[go: up one dir, main page]

WO2024185847A1 - Genetically modified mouse - Google Patents

Genetically modified mouse Download PDF

Info

Publication number
WO2024185847A1
WO2024185847A1 PCT/JP2024/008753 JP2024008753W WO2024185847A1 WO 2024185847 A1 WO2024185847 A1 WO 2024185847A1 JP 2024008753 W JP2024008753 W JP 2024008753W WO 2024185847 A1 WO2024185847 A1 WO 2024185847A1
Authority
WO
WIPO (PCT)
Prior art keywords
genetically modified
protein
mouse
cre recombinase
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/JP2024/008753
Other languages
French (fr)
Japanese (ja)
Inventor
一郎 谷内
将人 鐘巻
謙一郎 林
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Inter University Research Institute Corp Research Organization of Information and Systems
Kake Educational Institution
RIKEN
Original Assignee
Inter University Research Institute Corp Research Organization of Information and Systems
Kake Educational Institution
RIKEN
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 Inter University Research Institute Corp Research Organization of Information and Systems, Kake Educational Institution, RIKEN filed Critical Inter University Research Institute Corp Research Organization of Information and Systems
Publication of WO2024185847A1 publication Critical patent/WO2024185847A1/en
Anticipated expiration legal-status Critical
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material

