expressing siRNA or shRNA is introduced into cells. Then, after siRNA or shRNA is expressed in the cell, siRNA or shRNA becomes single-stranded, and then RISC (RNA-induced Silencing Complex) is formed.
  • RISC RNA-induced Silencing Complex
  • RISC uses the incorporated single-stranded RNA as a guide molecule and recognizes a target RNA strand having a sequence highly complementary to this single-stranded RNA.
  • the target RNA strand is cleaved by an enzyme such as AGO2 in RISC. Thereafter, the cleaved target RNA strand is degraded.
  • a plurality of RNAi molecules may be introduced into a cell at the same time, for example, 1, 2, 3, 4, 5, 6, or 10 may be introduced, and any one of these two values is included. Numbers may be introduced.
  • the RNAi molecule is preferably single-stranded or double-stranded from the viewpoint of more stably suppressing the function of the target gene or mRNA.
  • RNAi molecules Stealth RNAi designer (Invitrogen), siDirect 2.0 (Naito et al., BMC Bioinformatics. 2009 2009 Nov 30; 10: 392.), Etc. can be used. Further, it may be entrusted to a trust company (for example, Thermo Scientific).
  • the RNAi action can be confirmed by quantifying the expression level of the RNA strand by real-time RT-PCR. Alternatively, it can also be performed by methods such as analysis of RNA strand expression level by Northern blot, analysis of protein amount by Western blot, and observation of phenotype. In particular, the method using real-time RT-PCR is efficient.
  • siRNA includes an RNA strand capable of inducing RNAi.
  • the duplex of siRNA can be divided into a guide strand and a passenger strand, and the guide strand is incorporated into RISC.
  • the guide strand incorporated into RISC is used to recognize the target RNA.
  • Artificially produced RNAi research is mainly used in RNAi research, but some that exist endogenously in the living body are also known.
  • the guide strand may be composed of RNA having 15 or more bases. If it is 15 bases or more, the possibility of binding to the target polynucleotide with high accuracy increases.
  • the guide strand may be composed of RNA having 40 bases or less. If it is 40 bases or less, the risk that disadvantageous phenomena such as interferon response occur will be lower.
  • shRNA includes a single-stranded RNA strand that can induce RNAi and can form a hairpin-like structure (hairpin-like structure).
  • shRNA is cleaved by Dicer in the cell, and siRNA is excised. It is known that target RNA is cleaved by this siRNA.
  • the shRNA may be composed of 35 or more nucleotides. If it is 35 or more, the possibility that a hairpin-like structure peculiar to shRNA can be formed with high accuracy increases.
  • the shRNA may be composed of RNA of 100 bases or less. If it is 100 bases or less, the risk that disadvantageous phenomena such as interferon response occur will be reduced.
  • the length of shRNA is not necessarily 100 bases or less. However, it is thought that it can function as shRNA.
  • miRNA includes an RNA strand having a function similar to that of siRNA, and is known to suppress or degrade the translation of a target RNA strand.
  • the difference between miRNA and siRNA generally lies in the production pathway and detailed mechanism.
  • small RNA refers to a relatively small RNA strand, and examples thereof include siRNA, shRNA, miRNA, antisense RNA, and single- or multiple-stranded small RNA. , But not limited to them. When small RNA is used, disadvantageous phenomena such as interferon response can be suppressed.
  • the RNA strand may contain an overhang consisting of 1 to 5 bases at the 5 ′ end or 3 ′ end. In this case, it is considered that the efficiency of RNAi increases. This number may be, for example, 5, 4, 3, 2, or 1 base, and may be within the range of any two of them.
  • the overhang can be, for example, ac, c, uc, ag, aa, or uu. From the viewpoint of stably exhibiting RNAi action, the overhang is preferably 3'-terminal ac, c, or uc.
  • the number may be, for example, 1, 2, 3, 4, 5, or 10 or less, and may be in the range of any two of them.
  • the RNA strand may include a hairpin loop, and the number of bases of the hairpin loop may be, for example, 10, 8, 6, 5, 4, or 3 bases, and any two values thereof. It may be within the range.
  • the base sequence of the hairpin loop may be, for example, gugcuc or cucuuga. As long as this base sequence has a desired effect, one or a plurality of base sequences may be deleted, substituted, inserted, or added. In addition, the notation of each base sequence is the 5 ′ end on the left side and the 3 ′ end on the right side.
  • the length of the RNA strand is, for example, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34. , 40, 50, 60, 80, 100, 200, or 500 bases, or any of these two values. If this number is 15 or more, the possibility of being able to bind to the target polynucleotide with high accuracy increases. Further, if this number is 100 or less, the risk of adverse phenomena such as interferon response occurring when RNA strands are administered in vivo is reduced. Interferon response is generally known as a phenomenon in which cells enter an antiviral state by sensing dsRNA.
  • the “polynucleotide” includes those in which a plurality of nucleotides, nucleobases, or their equivalents are combined.
  • the polynucleotide includes a DNA strand or an RNA strand.
  • the “RNA strand” includes those in which a plurality of RNAs or equivalents thereof are combined.
  • the “DNA strand” includes those in which a plurality of DNAs or their equivalents are combined.
  • This RNA strand or DNA strand includes an RNA strand or a DNA strand in the form of a single strand or a plurality of strands (for example, a double strand).
  • the RNA strand or DNA strand may be a cell uptake promoting substance (eg, PEG or a derivative thereof), a label tag (eg, a fluorescent label tag), a linker (eg, a nucleotide linker), or a chemotherapeutic agent (eg, an anti-malignant).
  • a tumor substance or the like The RNA strand or DNA strand can be synthesized using a nucleic acid synthesizer. In addition, it can also be purchased from a trust company (for example, Invitrogen). In vivo RNA strands or DNA strands may form salts or solvates. In addition, RNA strands or DNA strands in vivo may be subjected to chemical modification.
  • RNA strand or DNA strand includes, for example, an RNA strand or DNA strand that forms a salt or solvate, or an RNA strand or DNA strand that has undergone chemical modification.
  • the RNA strand or DNA strand may be an RNA strand analog or a DNA strand analog.
  • the “salt” is not particularly limited, but includes, for example, an anion salt formed with any acidic (eg, carboxyl) group, or a cation salt formed with any basic (eg, amino) group.
  • the salts include inorganic salts or organic salts, for example, salts described in “Berge” et al., “J. Pharm. Sci.,” 1977, 66, 1-19.
  • the “solvate” is a compound formed by a solute and a solvent.
  • the solvent is water, the solvate formed is a hydrate.
  • This solvent is preferably one that does not interfere with the biological activity of the solute. Examples of such preferred solvents include, but are not limited to, water or various buffers.
  • the “chemical modification” include modification with PEG or a derivative thereof, fluorescein modification, or biotin modification.
  • the RNA strand preferably includes a base sequence complementary to a part of the base sequence of mRNA derived from the target gene.
  • the “part” is, for example, 5, 10, 15, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, or 50 bases or more. It may be within the range of any two of these values.
  • a plasmid that generates siRNA or shRNA for a specific gene can be purchased from, for example, a trust company (for example, Thermo Scientific, GeneCopoeia, etc.).
  • the nucleotide sequences of the four plasmids capable of generating ELAVL2 siRNA used in Examples described later are SEQ ID NO: 25, 26, 27, or 28.
  • the eight plasmids capable of generating TEAD1 siRNA or GATAD2B siRNA used in Examples described later are those in which DNA sequences encoding TEAD1 shRNA or GATAD2B shRNA are mounted on the psiLv-U6 TM vector.
  • shRNAs containing the nucleotide sequences of SEQ ID NOs: 5, 6, 7, or 8 are respectively generated from the four plasmids that can generate ELAVL2 siRNA used in Examples described later. These shRNAs are thought to be cleaved by enzymes in cells to generate siRNA. These siRNAs contain the nucleotide sequences of SEQ ID NO: 1 (uuauugguguuaaagucacgg), 2 (aauacgagaaguaauaaugcg), 3 (uuguuugucuuaaggag), or 4 (auuugcaucucugauagaagc), respectively.
  • This base sequence of SEQ ID NO: 1, 2, 3, or 4 is a base sequence that is complementary to a part of ELAVL2 mRNA, and is considered to be a part that functions as a guide strand.
  • shRNAs containing the nucleotide sequence of SEQ ID NO: 13, 14, 15, or 16 are generated from the four plasmids that can generate TEAD1 siRNA used in the examples described later. These shRNAs are thought to be cleaved by enzymes in cells to generate siRNA. These siRNAs each contain the nucleotide sequence of SEQ ID NO: 9 (uuggcuuaucugcagaguc), 10 (gcuuguuaugaauggcag), 11 (guaagaaugguuggcaugc), or 12 (aguuccuuuaagccaccuu).
  • the base sequence of SEQ ID NO: 9, 10, 11, or 12 is a base sequence complementary to a part of TEAD1EAD mRNA, and is considered to be a part that functions as a guide strand.
  • shRNAs containing the nucleotide sequence of SEQ ID NO: 21, 22, 23, or 24 are generated from the four plasmids that can generate GATAD2B siRNA used in the examples described later. These shRNAs are thought to be cleaved by enzymes in cells to generate siRNA. These siRNAs each contain the nucleotide sequence of SEQ ID NO: 17 (caacagauucaagcgaaga), 18 (caauagaugcugcauucug), 19 (caucaacauguguggaagg), or 20 (aggauguuguacgcugaca).
  • the base sequence of SEQ ID NO: 17, 18, 19, or 20 is a base sequence that is complementary to a part of GATAD2B mRNA, and is considered to be a part that functions as a guide strand.
  • ELAVL2 siRNA is a base sequence complementary to the base sequence of SEQ ID NO: 1, 2, 3, or 4 (for example, the base sequence at positions 1 to 21 of the base sequence of SEQ ID NO: 5, the sequence The base sequence at positions 1-21 of the base sequence of No. 6, the base sequence at positions 1-19 of the base sequence of SEQ ID NO: 7, or the base sequence of positions 1-21 of the base sequence of SEQ ID NO: 8) Good.
  • TEAD1 siRNA is a base sequence complementary to the base sequence of SEQ ID NO: 9, 10, 11, or 12 (for example, the base sequence at positions 1-19 of the base sequence of SEQ ID NO: 13, sequence The base sequence at positions 1-18 of the base sequence of number 14, the base sequence at positions 1-19 of the base sequence of sequence number 15, or the base sequence at positions 1-19 of the base sequence of sequence number 16) Good.
  • GATAD2B siRNA is a nucleotide sequence complementary to the nucleotide sequence of SEQ ID NO: 17, 18, 19, or 20 (for example, 1- 1 of the nucleotide sequence of SEQ ID NO: 21, 22, 23, or 24). 19-base sequence).
  • the base sequences represented by SEQ ID NOs: 1 to 31 have a desired effect, (c) one or a plurality of base sequences is deleted or substituted in any base sequence, Inserted or added base sequence, (d) a base sequence having 90% or more homology with any base sequence, (e) a polymorphic base sequence complementary to any base sequence It may be one or more base sequences selected from the group consisting of a base sequence encoded by a polynucleotide that specifically hybridizes to a nucleotide under stringent conditions.
  • the “complementary base sequence” is a base sequence possessed by another highly complementary polynucleotide capable of hybridizing to one polynucleotide.
  • Hybridization refers to the property that a base pair can be formed by hydrogen bonding between bases between a plurality of polynucleotides. Base pairs can occur in Watson-Crick base pairs, Hoogsteen base pairs, or any other sequence specific form. A state in which two single strands are hybridized is called a double strand.