Definitions

  • the present invention relates to genetically modified mice and methods for inducing the degradation of target proteins.
  • a representative example is the technology for producing knockout mice, which is based on gene targeting of the mouse ES cell genome using homologous recombination, and has made great progress in elucidating gene functions in vivo and the mechanisms of disease onset.
  • conditional knockout technology was developed, making it possible to knock out genes in specific tissues or cell types.
  • mice called flox mice
  • flox mice mice
  • a pair of loxP sequences recognized by Cre recombinase is introduced before and after the target gene region, and the target gene is destroyed in a tissue- or cell-type-specific manner by mating this flox mouse with a mouse that expresses Cre recombinase in a tissue- or cell-type-specific manner.
  • Conditional knockout technology has made it possible to study gene functions in adults even for genes that are lethal during development when knocked out throughout the body.
  • CreER recombinase which has the property of inducing recombination activity by drug administration, is used as the Cre recombinase, making it possible to perform tissue- or cell-type-specific and time-specific gene destruction.
  • conditional knockout technology is still unable to transiently suppress the function of the protein encoded by the manipulated gene with high temporal resolution.
  • CreER recombinase which can induce recombination activity by drug administration
  • the protein expressed from the gene before destruction remains for a certain period of time, so it takes a long time for the expression of the target protein to actually decrease and its activity to disappear.
  • the above problems are a major issue in drug discovery when selecting target proteins before developing a therapeutic drug.
  • the impact on biological functions when inhibiting the function of a candidate protein over the long term by destroying the candidate gene using whole-body knockout or conditional knockout does not necessarily match the impact when the protein's function is inhibited temporarily by administering the actual therapeutic drug to an animal, and sometimes unexpected side effects become apparent only after the actual therapeutic drug is administered. In such cases, the enormous amount of effort and investment required to develop the therapeutic drug is wasted, which is a major problem.
  • the objective of the present invention is to provide a new technology that enables tissue- or cell-type-specific control of protein function in vivo with high temporal resolution.
  • the auxin degron method is a technology that involves introducing the Transport Inhibitor Response 1 (TIR1) family protein, which constitutes an auxin-responsive ubiquitin ligase, into cells and controlling the degradation of target proteins fused with a degradation tag (called a degron) by adding the plant hormone auxin or an auxin analogue.
  • TIR1 Transport Inhibitor Response 1
  • the present inventors have generated Rosa26 OsTIR1(F74G) mice by introducing a construct in which a LoxP-STOP-LoxP (LSL) sequence is arranged upstream of a cDNA sequence encoding a rice-derived TIR1 protein having an F74G mutation (hereinafter referred to as "OsTIR1(F74G) protein") into the Rosa26 locus.
  • LSL LoxP-STOP-LoxP
  • OsTIR1(F74G) protein a construct in which a LoxP-STOP-LoxP (LSL) sequence is arranged upstream of a cDNA sequence encoding a rice-derived TIR1 protein having an F74G mutation (hereinafter referred to as "OsTIR1(F74G) protein") into the Rosa26 locus.
  • LSL LoxP-STOP-LoxP
  • OsTIR1(F74G) protein a LoxP-STOP-LoxP
  • the present inventors have found that administration of an auxin analog to the mice induced transient degradation of a degron-fused target protein very quickly, and the original expression level was then rapidly restored. Furthermore, administration of an auxin analogue to pregnant mice induced protein degradation in fetal mice, and administration of an auxin analogue to lactating female mice induced protein degradation in newborn mice, demonstrating that degradation of membrane proteins as target proteins can also be induced in vivo in mice.
  • the present invention is based on the above research results and provides the following.
  • a genetically modified mouse A pair of Cre recombinase recognition sequences, A transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences;
  • the genetically modified mouse comprises: a Transport Inhibitor Response 1 (TIR1) family gene sequence encoding a TIR1 family protein, which is located downstream of the pair of Cre recombinase recognition sequences; and a promoter sequence, which is located upstream of the pair of Cre recombinase recognition sequences and is capable of driving expression of the TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences.
  • TIR1 Transport Inhibitor Response 1
  • the genetically modified mouse described in (7), wherein the function of the gene encoding the target protein at the site where the site-specific promoter drives expression is essential for development and/or survival.
  • (11) A method for inducing degradation of a target protein in a mouse body, comprising: A degradation induction step of administering an auxin or an auxin analog to a genetically modified mouse to induce degradation of a target protein to which a degron sequence has been added,
  • the genetically modified mouse comprises a TIR1 family gene expression system, a gene encoding a target protein to which the degron sequence has been added, and a gene encoding a Cre recombinase;
  • the TIR1 family gene expression system comprises: A pair of Cre recombinase recognition sequences, A transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences; the method comprising: a TIR1 family gene sequence encoding a Transport Inhibitor Response 1 (TIR1) family protein, located downstream of the pair of Cre recombinase recognition sequences; and a promoter sequence located upstream
  • a method for inducing degradation of a target protein in a mouse-derived cell comprising the steps of: (10) A culturing step of culturing the cell according to (10); The method described above, further comprising a degradation inducing step of adding an auxin or an auxin analog to the cells to induce degradation of the target protein to which the degron sequence has been added.
  • This specification includes the disclosure of Japanese Patent Application No. 2023-036211, which is the priority basis of this application.
  • the present invention provides a genetically modified mouse that allows for tissue- or cell-type-specific control of protein function with high temporal resolution in vivo.
  • FIG. 13 shows that the loxP-neor-STOP-loxP sequence is removed by site-specific recombination of the loxP sequence by Cre recombinase, thereby inducing expression of the OsTIR1(F74G) protein.
  • FIG. 3A shows the crossbreeding for producing Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice.
  • FIG. 3B shows the results of flow cytometry analysis of the fluorescence intensity of Satb1-Venus in peripheral blood T cells after administration of 5-ph-IAA at a dose of 0.1 mg/body.
  • FIG. 3C shows the results of flow cytometry analysis of the fluorescence intensity of Satb1-Venus in peripheral blood T cells after administration of 5-ph-IAA at a dose of 0.02 mg/body.
  • FIG. 4 is a graph showing the results of FIG. 3B and FIG. 3C.
  • 5-ph-IAA was administered to pregnant or lactating female mice, and the fluorescence intensity of Satb1-Venus in T cells in the thymus excised from fetal or neonatal mice was analyzed by flow cytometry.
  • This figure shows the results of inducing protein degradation in vitro by targeting the membrane protein PD-1.
  • Figure 7 shows the results of inducing protein degradation in vivo in mice by targeting the membrane protein PD-1.
  • Figure 7A shows the crossbreeding for generating Rosa26 OsTIR1/+ :Pdcd1-mAID:Vav1Cre mice.
  • FIG. 7B shows the results of flow cytometry analysis of PD-1 protein expression in follicular T cells in Peyer's patches 12 hours after intraperitoneal administration of 5-ph-IAA or PBS.
  • FIG. 8 shows the results of inducing PD1 degradation in PD1 degron mice (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; Vavcre) expressing OsTIR1 in whole blood cells transplanted with MC38 colon cancer.
  • FIG. 8A shows an outline of the experiment.
  • FIG. 8B shows the results of measuring the expression level of PD1 in tumor-infiltrating cells (TIL).
  • TIL tumor-infiltrating cells