  • the “plurality” may be, for example, 10, 8, 6, 4, or 2, or may be less than any of these values. From the viewpoint of causing cell dedifferentiation more strongly or stably, the smaller the number, the better. It is known to those skilled in the art that an RNA strand that has undergone deletion, addition, insertion, or substitution of one or more bases maintains its biological activity.
  • the above “90% or more” may be, for example, 90, 95, 97, 98, 99, or 100% or more, and may be in the range of any two of them. From the viewpoint of causing cell dedifferentiation more powerfully or stably, a larger number is preferable.
  • the above-mentioned “homology” may be calculated according to a method known in the art in the ratio of bases that are homologous in two or more base sequences. Before calculating the ratio, the bases of the base sequence groups to be compared are aligned, and a gap is introduced into a part of the base sequence if necessary to maximize the ratio of the same bases.
  • the following conditions can be adopted as the “stringent conditions”.
  • Use low ionic strength and high temperature for washing e.g., 0.015M sodium chloride / 0.0015M sodium citrate / 0.1% sodium dodecyl sulfate at 50 ° C
  • a denaturing agent such as formamide (for example, at 42 ° C., 50% (v / v) formamide and 0.1% bovine serum albumin / 0.1% ficoll / 0.1% polyvinylpyrrolidone / 50 mM sodium phosphate buffer pH 6.5, And 750 mM sodium chloride, 75 mM sodium citrate), or (3) 20% formamide, 5 ⁇ SSC, 50 mM sodium phosphate (pH 7.6), 5 ⁇ Denhardt's solution, 10% dextran sulfate, and 20 mg / ml denaturation Incubate overnight at 37 ° C.
  • the introduction of the inhibitor into cells and the method for culturing the cells can be performed according to methods known in the art.
  • introduction into cells for example, infection introduction using a viral vector, calcium phosphate method, lipofection method, electroporation method, or microinjection can be used.
  • only introduced cells can be selected using drug resistance, cell sorter or the like.
  • the medium for example, ReproStem, a medium for primate ES cells (Cosmo Bio), a medium for maintaining undifferentiation, a medium for normal human cells (for example, a medium based on DMEM or RPMI), and the like can be used.
  • the cells may be cultured in a ReproStem under conditions of 37 ° C., 5% CO 2 and 10% FBS.
  • This numerical value may be moved back and forth within a range of, for example, plus or minus 10, 20, or 30%. It may be established or cultured in the absence of feeder cells.
  • the number of days of culture when establishing pluripotent stem cells is not particularly limited, and may be, for example, 1, 2, 3, 4, 5, 6, 8, 10, 15, 30, or 40 days or more, It may be within the range of any two of them.
  • the cancer cells to be dedifferentiated are preferably undifferentiated cancer cells.
  • a plurality of RNA chains may be introduced into the cell.
  • the DNA strand when the DNA strand is introduced into a cell, a plurality of DNA strands may be introduced into the cell.
  • the number of “plural RNA strands” or “plurality of DNA strands” may be, for example, 2, 3, 4, 5, 6, 10, 12, 16, or 20 or more, It may be within the range of any two values.
  • the “cell population” may be a substantially uniform cell population.
  • the “inhibitor” may be an RNA strand that binds to one or more 3′UTRs such as ELAVL2 and inhibits the transcription mechanism, but excludes miR-520d-5p.
  • the act of inhibiting using miR-520d-5p is excluded.
  • the “vector” refers to a viral (eg, lentivirus, adenovirus, retrovirus, or HIV) vector, a plasmid derived from E. coli (eg, pBR322), a plasmid derived from Bacillus subtilis (eg, pUB110).
  • a viral vector eg, lentivirus, adenovirus, retrovirus, or HIV
  • a plasmid derived from E. coli eg, pBR322
  • Bacillus subtilis eg, pUB110
  • the vector may contain components necessary for the expression of a DNA strand, such as a promoter, a replication origin, or an antibiotic resistance gene.
  • the vector may be a so-called expression vector.
  • the “cell population” is a population including a plurality of cells.
  • This cell population may be, for example, a population containing substantially uniform cells.
  • the cell population may also be a cell preparation.
  • a cell preparation may contain, for example, cells and buffer or media components.
  • the pluripotent stem cell population may contain pluripotent stem cells, for example, 80, 90, 95, 96, 97, 98, 99, or 100% or more, and any one of these two values. May be.
  • the “therapeutic agent for malignant tumor” may further include DDS (Drug-Delivery System).
  • DDS Drug-Delivery System
  • the polynucleotide can be more efficiently introduced into the cell.
  • DDS include gelatin hydrogel and atelocollagen.
  • “treatment” refers to the ability to exert a symptom improving effect or a preventive effect on one or more symptoms associated with a patient's disease or disease.
  • a “therapeutic agent” may be a pharmaceutical composition comprising one or more pharmacologically acceptable carriers.
  • the pharmaceutical composition can be produced by any method known in the technical field of pharmaceutics, for example, by mixing an active ingredient (for example, an inhibitor against one or more of ELAVL2 and the like) and the carrier.
  • the form of use of the therapeutic agent is not limited as long as it is a substance used for treatment, and it may be an active ingredient alone or a mixture of an active ingredient and an arbitrary ingredient.
  • the shape of the carrier is not particularly limited, and may be, for example, a solid or a liquid (for example, a buffer solution).
  • the therapeutic agent of a malignant tumor contains the drug (preventive agent) used for the prevention of a malignant tumor, the benign improvement inducer of normal malignant tumor, or a normal stem cell induction agent.
  • the administration route of the therapeutic agent is preferably one that is effective in the treatment, and may be, for example, intravenous, subcutaneous, intramuscular, intraperitoneal, or oral administration.
  • the administration form may be, for example, an injection, capsule, tablet, granule or the like.