  • FIG. 8C shows the results of measuring the tumor diameter in the PBS-administered group and the 5-ph-IAA-administered group.
  • FIG. 9A shows the results of inducing PD1 degradation in PD1 degron mice (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; E8Icre mice) expressing OsTIR1 only in CD8 T cells transplanted with MC38 colon cancer.
  • FIG. 9A shows the results of measuring the tumor diameter in the PBS-administered group and the 5-ph-IAA-administered group of the PD1 degron mice.
  • FIG. 9B shows the results of comparing the tumor diameter when 5-ph-IAA was administered to PD1 degron mice expressing OsTIR1 only in CD8 T cells (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; E8Icre mice) and PD1 degron mice expressing OsTIR1 in all blood cells (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; Vavcre).
  • the first aspect of the present invention is a genetically modified mouse.
  • the genetically modified mouse of this aspect includes a gene expression system capable of inducing expression of a TIR1 family gene after recombination between a pair of Cre recombinase recognition sequences.
  • expression of a TIR1 family protein is induced by tissue- or cell-type-specific expression of Cre recombinase, and further, degradation of a target protein to which a degron sequence has been added can be transiently induced by administration of auxin or an auxin analog at any time point.
  • the mouse strain or genetic background of the "genetically modified mouse (transgenic mouse)" is not limited, and may be, for example, a C57BL/6N strain, a C57BL/6J strain, a BALB/C strain, a 129 strain, or the like, or may be a hybrid strain obtained by crossing a plurality of strains.
  • the genetically modified mouse may be, for example, a knock-in (KI) mouse, or a transgenic mouse other than a KI mouse (e.g., a Tg mouse in which a foreign gene sequence is introduced at a random position on the genome).
  • the method of producing the genetically modified mouse is not limited, and may be, for example, a genetically modified mouse produced by gene targeting based on conventional homologous recombination or a genome editing method using the CRISPR/Cas9 system.
  • the genetically modified mouse may be at any developmental stage or week/fetal age, and may be, for example, a fetal mouse, a perinatal mouse, a neonatal mouse, a preweaning mouse, a young adult mouse, or an adult mouse.
  • the genetically modified mouse may be either male or female.
  • the genetically modified mouse may be a disease model mouse, for example, a mouse that has developed a disease or may develop a disease due to genetic mutation, genetic abnormality, gene introduction, drug treatment, and/or surgical intervention, etc.
  • TIR1 family gene sequence refers to the base sequence or nucleic acid sequence of a gene encoding a TIR1 family protein.
  • type of TIR1 family gene sequence so long as it is a gene encoding a plant-derived TIR1 family protein.
  • type of plant from which it is derived and it may be derived, for example, from Arabidopsis thaliana, rice, zinnia, pine, fern, Physcomitrella patens, etc.
  • Specific examples of TIR1 family gene sequences include the base sequences of the TIR1 gene, AFB1 gene, AFB2 gene, AFB3 gene, FBX14 gene, and AFB5 gene.
  • the OsTIR1 gene which is a TIR1 family gene derived from rice, is preferred, and examples of such genes include the genes with NCBI accession numbers NM_001059194 (GeneID: 4335696), Os04g0395600, or accession numbers EAY93933 and OsI_15707, and more specifically, the OsTIR1 gene sequence consisting of the nucleotide sequence shown in SEQ ID NO: 2, which encodes the OsTIR1 protein consisting of the amino acid sequence shown in SEQ ID NO: 1.
  • the TIR1 family gene sequences such as the OsTIR1 gene sequence may also be codon-optimized for mouse cells, etc.
  • auxin degron system refers to a protein degradation control system that applies the plant protein degradation system based on the plant hormone auxin to host cells such as eukaryotic cells of non-plant origin.
  • this system is a system in which the above-mentioned TIR1 family protein guides a target protein labeled with a degron to degradation using the ubiquitin/proteasome degradation system of the host cell in a manner dependent on binding with auxin or an auxin analog.
  • auxin refers to a group of plant hormones that control plant growth.
  • auxin includes both naturally occurring auxins and non-natural auxins (synthetic auxins) that exhibit physiological actions similar to those of auxins.
  • Known naturally occurring auxins include indole-3-acetic acid (IAA) and 4-chloroindole-3-acetic acid (4Cl-IAA).
  • Known synthetic auxins include naphthaleneacetic acid, naphthoxyacetic acid, phenylacetic acid, 2,4-dichlorophenoxyacetic acid (2,4-D), and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T).
  • IAA indole-3-acetic acid
  • IAA one of the natural auxins
  • auxin analog is not particularly limited as long as it has a structure similar to any of the above-mentioned auxins, such as natural auxins such as indole-3-acetic acid (IAA) or 4-chloroindole-3-acetic acid (4Cl-IAA), and can bind to a TIR1 family protein to induce degradation of a target protein labeled with a degron.
  • auxin analogs are disclosed in WO 2021/009993, and may be, for example, a compound represented by the following general formula (II) or an ester thereof, or a compound represented by the following general formula (III).
  • R 1 is a cyclic aliphatic hydrocarbon group which may have a substituent and in which a portion of the carbon atoms constituting the ring may be substituted with a heteroatom, or an aromatic hydrocarbon group which may have a substituent and in which a portion of the carbon atoms constituting the ring may be substituted with a heteroatom.
  • R 2 represents a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, or a halogen atom
  • R 3 represents a hydrogen atom or an alkyl group having 1 to 6 carbon atoms.
  • R 2 is a hydrogen atom or an alkyl group having 1 to 6 carbon atoms
  • R 3 is an alkyl group having 1 to 6 carbon atoms.
  • auxin analog is 5-phenyl-indole-3-acetic acid (hereinafter referred to as "5-ph-IAA”), shown in the following formula (IV).
  • multiple refers to an integer of 2 or more, for example, 2 to 10, 2 to 7, 2 to 5, 2 to 4, or 2 to 3.
  • everal refers to, for example, 2 to 10, 2 to 7, 2 to 5, 2 to 4, or 2 to 3.
  • amino acid identity refers to the percentage of matching amino acid residues out of the total number of amino acid residues in the amino acid sequences of two polypeptides being compared, when aligned by inserting appropriate gaps into one or both, as necessary, to maximize the number of matching amino acid residues.
  • Base identity base sequence identity
  • amino acid substitution refers to substitutions between the 20 types of amino acids that make up natural proteins. Amino acid substitutions are preferably within conservative amino acid groups that have similar properties such as charge, side chain, polarity, and aromaticity. Examples include substitutions within the uncharged polar amino acids with low polarity side chains (Gly, Asn, Gln, Ser, Thr, Cys, Tyr), branched-chain amino acids (Leu, Val, Ile), neutral amino acids (Gly, Ile, Val, Leu, Ala, Met, Pro), neutral amino acids with hydrophilic side chains (Asn, Gln, Thr, Ser, Tyr, Cys), acidic amino acids (Asp, Glu), basic amino acids (Arg, Lys, His), and aromatic amino acids (Phe, Tyr, Trp).
  • the genetically modified mouse of the present invention comprises a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences, a TIR1 family gene sequence located downstream of the pair of Cre recombinase recognition sequences, and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences.
  • the entire sequence region comprising a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences, a TIR1 family gene sequence located downstream of the pair of Cre recombinase recognition sequences, and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences, which is capable of expressing a TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences, and which is composed of the TIR1 gene and an expression control region necessary for the gene expression, is referred to as a "TIR1 gene expression system" or simply a "gene expression system.”
  • the genetically modified mouse of the present invention comprises a TIR1 gene expression system.
  • the promoter sequence is located upstream of a pair of Cre recombinase recognition sequences
  • the TIR1 family gene sequence is located downstream of a pair of Cre recombinase recognition sequences.
  • upstream and downstream are defined based on the direction of transcription induced from the promoter. In other words, in the transcript transcribed from the promoter, the 5' side corresponds to the upstream side, and the 3' side corresponds to the downstream side.
  • the TIR1 gene expression system may be contained in the genome, or a vector such as an expression vector (e.g., a plasmid vector) containing the TIR1 gene expression system may be introduced into the genetically modified mouse.
  • a vector such as an expression vector (e.g., a plasmid vector) containing the TIR1 gene expression system may be introduced into the genetically modified mouse.
  • the TIR1 gene expression system When the TIR1 gene expression system is contained in the genome, it can be introduced into any position in the genome, but it is preferable to introduce it into a gene locus that can be expressed widely regardless of cell type or developmental stage and that is unlikely to affect the expression or function of other endogenous genes, such as the Rosa26 gene locus or the ⁇ -actin gene locus.
  • the genetically modified mouse comprises a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence, a TIR1 family gene sequence, and a promoter sequence in the Rosa26 locus (i.e., comprises a TIR1 gene expression system in the Rosa26 locus).
  • the "Rosa26 locus” is a genomic region on mouse chromosome 6, also referred to as Gt(ROSA)26Sor.
  • the position at which the TIR1 gene expression system is introduced in the Rosa26 locus is not particularly limited. For example, stable expression can be obtained by introducing the TIR1 gene expression system into the intron region between exon 1 and exon 2 of the Rosa26 locus.
  • the TIR1 family gene sequence is a nucleotide sequence or nucleic acid sequence encoding any TIR1 family protein.
  • examples include nucleotide sequences or nucleic acid sequences encoding wild-type or mutant TIR1 family proteins derived from Arabidopsis thaliana, rice, zinnia, pine, fern, or Physcomitrella patens.
  • nucleotide sequence or nucleic acid sequence encoding a wild-type TIR1 family protein derived from rice is a nucleotide sequence encoding the OsTIR1 protein derived from rice having the amino acid sequence shown in SEQ ID NO:1, for example an OsTIR1 gene sequence having the nucleotide sequence shown in SEQ ID NO:2.
  • mutant TIR1 family proteins include an amino acid sequence in which one or several (e.g., 1 to 100, 1 to 50, 1 to 20, 1 to 10, or 1 to 5) amino acids have been deleted, inserted, substituted, or added in the amino acid sequence shown in SEQ ID NO: 1, or a protein consisting of or containing an amino acid sequence having 70% or more, 80% or more, 90% or more, 95% or more, 98% or more, or 99% or more identity to the amino acid sequence shown in SEQ ID NO: 1.
  • This identity of the amino acid sequence can be calculated for the entire length of the amino acid sequence shown in SEQ ID NO: 1, or can be calculated for the amino acid sequence shown in SEQ ID NO: 1 excluding position 74, as described below.
  • the wild-type or mutant TIR1 family protein has an activity equivalent to or greater than that of wild-type TIR1, i.e., the activity of recognizing a degron in the presence of auxin or an auxin analog and leading to the degradation of a target protein.
  • the Phe residue corresponding to position 74 of the amino acid sequence shown in SEQ ID NO:1 is replaced with a Gly residue, an Ala residue, or a Ser residue (referred to as "F74G mutation", “F74A mutation”, “F74S mutation”, etc. in the present specification).
  • An example of a rice-derived TIR1 protein in which the Phe residue corresponding to position 74 of the amino acid sequence shown in SEQ ID NO:1 is replaced with a Gly residue is an example of the amino acid sequence shown in SEQ ID NO:3.
  • TIR1 family protein in this embodiment may be one that consists of or contains an amino acid sequence that has 70% or more, 80% or more, 90% or more, 95% or more, 98% or more, or 99% or more identity to the amino acid sequence shown in SEQ ID NO:1, except for position 74 of the amino acid sequence shown in SEQ ID NO:1.
  • the term "degron sequence” refers to an amino acid sequence that can induce degradation by the auxin degron system when added to a target protein.
  • a plant-derived Aux/IAA family protein or a fragment thereof functions as a degron sequence and can therefore be used as a degron sequence in the genetically modified mouse or TIR1 gene expression system of the present invention.
  • the gene encoding the Aux/IAA family protein is not particularly limited in type, so long as it is a plant-derived Aux/IAA family gene.
  • Examples of Arabidopsis-derived Aux/IAA family genes include the IAA1 to IAA34 genes, and preferably the IAA17 gene.
  • the sequences of Arabidopsis-derived Aux/IAA family genes are registered in TAIR (The Arabidopsis Information Resource) and are available as needed.
  • a fragment of a plant-derived Aux/IAA family protein that can be used as a degron sequence preferably has an activity of binding to a TIR1 family protein/auxin or auxin analog complex and leading to the degradation of a target protein, and may be, for example, a full-length or partial sequence of mAID, which will be described later.
  • mAID Mini-auxin-inducible degron
  • mAID refers to a degron sequence consisting of a partial sequence of Arabidopsis thaliana IAA17, which is one of the Aux/IAA family proteins.
  • mAID can be a sequence consisting of a region containing at least two Lys residues on the N-terminal and C-terminal sides of the domain II region of an Aux/IAA family protein, or a sequence consisting of two or more such sequences linked together.
  • a specific example of an mAID is the amino acid sequence shown in SEQ ID NO:5.
  • Cre recombinase recognition sequence refers to a sequence that is recognized by Cre recombinase and can induce recombination between two sequences.
  • Specific base sequences of Cre recombinase recognition sequences are known in the art, and examples include loxP sequences (ATAACTTCGTATAGCATACATTATACGAAGTTAT, SEQ ID NO: 6); lox511 sequences (ATAACTTCGTATAGTATACATTATACGAAGTTAT, SEQ ID NO: 7); lox2272 sequences (ATAACTTCGTATAGGATACTTTATACGAAGTTAT, SEQ ID NO: 8); and loxFAS sequences (ATAACTTCGTATATACCTTTCTATACGAAGTTAT, SEQ ID NO: 9).
  • a pair of Cre recombinase recognition sequences is located between the TIR1 family gene sequence and the promoter sequence, and a transcription termination polyA sequence is located between the pair of Cre recombinase recognition sequences.
  • a pair of Cre recombinase recognition sequences are generally arranged in the same direction. When a pair of Cre recombinase recognition sequences are present in the same direction, Cre recombinase recognizes the two sequences and induces site-specific recombination between them, resulting in the excision and removal of the sequence sandwiched between the two sequences.
  • transcription termination polyA sequence refers to a base sequence capable of causing the termination of transcription.
  • the transcription termination polyA sequence is also called a polyadenylation signal or transcription terminator, and can induce the termination of transcription and the polyadenylation of the 3' end of the transcription product.
  • the transcription termination polyA sequence is arranged between a pair of Cre recombinase recognition sequences, and transcription can be terminated on that sequence.
  • the type of transcription termination polyA sequence is not particularly limited, and any transcription termination polyA sequence can be used.
  • a transcription termination polyA sequence derived from the human growth hormone (HGH) gene, the SV40 gene, the rabbit ⁇ -globin gene, the bovine growth hormone (BGH) gene, or the phosphoglycerate kinase (PGK) gene may be used, or any known transcription termination polyA sequence that can function in eukaryotic cells may be used.
  • HGH human growth hormone
  • BGH bovine growth hormone
  • PGK phosphoglycerate kinase
  • one or more transcription termination polyA sequences can be arranged between a pair of Cre recombinase recognition sequences. For example, placing two or more transcription termination polyA sequences is advantageous because it is possible to suppress leaky expression of genes placed downstream.
  • the "promoter sequence” is a promoter that is located upstream of a pair of Cre recombinase recognition sequences and can drive the expression of the TIR1 family gene located downstream thereof after recombination between the pair of Cre recombinase recognition sequences.