  • An aqueous solution for injection may be stored in, for example, a vial or a stainless steel container.
  • the aqueous solution for injection may contain, for example, physiological saline, sugar (for example, trehalose), NaCl, or NaOH.
  • the therapeutic agent may contain, for example, a buffer (for example, phosphate buffer), a stabilizer and the like.
  • the dose is not particularly limited, but may be, for example, 0.0001 mg to 1000 mg / kg body weight per dose.
  • the dosing interval is not particularly limited, but may be administered once every 1 to 10 days, for example.
  • the dose, administration interval, and administration method may be appropriately selected depending on the age, weight, symptoms, target organ, etc. of the patient. It may also be administered in combination with other appropriate chemotherapeutic drugs.
  • the therapeutic agent preferably contains a therapeutically effective amount or an effective amount of an active ingredient that exhibits a desired action. If the malignant tumor marker decreases after administration, it may be determined that there was a therapeutic effect.
  • This therapeutic agent may be reprogrammed in vivo and further differentiated in vivo.
  • the characteristics of malignant tumor cells may be changed in vivo and assimilated into surrounding tissues.
  • malignant tumor cells may be changed to non-malignant tumor cells in vivo.
  • a “patient” is a human or non-human mammal (eg, mouse, guinea pig, hamster, rat, mouse, rabbit, pig, sheep, goat, cow, horse, cat, dog, marmoset. , Monkey or chimpanzee).
  • a human or non-human mammal eg, mouse, guinea pig, hamster, rat, mouse, rabbit, pig, sheep, goat, cow, horse, cat, dog, marmoset. , Monkey or chimpanzee.
  • an embodiment of the present invention includes a method for inducing dedifferentiation, a method for inducing pluripotent stem cells, or a treatment for malignant tumor, which comprises a step of inhibiting the activity of an RNA chain or protein derived from a miR-520d-5p-controlled gene. Including methods.
  • One embodiment of the present invention also includes a method for producing pluripotent stem cells, which comprises the step of inhibiting the activity of an RNA chain or protein derived from a miR-520d-5p-controlled gene.
  • a method for producing pluripotent stem cells which comprises the step of inhibiting the activity of an RNA chain or protein derived from a miR-520d-5p-controlled gene.
  • one embodiment of the present invention provides a dedifferentiation inducer, a pluripotent stem cell inducer, a malignant tumor therapeutic agent, or the like containing an activity inhibitor of an RNA chain or protein derived from a miR-520d-5p-controlled gene. Including.
  • ELAVL2 is a gene encoding an RNA binding protein according to a summary published by NCBI. The present inventor discovered that when the expression of this gene was inhibited, the cells became pluripotent stem cells. Specific experimental procedures and results are described below.
  • ELAVL2 siRNA expressing siRNA against ELAVL2 mRNA were purchased from Thermo Scientific (B-9 (SEQ ID NO: 25), B-10 (sequence) No. 26), B-11 (SEQ ID NO: 27), B-12 (SEQ ID NO: 28)). These plasmids are DNAs encoding ELAVL2 siRNA incorporated into the pLKO.1 vector. Each of these plasmids was introduced into 293FT cells (Clontech, human embryonic kidney cell line). Thereafter, virus particles released to the culture supernatant were purified by ultracentrifugation (27000 rpm) and stored at -80 degrees.
  • ELAVL2 siRNAs four types of ELAVL2 siRNA (hereinafter also referred to as “ELAVL2 siRNAs”) were introduced into HLF cells by co-introduction.
  • This ELAVL2 siRNA s has a base sequence complementary to ELAVL2 mRNA (SEQ ID NOs: 1, 2, 3, and 4 respectively).
  • FIG. 1 shows the result of observation with an inverted microscope manufactured by Olympus.
  • the ELAVL2 siRNAs-introduced HLF cells obtained by the above procedure are hereinafter referred to as siELAVL2-HLF.
  • siELAVL2-HLF Five days later, siELAVL2-HLF grew and proliferated little by little in places where it depended on the scaffold, unlike cancer cells.
  • radially-like protrusion-like structures were observed from the spherical cell population over the entire circumference (FIG. 1, bottom x40).
  • ELAVL2 siRNAs were introduced into well-differentiated cancer cells, after introduction of ELAVL2 siRNAs, normal undifferentiated cells were induced in vivo via moderately differentiated cancer, poorly differentiated cancer, and undifferentiated cancer. Yes.
  • ELAVL2 siRNAs were introduced into 293FT cells instead of the above HLF cells, iPS cell-like spherical cells were observed.
  • HLF into which the plasmids B-9, B-10, B-11, and B-12 were respectively introduced was prepared, and the expression suppression rate of ELAVL2 was examined by RT-PCR. The results are as shown in FIG. 3, and in all cases, the expression of ELAVL2 was suppressed.
  • siELAVL2-HLF was cultured separately when RPMI 1640 (WAKO) was used as the culture medium and when undifferentiated maintenance culture medium ReproStem (ReproCELL) was used.
  • RPMI 1640 WAKO
  • ReproCELL undifferentiated maintenance culture medium
  • siELAVL2-HLF was not a heterogeneous cell population peculiar to cancer cells, but had a special growth direction similar to normal cells toward a homogeneous growth direction.
  • the S period increased and the GoG1 period decreased. Since siELAVL2-HLF has few apoptotic cells unlike cancer cells, it can be considered that ELAVL2 siRNA is different from conventional anticancer components.
  • siELAVL2-HLF Growth form siELAVL2-HLF and mock-introduced HLF were cultured in ReproStem for 14 days and then observed with a microscope. The results are shown in FIG. siELAVL2-HLF proliferated while spreading so as to be connected to the protrusions radiating from the spherical cells.
  • the upper row shows the growth form of mock-HLF and the lower row shows the growth form of siELAVL2-HLF.
  • siELAVL2-HLF was confirmed to be stained by AP staining (alkaline phosphatase staining) (FIG. 7).