  • transcription induced from the promoter sequence is terminated on the above-mentioned transcription termination polyA sequence, so that the TIR1 family gene sequence located downstream thereof is not transcribed or is not substantially transcribed.
  • the type of promoter sequence is not particularly limited as long as it can drive the expression of genes in the target cell type in which the auxin degron system is to function, and may be either an exogenous promoter or an endogenous promoter, a ubiquitous promoter (systemic promoter) or a site-specific promoter, or may be a constitutively active promoter, an expression-inducible promoter, or a time-specific active promoter.
  • the exogenous promoter may be a promoter derived from a mammal such as a mouse, or a promoter derived from a non-mammalian organism, and for example, a CAG promoter, a CMV promoter, an SV40 promoter, an EF1a promoter, or an RSV promoter can be used.
  • the endogenous promoter may be an endogenous promoter of a locus in the genome into which the TIR1 gene expression system is introduced. For example, when the TIR1 gene expression system is introduced into the Rosa26 gene locus, the expression of the TIR1 family gene may be driven by the endogenous promoter of the Rosa26 gene.
  • the above promoter sequence is preferably operably linked to the TIR1 family gene located downstream thereof after recombination between a pair of Cre recombinase recognition sequences.
  • operably linked refers to a functional link between the above promoter sequence and a TIR1 family gene sequence that encodes a TIR1 family protein. This functional link refers to a link that allows transcription of the TIR1 family gene sequence after recombination between a pair of Cre recombinase recognition sequences.
  • the genetically modified mouse of the present invention further comprises a gene encoding a target protein to which a degron sequence has been added and/or a gene encoding a Cre recombinase.
  • target protein to which the degron sequence is added there is no limit to the type of target protein to which the degron sequence is added.
  • it may be an exogenous protein or an endogenous protein.
  • the target protein may be, for example, a drug discovery target protein such as a disease-causing protein.
  • the position at which the degron sequence is added to the target protein is not limited, and may be, for example, the N-terminus and/or C-terminus of the target protein, or may be inserted into the middle of the amino acid sequence of the target protein.
  • the number of degron sequences added may be one or more.
  • the gene encoding the target protein to which the degron sequence has been added may be an endogenous gene into which a base sequence encoding the degron sequence has been introduced by knock-in or the like.
  • the gene encoding the target protein is essential for development and/or survival.
  • a gene essential for development here is, for example, a gene that causes developmental abnormalities in an individual when the gene is disrupted by whole-body knockout.
  • a gene essential for survival here is, for example, a gene that causes lethality in at least some individuals when the gene is disrupted by whole-body knockout. Such genes were difficult to analyze using conventional whole-body knockout methods, but can be analyzed by transient functional inhibition using the genetically modified mouse of the present invention.
  • the target protein is a membrane protein.
  • the membrane protein may be, for example, an integral membrane protein, a peripheral membrane protein, or a lipid-anchored protein.
  • the type of Cre recombinase is not limited, and may be, for example, a Cre recombinase derived from Escherichia coli P1 phage or a modified form thereof.
  • modified Cre recombinase include tamoxifen-inducible Cre recombinases such as Cre-ERT and Cre-ERT2.
  • the gene encoding the Cre recombinase is placed under the control of a site-specific promoter, such as a tissue-specific or cell type-specific promoter.
  • a site-specific promoter such as a tissue-specific or cell type-specific promoter.
  • Various Cre mice are available in the art, and by selecting and mating Cre mice according to the tissue or cell type of interest, it is possible and convenient to introduce the gene encoding the Cre recombinase placed under the control of a site-specific promoter into a genetically modified mouse.
  • the function of the gene encoding the target protein at the site where the expression is driven by the site-specific promoter is essential for development and/or survival.
  • a gene essential for development is, for example, a gene that causes developmental abnormalities in an individual when the gene is disrupted by conditional knockout based on the expression of Cre recombinase under the control of the same site-specific promoter.
  • a gene essential for survival is, for example, a gene that causes lethality in at least some individuals when the gene is disrupted by conditional knockout based on the expression of Cre recombinase under the control of the same site-specific promoter.
  • recombination between a pair of Cre recombinase recognition sequences is specifically induced in tissues or cell types where the expression of Cre recombinase is induced by a site-specific promoter, and as a result, the transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences is removed from the TIR1 gene expression system, and the expression of the TIR1 family gene located downstream of the Cre recombinase recognition sequence is induced, resulting in tissue- or cell-type-specific expression of the TIR1 family protein.
  • the TIR1 family protein binds to the auxin or auxin analog, and the target protein labeled with a degron can be degraded using the ubiquitin/proteasome degradation system of the host cell.
  • the present invention also provides sperm, fertilized eggs, or embryos that develop in the genetically modified mouse of the present invention.
  • the sperm, fertilized eggs, or embryos may be frozen sperm, frozen fertilized eggs, or frozen embryos, respectively.
  • cells derived from the genetically modified mouse of the present invention for example, mouse cells described below that are derived from the genetically modified mouse of the present invention.
  • the second aspect of the present invention is a method for degrading a target protein.
  • the method for degrading a target protein of this aspect can induce degradation of a target protein to which a degron sequence has been added by administering auxin or an auxin analog to a genetically modified mouse or adding it to a mouse cell.
  • degradation of a target protein can be induced in a fetus or a mouse pup before weaning by administering auxin or an auxin analog to a mother mouse.
  • Degradation of a candidate protein as a target for drug discovery can also be induced in a disease model mouse.
  • the target protein degradation method of the present invention can be carried out on a genetically modified mouse (mouse individual) or mouse cells derived from a genetically modified mouse.
  • the specific configuration of the target protein degradation method of the present invention differs depending on whether the target is a mouse individual or mouse cells.
  • the target protein degradation method of the present invention includes, as an essential step, a degradation induction step in which auxin or an auxin analog is administered to the genetically modified mouse to induce degradation of a target protein to which a degron sequence has been added, and includes, as a selection step, an evaluation step in which the phenotype of the genetically modified mouse after the degradation induction step is evaluated.
  • the target protein degradation method of the present invention includes, as an essential step, a culture step in which mouse cells are cultured, and a degradation induction step in which auxin or an auxin analog is added to the mouse cell to induce degradation of a target protein to which a degron sequence has been added, and includes, as a selection step, an evaluation step in which the phenotype of the mouse cell after the degradation induction step is evaluated.
  • the “degradation inducing step” refers, when a genetically modified mouse is the target, to a step of administering auxin or an auxin analogue to the genetically modified mouse to induce degradation of the target protein to which a degron sequence has been added, and, when a mouse cell is the target, to a step of adding auxin or an auxin analogue to the mouse cell to induce degradation of the target protein to which a degron sequence has been added.