  • AP is an undifferentiated cell marker. Therefore, it was suggested that the cell population in FIG. 6 is an undifferentiated cell population.
  • Transplantation siELAVL2-HLF was cultured in ReproStem for 1 week and then implanted subcutaneously in immunodeficient mice (KSN / Slc, Charles River). The transplanted tissue was observed 2 months after transplantation (FIG. 8). As a result, all the cases were non-tumorous.
  • the upper photograph is an appearance of a mouse
  • the middle photograph is a photograph when the liver tissue is observed after incision of the abdomen
  • the lower photograph is an enlarged photograph of the liver tissue. Scars were seen in the transplanted tissue. This becomes a trace in the transplanted living body after transplantation.
  • ELAVL2 reporter assay The present inventor has confirmed that miR-520d-5p binds to the 3 'UTR (SEQ ID NO: 10) of ELAVL2 mRNA (SEQ ID NO: 9), and the expression of ELAVL2 can be inhibited. , Found in the luciferase reporter assay. Specific experimental procedures and results are described below.
  • Synthetic miR-520d-5p (consigned to IDT) and pMIR-520d-5p expression lentivirus were co-introduced into HLF cells, and assayed with the Promega psiCHECK2 reporter vector.
  • the expression level (RLU) of luciferin was measured with a microplate reader (TECAN), and the value corrected by dividing lenilla (RLU) with firefly (RLU) was used as a control (synthetic miR-520d-3p (IDT) ) Or pLKO.1 lentiviral vector (Addgene)).
  • RLU expression level
  • miR-520d-5p or pMIR-520d-5p means that those genes were expressed. “-” Means not expressed.
  • binding site 1 or 2 means that ELAVL2 3′UTR is a base sequence having no mismatch. “-” Means that ELAVL2 3′UTR is a base sequence having a mismatch.
  • miR-520d-5p specifically binds to ELAVL2 3 'UTR region 1 (3'UTR: 508-525) and 2 (3'UTR: 880-894) and suppresses ELAVL2 expression. It means that it was done. This indicates that ELAVL2 3 ′ UTR is the target of miR-520d-5p.
  • Example 3 Production of pluripotent stem cells by inhibition of TEAD1
  • the present inventor used the database of DIANA-MICROT, miRDB, MicroRNA.org, and TargetScan-VERT, and the 3 ′ UTR of TEAD1 was miR-520d- We identified it as a target of 5p.
  • a method for producing pluripotent stem cells using the inhibitory effect of TEAD1 will be described.
  • a plasmid expressing siRNA against TEAD1 mRNA is purchased from GeneCopoeia. This plasmid is introduced into malignant tumor cells. Thereafter, pluripotent stem cells can be obtained by culturing for 5 days.
  • a spherical cell population can be observed.
  • tumor non-formation or differentiation can occur in the transplanted organ.
  • candidate gene candidates for miR-520d-5p are considered, including those that are not actually targeted.
  • target genes such as TEAD1 were identified using DIANA-MICROT, miRDB, MicroRNA.org, or TargetScan-VERT.
  • the miR- Genes were identified on the basis of the fact that there are many bases that match 520d-5p and excellent overall complementarity.
  • Example 4 Production of Pluripotent Stem Cells by Inhibition of GATAD2B
  • 3 ′ UTR of GATAD2B can be a target of miR-520d-5p using miRDB and TargetScan-VERT.
  • a method for producing pluripotent stem cells using the inhibitory effect of GATAD2B will be described.
  • a plasmid expressing siRNA or shRNA against GATAD2B mRNA is purchased from GeneCopoeia. This plasmid is introduced into malignant tumor cells.
  • pluripotent stem cells can be obtained by culturing for 7 days. When the cultured cells are observed with a microscope, a spherical cell population can be observed. When this cell population is transplanted into a mouse, tumor non-formation or differentiation can occur in the transplanted organ.
  • Example 5 Production of pluripotent stem cells by inhibition of SBF2, PUM2, or NBEA
  • the present inventor used MicroRNA.org and TargetScan-VERT, and SBF2, PUM2, and NBEA 3'UTR was miR- It was identified that it could be a target of 520d-5p.
  • a method for producing pluripotent stem cells using the inhibitory effect of SBF2, PUM2, or NBEA will be described.
  • a plasmid expressing siRNA or shRNA against SBF2 mRNA, PUM2 mRNA, or NBEA mRNA is purchased from Thermo Scientific. This plasmid is introduced into malignant tumor cells.
  • pluripotent stem cells can be obtained by culturing for 7 days.
  • a spherical cell population can be observed.
  • tumor non-formation or differentiation can occur in the transplanted organ.
  • ⁇ Example 7 Production of pluripotent stem cells by inhibition of each gene shown in Table 2
  • the present inventor can use miRDB to target the 3R UTR of each gene shown in Table 2 as miR-520d-5p It was identified.
  • a method for producing pluripotent stem cells using the inhibitory effect of each gene shown in Table 2 will be described.
  • a plasmid expressing siRNA or shRNA against mRNA derived from each gene shown in Table 2 is purchased from Thermo Scientific. This plasmid is introduced into malignant tumor cells.
  • pluripotent stem cells can be obtained by culturing for 7 days. When the cultured cells are observed with a microscope, a spherical cell population can be observed. When this cell population is transplanted into a mouse, tumor non-formation or differentiation can occur in the transplanted organ.
  • Example 8 Production of pluripotent stem cells by inhibition of each gene shown in Table 3
  • the present inventor used MicroRNA.org to target that the 3 'UTR of each gene shown in Table 3 is miR-520d-5p Identified that could be.
  • a method for producing pluripotent stem cells using the inhibitory effect of each gene shown in Table 3 will be described.
  • a plasmid expressing siRNA or shRNA against mRNA derived from each gene shown in Table 3 is purchased from Thermo Scientific. This plasmid is introduced into malignant tumor cells.
  • pluripotent stem cells can be obtained by culturing for 7 days. When the cultured cells are observed with a microscope, a spherical cell population can be observed. When this cell population is transplanted into a mouse, tumor non-formation or differentiation can occur in the transplanted organ.