  • the genetically modified mouse or mouse cell comprises a TIR1 family gene expression system, a gene encoding a target protein to which the degron sequence has been added, and a gene encoding a Cre recombinase.
  • the TIR1 family gene expression system comprises a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences, a TIR1 family gene sequence encoding a Transport Inhibitor Response 1 (TIR1) family protein located downstream of the pair of Cre recombinase recognition sequences, and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences and capable of driving expression of the TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences.
  • TIR1 family gene expression system comprises a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence located between the pair of Cre recombin
  • the developmental stage or week/fetal age of the genetically modified mouse does not matter.
  • the genetically modified mouse may be a fetal mouse, a perinatal mouse, a newborn mouse, a pre-weaning mouse, a young adult mouse, or an adult mouse.
  • the method of administration to genetically modified mice is not limited as long as it is capable of delivering auxin or auxin analogues to tissues or cell types that induce degradation of the target protein.
  • Examples include intravenous administration (tail vein injection), intraperitoneal administration, and subcutaneous administration.
  • auxin or auxin analogues can be administered indirectly to the mother mouse via the plasma that crosses the placenta or the milk that is fed.
  • auxin or auxin analogues can be administered to pregnant mice, and in the case of preweaned pups, auxin or auxin analogues can be administered indirectly to fetal mice or pups by administering them to lactating female mice.
  • the number of times the auxin or auxin analogue is administered to the genetically modified mouse is not limited. For example, it may be a single administration or multiple administrations, for example, once, twice, three times, four times, or more.
  • the multiple administrations may also be periodic administrations, for example repeated administrations every day, every two days, every three days, or once a week. The same applies to the number of times the auxin or auxin analogue is added to the mouse cells.
  • the dosage of the auxin or auxin analogue is not particularly limited as long as it is capable of inducing degradation of the target protein and does not have a deleterious effect on the genetically modified mouse, and may be, for example, 0.001 mg/kg to 10 g/kg body weight, 0.01 mg/kg to 1 g/kg body weight, or 0.1 mg/kg to 50 mg/kg body weight based on the body weight of the genetically modified mouse, and preferably 0.5 mg/kg to 10 mg/kg body weight or 1 mg/kg to 5 mg/kg body weight.
  • the dosage per administration is not particularly limited, and may be, for example, 0.02 mg to 200 g, 0.2 mg to 20 g, or 2 mg to 1 g, and preferably 10 mg to 200 mg or 20 mg to 100 mg.
  • the dosage can be adjusted depending on, for example, the desired degree of degradation; for example, a dosage aimed at a high level of degradation is 5 mg/kg body weight or more or 10 mg/kg body weight or more, and for example, a dosage aimed at a low level of degradation is 2 mg/kg body weight or less or 1 mg/kg body weight or less.
  • the amount of auxin or auxin analogue added to mouse cells is not particularly limited as long as it is capable of inducing degradation of the target protein and does not have a deleterious effect on the mouse cells, and may be, for example, 0.001 mg/L to 10 g/L, 0.01 mg/L to 1 g/L, or 0.1 mg/L to 50 mg/L.
  • the “evaluation step” refers to a step of evaluating the phenotype of a genetically modified mouse after the degradation induction step, when a genetically modified mouse is the target, and a step of evaluating the phenotype of mouse cells after the degradation induction step, when mouse cells are the target.
  • the "phenotype of a genetically modified mouse” is not particularly limited as long as it is an observable characteristic exhibited by an individual mouse, or an observable characteristic exhibited by an organ, tissue, cell, body fluid, etc. taken from an individual mouse.
  • the individual-level phenotype may be the appearance or shape of the whole body and body parts; weight, body fat percentage, electrocardiogram, muscle strength such as grip strength; or movement such as walking, sensory function, learning, information processing, feeding, excretion, etc. (behavioral phenotype).
  • the phenotype of an organ or tissue is not limited, and may be, for example, a characteristic observable in an organ such as the skin, heart, liver, intestine, or the central nervous system or peripheral nervous system such as the brain, a characteristic observable in the cells of any organ or tissue, or a characteristic observable in a body fluid such as blood or urine (for example, biomolecules such as proteins, lipids, and nucleic acids in a body fluid).
  • the "phenotype of mouse cells” is not limited to any characteristic that can be observed in a cell, whether it is the whole cell, the inside of the cell (e.g., organelles such as the cytoplasm, nucleus, mitochondria, endoplasmic reticulum, and Golgi apparatus), or the cell surface (e.g., cell membrane and cell surface), but may be, for example, the proliferation ability of a cell or the detected amount of a marker; the expression level or activity of a gene (e.g., nucleic acid such as mRNA or miRNA) or a protein; or the amount of an antigen or a metabolite.
  • organelles such as the cytoplasm, nucleus, mitochondria, endoplasmic reticulum, and Golgi apparatus
  • the cell surface e.g., cell membrane and cell surface
  • the proliferation ability of a cell or the detected amount of a marker e.g., the expression level or activity of a gene (e.g., nucleic acid such as m
  • mouse cells is not limited, and may be somatic cells or germ cells, or may be stem cells, precursor cells, undifferentiated cells, differentiated cells, or established cell lines (e.g., primary culture cells, cancer cells).
  • differentiated cells include fibroblasts, epithelial cells, hepatocytes, blood cells, immune cells, mesenchymal cells, nerve cells, and muscle cells.
  • the specific method of evaluating a phenotype is not particularly limited and can be selected according to the type of phenotype to be evaluated.
  • Specific examples of evaluation methods and means include serum biochemistry tests, immunological tests, electrocardiograms, body weight measurements, body fat measurements, organ weight measurements, behavioral observations, morphological observations, Y-mazes, grip strength tests, open field tests, flow cytometry, northern blotting, in situ hybridization, microarrays, RNA seq, PCR such as RT-PCR, ELISA, immunohistochemistry, and western blotting.
  • the evaluation step can be carried out at any time after the degradation induction step.
  • the evaluation step may be carried out 1 minute to 1 month, 30 minutes to 1 week, 1 hour to 3 days, 2 hours to 1 day, 3 hours to 12 hours, 5 hours to 10 hours, 7 hours to 8 hours after the degradation induction step, and preferably 3 hours or more later.
  • the evaluation step may be carried out during the period in which the target protein is degraded after the degradation induction step, or may be carried out after the target protein has been degraded and the original expression level has been restored.
  • the time point for carrying out the evaluation step may be selected depending on the desired phenotype.
  • the target protein is a candidate drug discovery target protein.
  • the term “candidate drug discovery target protein” refers to a protein that is a candidate when searching for a protein that is a target for drug discovery. Specifically, it is a candidate protein whose degradation is expected to produce a desired effect, such as a therapeutic or preventive effect on a disease or disease model, or a reduction or avoidance of side effects.
  • the genetically modified mouse may be a disease model mouse.
  • the specific type of the candidate drug discovery target protein is not limited.
  • it may be a protein whose beneficial effect has been overlooked in previous drug discovery because destruction based on whole-body knockout or conditional knockout of the gene encoding the candidate drug discovery target protein did not show a therapeutic or preventive effect on the disease model mouse or showed (harmful) side effects (e.g., lethality).
  • the term "disease” is not limited, and may be, for example, cancer, hepatitis, infectious disease, etc.