  • plasmids that express siRNA or shRNA against mRNA derived from each gene shown in Table 4 are purchased from Thermo Scientific. This plasmid is introduced into malignant tumor cells. Thereafter, pluripotent stem cells can be obtained by culturing for 5 days. When the cultured cells are observed with a microscope, a spherical cell population can be observed. When this cell population is transplanted into a mouse, tumor non-formation or differentiation can occur in the transplanted organ.
  • Example 10 Evaluation of inhibitory effect of ELAVL2, TEAD1, and GATAD2B
  • plasmids expressing siRNA against TEAD1 mRNA and four types of plasmids expressing siRNA against GATAD2B mRNA were purchased from GeneCopoeia. These plasmids are obtained by incorporating DNA strands encoding TEAD1 siRNA or GATAD2B siRNA into a psiLv-U6 TM vector.
  • the TEAD1 siRNA has a base sequence complementary to TEAD1 mRNA (SEQ ID NOs: 9, 10, 11, 12 respectively).
  • the GATAD2B siRNA has a base sequence complementary to GATAD2B mRNA (SEQ ID NOs: 17, 18, 19, and 20 respectively).
  • the expression suppression rate of ELAVL2, TEAD1, and GATAD2B was examined by RT-PCR.
  • the results are as shown in FIGS. 10 to 12, and the expression of any gene was suppressed.
  • D is the number of culture days
  • R1 is a group of cells that have formed liver tissue or teratoma in about 1 month after in vivo
  • R2 is a group of cells that has not shown tumor formation after about 1 month in in vivo. I mean.
  • Example 11 Evaluation of DNA methylation level The DNA methylation level as an index of pluripotent stem cell formation was examined by the following procedure. In the same procedure as in Example 10, 4 types of ELAVL2 siRNA co-introduced HLF cells (siE), 4 types of TEAD1 siRNA co-introduced HLF cells (siT), and 4 types of GATAD2B siRNA were co-introduced. HLF cells (siG) were prepared.
  • ELAVL2 siRNA and 4 types of TEAD1 siRNA co-introduced HLF cells (siET), 4 types of ELAVL2 siRNA and 4 types of GATAD2B siRNA co-introduced HLF cells (siEG), 4 types of TEAD1 HLF cells (siTG) into which siRNA and 4 types of GATAD2B siRNA were co-introduced were prepared.
  • HLF cells (siETG) into which four types of ELAVL2 siRNA, four types of TEAD1 siRNA, and four types of GATAD2B siRNA were co-introduced were prepared.
  • HLF cells were changed to Huh7 cells (well-differentiated liver cancer cells), and the DNA methylation level was examined by the same procedure. The results are shown in FIG.
  • siETG was able to induce a demethylation level equivalent to hiPSC.
  • Huh7 cells each siRNA was introduced to induce a demethylation level equivalent to hiPSC.
  • HLF cells into which siETG had been introduced were transplanted subcutaneously or intraperitoneally into immunodeficient mice (KSN / Slc), no tumor was formed.
  • Example 12 Evaluation of tumorigenicity
  • HLF cells into which four types of ELAVL2 siRNA, four types of TEAD1 siRNA, and four types of GATAD2B siRNA were co-introduced were prepared. Two days after co-introduction, HLF cells (1 ⁇ 10 7 cells or more) were transplanted subcutaneously (12 mice) and intraperitoneally (12 mice) into immunodeficient mice. As a result, tumor formation ability was not observed in all cases in observations over 4 months.
  • HLF cells (1 ⁇ 10 7 cells or more) were transplanted subcutaneously (8 mice) and intraperitoneally (8 mice) into immunodeficient mice. Again, no tumor-forming ability was observed in all cases over 4 months of observation.
  • HLF cells into which four types of ELAVL2 siRNAs were co-introduced were prepared by the same procedure as in Example 11. Two days after co-introduction, HLF cells (1x10 7 cells or more) were transplanted subcutaneously into immunodeficient mice, but only 2 of 12 cases formed subcutaneous tumors, but not malignant tumors, such as "adipocytes and collagen fibers. It was a diagnosis of “non-undifferentiated tumor tissue having a differentiating tendency including normal cells”, and it was an unprecedented benign tumor difficult to make a clear diagnosis on the pathological tissue. In the other 10 cases, no tumor formation was observed after 4 months of observation.
  • Examples 1 to 12 above show that cells are reprogrammed into pluripotent stem cells by inhibiting the expression of one or more of the genes listed above.
  • reprogramming of malignant tumor cells is an area where little research results have been made worldwide, and it has new possibilities for the treatment of malignant tumors in the future.
  • the technique described in this example can be used for the production of pluripotent stem cells for research and regenerative medicine.
  • the pluripotent stem cells obtained in this example did not become malignant even after a long period of 4 months or longer after in vivo administration. Therefore, it can be said that the pluripotent stem cell obtained in the present Example is very excellent in safety.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Biophysics (AREA)
  • Transplantation (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Physics & Mathematics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Developmental Biology & Embryology (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Le but de la présente invention est d'obtenir : un nouveau procédé de production de cellules souches pluripotentes; ou un agent thérapeutique pour les tumeurs malignes. L'invention concerne ainsi l'utilisation d'un procédé destiné à induire la dédifférenciation d'une cellule, ledit procédé comprenant une étape consistant à inhiber l'expression ou la fonction d'un gène contrôlé par miR-520d-5p. L'invention concerne également l'utilisation d'un agent thérapeutique pour les tumeurs malignes, qui comprend un inhibiteur de l'expression ou de la fonction du gène contrôlé par miR-520d-5p.
PCT/JP2013/082399 2012-12-20 2013-12-02 Développement de cellules souches pluripotentes à l'aide d'un nouveau procédé d'induction d'une dédifférenciation WO2014097875A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2014553063A JPWO2014097875A1 (ja) 2012-12-20 2013-12-02 新規の脱分化誘導方法を用いた多能性幹細胞化