  • the type of “cancer” is not limited, and examples include adenocarcinoma, squamous cell carcinoma, small cell carcinoma, and large cell carcinoma.
  • Specific types of cancer include, for example, malignant melanoma, thyroid cancer, lung cancer, breast cancer, esophageal cancer, gastric cancer, colon cancer, small intestine cancer, prostate cancer, ovarian cancer, kidney cancer, liver cancer, pancreatic cancer, biliary tract cancer, brain tumor, head and neck cancer, mesothelioma, osteosarcoma, blood cancer, lymphoma, myeloma, etc.
  • the target protein degradation method of this aspect further includes a step of administering to the genetically modified mouse a drug targeting a factor other than the candidate drug target protein.
  • the "factor other than the candidate drug target protein” is a factor that is expected to enhance the desired effect, such as a therapeutic or preventive effect on a disease or disease model, or a reduction or avoidance of side effects, by targeting it in combination with the candidate drug target protein, and is, for example, any biological substance such as a protein or nucleic acid other than the candidate drug target protein.
  • the drug targeting a factor other than the candidate drug target protein is not limited and may be a low molecular weight compound, an antibody drug, or a nucleic acid drug such as siRNA or antisense nucleic acid, and may be administered simultaneously with or without the administration of auxin or auxin analog.
  • the evaluation step can compare the phenotype when both the candidate drug target protein and the factor are targeted with the phenotype when only the factor is targeted. This comparison makes it possible to determine the degree and presence of an effect of enhancing the desired effect, such as a therapeutic or preventive effect against a disease or a reduction or avoidance of side effects, by inducing the degradation of a candidate drug target protein in combination with the administration of a drug that targets a factor different from the candidate drug target protein.
  • the method for degrading a target protein of the present invention can control degradation of a target protein in a living mouse body or in mouse cells with extremely high time resolution, thereby achieving transient degradation and subsequent rapid recovery.
  • the function of the candidate protein is inhibited for a long period of time by destroying the candidate gene.
  • the phenotype of the mouse in this case may not necessarily match the phenotype when the developed therapeutic drug is actually administered to the animal to transiently control the function of the protein, and unexpected side effects may only become apparent after the developed therapeutic drug is actually administered. In such cases, the enormous investment of labor and cost required for therapeutic development is wasted.
  • the targeted protein degradation method of the present invention makes it possible to observe the effects of transiently controlling the function of a protein, making it possible to more accurately verify in advance the actual effects of the therapeutic drug to be developed in the process of searching for candidate proteins for drug discovery, reducing unnecessary investment costs and streamlining the initial stages of drug discovery.
  • Example 1 Establishment of Rosa26 OsTIR1(F74G) mouse line (the purpose) We will generate the Rosa26 OsTIR1(F74G) mouse line, which allows cell type-specific expression of the OsTIR1(F74G ) protein.
  • TIR1 Rice-derived Transport Inhibitor Response 1 family protein in which the Phe residue at position 74 in the amino acid sequence of the OsTIR1 protein (SEQ ID NO: 1) was replaced with a Gly residue, and the ires-EGFP sequence was placed downstream of a CAG promoter (pCAG) and a loxP-neor-STOP-loxP sequence (SEQ ID NO: 11) (Fig. 1A).
  • OsTIR1 (F74G) protein from the Rosa26 OsTIR1 (F74G) allele requires the removal of the loxP-neor-STOP-loxP sequence by site-specific recombination of the loxP sequence using Cre recombinase.
  • Tg transgenic mice expressing Cre recombinase in a tissue- or cell-specific manner
  • OsTIR1(F74G) protein can be expressed in a tissue- or cell-specific manner (Fig. 2).
  • Example 2 Time series analysis of protein degradation in vivo in mice (the purpose) Mice expressing a target protein fused with a mini AID degron sequence (mAID) are generated, and these mice are crossed with the Rosa26 OsTIR1(F74G) mice generated in Example 1 and Tg mice expressing Cre recombinase. An auxin analog is administered to the mice obtained by crossing to induce degradation of the target protein, and the time course of protein degradation in vivo is analyzed.
  • mAID mini AID degron sequence
  • Rosa26 OsTIR1(F74G) mice prepared in Example 1, Satb1 Venus-mAID mice, and Cd4Cre Tg mice (4-8 week-old male or female; provided by Dr. Chris Wilson, University of Washington) that express Cre recombinase specifically in T cells were crossed to generate Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice ( Figure 3A).
  • 5-Phenyl-indole-3-acetic acid (5-ph-IAA) was administered intraperitoneally (ip) to Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice at a dose of 0.1 mg/body or 0.02 mg/body. Blood was collected from the mice at each time point from 0 to 72 hours after administration, and the fluorescence intensity of Satb1-Venus in T cells in the peripheral blood was analyzed by flow cytometry.
  • Example 3 Induction of protein degradation in fetal and neonatal mice (the purpose) To verify the effect of target protein degradation in fetal and neonatal mice, 5-ph-IAA will be administered to pregnant or lactating female mice, and the degradation of target proteins will be analyzed in fetal or neonatal mice.
  • 5-ph-IAA was intraperitoneally administered at a dose of 0.1 mg/body to pregnant mice (17.5 days of pregnancy) or lactating female mice on the second day after birth for the Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice prepared in Example 2. 24 hours after administration, the thymus was removed from fetal mice or newborn mice lactated by female mice, and the fluorescence intensity of Satb1-Venus in T cells was analyzed by flow cytometry.
  • Example 4 Targeted degradation of membrane proteins (the purpose) The effect of targeting membrane proteins for protein degradation will be verified in cultured cells and in vivo in mice.
  • T cell lines treated with 5-ph-IAA showed reduced expression of PD-1 protein compared to control cells treated with DMSO.
  • Rosa26 OsTIR1(F74G) mice prepared in Example 1, Pdcd1-mAID mice, and Vav1Cre Tg mice (4-8 week-old male or female; purchased from Jackson Laboratory) that express Cre recombinase specifically in blood cells were crossed to generate Rosa26 OsTIR1/+ :Pdcd1-mAID:Vav1Cre mice ( Figure 7A).
  • Rosa26 OsTIR1/+ :Pdcd1-mAID Vav1Cre mice were intraperitoneally administered 5-ph-IAA or PBS at a dose of 0.1 mg/body. Twelve hours after administration, follicular T cells were prepared from Peyer's patches, and PD-1 protein expression was measured by flow cytometry.
  • Example 5 Verification of antitumor effect based on targeted degradation of PD1 protein (the purpose)
  • MC38 colon cancer was transplanted into the PD1 degron mice (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; Vavcre) generated in Example 4. From 10 days after transplantation, PBS or 5-ph-IAA was administered continuously once every two days ( FIG. 8A ), and the tumor diameters in the PBS-administered group and the 5-ph-IAA-administered group were compared.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Environmental Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Behavior & Ethology (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Microbiology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention provides novel technology which makes it possible to control the function of a protein in the living body of mouse with high temporal resolution. Provided is a genetically modified mouse, said genetically modified mouse comprising: a pair of Cre recombinase recognition sequences; a transcription termination polyA sequence that is located between the pair of Cre recombinase recognition sequences; a transport inhibitor response 1 (TIR1) family gene sequence that is located downstream of the pair of Cre recombinase recognition sequences and that codes for a TIR1 family protein; and a promoter sequence that is located upstream of the pair of Cre recombinase recognition sequences, and that can drive expression of the TIR1 family gene after recombination of the pair of Cre recombinase recognition sequences.