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2012278330 2012-12-20
JP2012-278330 2012-12-20

Publications (1)

Publication Number Publication Date
WO2014097875A1 true WO2014097875A1 (fr) 2014-06-26

Family

ID=50978210

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2013/082399 WO2014097875A1 (fr) 2012-12-20 2013-12-02 Développement de cellules souches pluripotentes à l'aide d'un nouveau procédé d'induction d'une dédifférenciation

Country Status (2)

Country Link
JP (1) JPWO2014097875A1 (fr)
WO (1) WO2014097875A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015030149A1 (fr) * 2013-08-29 2015-03-05 国立大学法人鳥取大学 Groupe biomoléculaire lié à l'anti-vieillissement cellulaire
WO2018203553A1 (fr) * 2017-05-02 2018-11-08 株式会社ペジィー・ファーマ Procédé pour améliorer la qualité de cellules différenciées
CN110129443A (zh) * 2019-05-21 2019-08-16 中国人民解放军***福州总医院 Fcho2基因在制备***放疗增敏药物的应用
WO2021251466A1 (fr) * 2020-06-11 2021-12-16 国立大学法人京都大学 Méthode efficace de production de cellules souches pluripotentes induites

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003007793A2 (fr) * 2001-07-17 2003-01-30 Yale University Compositions, methodes et trousses de traitement et diagnostic de l'hypertension
WO2004056386A2 (fr) * 2002-12-19 2004-07-08 To-Bbb Holding B.V. Acides nucleiques intervenant dans la regulation de la barriere hemato-encephalique
WO2008008472A2 (fr) * 2006-07-13 2008-01-17 President And Fellows Of Harvard College Méthodes et compositions pour moduler une formation de synapses
WO2009045443A2 (fr) * 2007-10-02 2009-04-09 The University Of Rochester Méthodes et compositions associées aux réponses synergiques aux mutations oncogènes
WO2010108126A2 (fr) * 2009-03-19 2010-09-23 Fate Therapeutics, Inc. Compositions de reprogrammation et procédés d'utilisation de celles-ci
JP2010536367A (ja) * 2007-08-24 2010-12-02 オンコセラピー・サイエンス株式会社 癌関連遺伝子、cdca5、epha7、stk31及びwdhd1
WO2011146879A2 (fr) * 2010-05-20 2011-11-24 University Of Rochester Procédés et compositions liés à la modulation de l'autophagie

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2076599A2 (fr) * 2006-09-19 2009-07-08 Asuragen, Inc. Gènes et voies régulés par mir-200 servant de cibles dans le cadre d'une intervention thérapeutique
CN102007223B (zh) * 2008-02-28 2014-06-18 俄亥俄州立大学研究基金会 用于胃癌的诊断、预后和治疗的基于微rna的方法和组合物

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003007793A2 (fr) * 2001-07-17 2003-01-30 Yale University Compositions, methodes et trousses de traitement et diagnostic de l'hypertension
WO2004056386A2 (fr) * 2002-12-19 2004-07-08 To-Bbb Holding B.V. Acides nucleiques intervenant dans la regulation de la barriere hemato-encephalique
WO2008008472A2 (fr) * 2006-07-13 2008-01-17 President And Fellows Of Harvard College Méthodes et compositions pour moduler une formation de synapses
JP2010536367A (ja) * 2007-08-24 2010-12-02 オンコセラピー・サイエンス株式会社 癌関連遺伝子、cdca5、epha7、stk31及びwdhd1
WO2009045443A2 (fr) * 2007-10-02 2009-04-09 The University Of Rochester Méthodes et compositions associées aux réponses synergiques aux mutations oncogènes
WO2010108126A2 (fr) * 2009-03-19 2010-09-23 Fate Therapeutics, Inc. Compositions de reprogrammation et procédés d'utilisation de celles-ci
WO2011146879A2 (fr) * 2010-05-20 2011-11-24 University Of Rochester Procédés et compositions liés à la modulation de l'autophagie

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
KNIGHT J.F. ET AL.: "TEAD1 and c-Cbl are novel prostate basal cell markers that correlate with poor clinical outcome in prostate cancer", BRITISH JOURNAL OF CANCER, vol. 99, no. 11, 2008, pages 1849 - 1858 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015030149A1 (fr) * 2013-08-29 2015-03-05 国立大学法人鳥取大学 Groupe biomoléculaire lié à l'anti-vieillissement cellulaire
US10265347B2 (en) 2013-08-29 2019-04-23 Norimasa Miura Biomolecular group related to cell anti-aging
WO2018203553A1 (fr) * 2017-05-02 2018-11-08 株式会社ペジィー・ファーマ Procédé pour améliorer la qualité de cellules différenciées
JPWO2018203553A1 (ja) * 2017-05-02 2020-03-19 株式会社ペジィー・ファーマ 分化細胞の品質改善方法
CN110129443A (zh) * 2019-05-21 2019-08-16 中国人民解放军***福州总医院 Fcho2基因在制备***放疗增敏药物的应用
CN110129443B (zh) * 2019-05-21 2022-07-15 中国人民解放军***福州总医院 Fcho2基因在制备***放疗增敏药物的应用
WO2021251466A1 (fr) * 2020-06-11 2021-12-16 国立大学法人京都大学 Méthode efficace de production de cellules souches pluripotentes induites

Also Published As

Publication number Publication date
JPWO2014097875A1 (ja) 2017-01-12

Similar Documents

Publication Publication Date Title
US9476041B2 (en) Method for producing novel hipsc by means of siRNA introduction
Natsume et al. Glioma-initiating cells and molecular pathology: implications for therapy
JP2011522514A (ja) Rna干渉を介する多能性遺伝子の誘発による細胞の再プログラミング
WO2015030149A1 (fr) Groupe biomoléculaire lié à l'anti-vieillissement cellulaire
US20100088775A1 (en) Methods of modulating epithelial-mesenchymal transition and mesenchymal-epithelial transition in cells and agents useful for the same
WO2014097875A1 (fr) Développement de cellules souches pluripotentes à l'aide d'un nouveau procédé d'induction d'une dédifférenciation
JP2022130675A (ja) メチル化関連酵素hat1とkat8の阻害薬
US20170137808A1 (en) Improved small interfering ribonucleic acid molecules
Hoffmann et al. Selectively expressed RNA molecules: a new dimension in functionalized cell targeting
EP3169770A1 (fr) Procédés de production de cellules souches pluripotentes induites
JP5781732B2 (ja) がん抑制miRNA
WO2017073689A1 (fr) Molécule d'arn pour la réparation de dommages à l'adn
CN109996878A (zh) 抗癌治疗干预法
Mukheed et al. Stem Cell Technology in Regenerative Medicines and Cancer Treatment: Towards Revolution in Clinical Success
Haase IGF2BP1 is the first positive marker for anaplastic thyroid carcinoma and an enhancer of a targetable gene expression signature
Jomaa Untangling Glioblastoma Invasion: Characterizing a Cell Culture Model of Glioblastoma Tumor Microtubes

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: 13866054

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: 2014553063

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: 13866054

Country of ref document: EP

Kind code of ref document: A1