Description

遺伝子改変マウスGenetically engineered mice

 本発明は、遺伝子改変マウス、及び標的タンパク質の分解を誘導する方法等に関する。 The present invention relates to genetically modified mice and methods for inducing the degradation of target proteins.

 タンパク質の機能を解析する方法として、生体内において特定の遺伝子の機能を選択的に阻害又は破壊する技術は、生命科学の研究に大きく貢献してきた。その代表例であるノックアウトマウス作製技術は、相同組換えを利用したマウスES細胞ゲノムへの遺伝子ターゲティングに基づいており、これにより生体内における遺伝子機能や疾患の発症メカニズムの解明が大きく進展してきた。 Technologies that selectively inhibit or destroy the function of specific genes in vivo as a method for analyzing protein functions have made a major contribution to life science research. A representative example is the technology for producing knockout mice, which is based on gene targeting of the mouse ES cell genome using homologous recombination, and has made great progress in elucidating gene functions in vivo and the mechanisms of disease onset.

 その後、条件付きノックアウト技術が開発され、特定の組織や細胞種で遺伝子をノックアウトすることも可能になった。条件付きノックアウト技術では、標的とする遺伝子領域の前後にCreリコンビナーゼが認識する一対のloxP配列を導入したマウス(floxマウスと呼ばれる)を作出し、このfloxマウスと組織又は細胞種特異的にCreリコンビナーゼを発現するマウスとを交配させることにより、標的遺伝子を組織又は細胞種特異的に破壊する。条件付きノックアウト技術により、全身でノックアウトすると発生途中で致死性を示す遺伝子についても成体における遺伝子機能を研究することが可能となった。  Subsequently, conditional knockout technology was developed, making it possible to knock out genes in specific tissues or cell types. In conditional knockout technology, mice (called flox mice) are created in which a pair of loxP sequences recognized by Cre recombinase is introduced before and after the target gene region, and the target gene is destroyed in a tissue- or cell-type-specific manner by mating this flox mouse with a mouse that expresses Cre recombinase in a tissue- or cell-type-specific manner. Conditional knockout technology has made it possible to study gene functions in adults even for genes that are lethal during development when knocked out throughout the body.

 条件付きノックアウト技術では、薬剤投与により組換え活性が誘導される特性を有するCreERリコンビナーゼをCreリコンビナーゼとして使用することで、組織又は細胞種特異的かつ時期特異的な遺伝子破壊も実施可能である。 In conditional knockout technology, CreER recombinase, which has the property of inducing recombination activity by drug administration, is used as the Cre recombinase, making it possible to perform tissue- or cell-type-specific and time-specific gene destruction.

 しかしながら、条件付きノックアウト技術では、操作対象遺伝子によってコードされるタンパク質の機能を高い時間分解能で一過的に抑制することは、依然としてできない。例えば、薬剤投与により組換え活性を誘導できるCreERリコンビナーゼを用いた場合では、薬剤投与により遺伝子破壊が誘導された後においても、破壊前の遺伝子から発現していたタンパク質が一定期間残存するため、解析対象のタンパク質の発現が実際に低下し、その活性が消失するまでには長い時間を要する。 However, conditional knockout technology is still unable to transiently suppress the function of the protein encoded by the manipulated gene with high temporal resolution. For example, when using CreER recombinase, which can induce recombination activity by drug administration, even after gene destruction is induced by drug administration, the protein expressed from the gene before destruction remains for a certain period of time, so it takes a long time for the expression of the target protein to actually decrease and its activity to disappear.

 上記のような問題点は、創薬において治療薬を開発する前に標的とするタンパク質を選定する際に、大きな問題となる。全身ノックアウトや条件付きノックアウトを用いて候補遺伝子を破壊することにより長期に亘って候補タンパク質の機能を阻害する場合の生体機能への影響と、実際に開発した治療薬を動物に投与して一過的にタンパク質の機能を阻害した場合の影響とは、必ずしも一致しない場合があり、実際に開発された治療薬を投与することによって初めて予想外の副作用が明らかになる場合もある。このような場合、治療薬の開発に要した膨大な労力やコストの投資が無駄になってしまうため大きな問題である。 The above problems are a major issue in drug discovery when selecting target proteins before developing a therapeutic drug. The impact on biological functions when inhibiting the function of a candidate protein over the long term by destroying the candidate gene using whole-body knockout or conditional knockout does not necessarily match the impact when the protein's function is inhibited temporarily by administering the actual therapeutic drug to an animal, and sometimes unexpected side effects become apparent only after the actual therapeutic drug is administered. In such cases, the enormous amount of effort and investment required to develop the therapeutic drug is wasted, which is a major problem.

国際公開第2021/009993号International Publication No. 2021/009993

 本発明の目的は、マウス生体内において組織又は細胞種特異的にタンパク質の機能を高い時間分解能で制御することを可能とする、新たな技術を提供することである。 The objective of the present invention is to provide a new technology that enables tissue- or cell-type-specific control of protein function in vivo with high temporal resolution.

 オーキシンデグロン法は、オーキシン応答性ユビキチンリガーゼを構成するTransport Inhibitor Response 1(TIR1)ファミリータンパク質を細胞に導入し、植物ホルモンのオーキシンやオーキシンアナログの添加により、分解タグ(デグロンと呼ばれる)を融合した標的タンパク質の分解を制御する技術である。 The auxin degron method is a technology that involves introducing the Transport Inhibitor Response 1 (TIR1) family protein, which constitutes an auxin-responsive ubiquitin ligase, into cells and controlling the degradation of target proteins fused with a degradation tag (called a degron) by adding the plant hormone auxin or an auxin analogue.

 本発明者らは、上記課題を解決するために、F74G変異を有するイネ由来TIR1タンパク質(以下、「OsTIR1(F74G)タンパク質」と称する)をコードするcDNA配列の上流にLoxP-STOP-LoxP(LSL)配列を配置した構築物をRosa26遺伝子座に導入したRosa26OsTIR1(F74G)マウスを作製した。このRosa26OsTIR1(F74G)マウスに特定の組織又は細胞種特異的にCreリコンビナーゼを発現するトランスジェニック(Tg)マウスを交配させることによって、組織又は細胞種特異的にOsTIR1(F74G)タンパク質を発現するマウスを作製した。本発明者らは、このマウスにオーキシンアナログを投与した結果、デグロンを融合した標的タンパク質の一過的な分解が極めて迅速に誘導され、その後速やかに元の発現レベルが回復し得ることを見出した。さらに、オーキシンアナログの妊娠マウスへの投与により胎仔マウスにおけるタンパク質分解の誘導や、授乳中の雌マウスへの投与により新生仔マウスにおけるタンパク質分解の誘導も達成され、膜タンパク質を標的タンパク質とする分解についてもマウス生体内において誘導できることを見出した。 In order to achieve the above object, the present inventors have generated Rosa26 OsTIR1(F74G) mice by introducing a construct in which a LoxP-STOP-LoxP (LSL) sequence is arranged upstream of a cDNA sequence encoding a rice-derived TIR1 protein having an F74G mutation (hereinafter referred to as "OsTIR1(F74G) protein") into the Rosa26 locus. The Rosa26 OsTIR1 ( F74G) mice were crossed with transgenic (Tg) mice expressing Cre recombinase in a specific tissue or cell type-specific manner to generate mice expressing OsTIR1(F74G) protein in a tissue or cell type-specific manner. The present inventors have found that administration of an auxin analog to the mice induced transient degradation of a degron-fused target protein very quickly, and the original expression level was then rapidly restored. Furthermore, administration of an auxin analogue to pregnant mice induced protein degradation in fetal mice, and administration of an auxin analogue to lactating female mice induced protein degradation in newborn mice, demonstrating that degradation of membrane proteins as target proteins can also be induced in vivo in mice.

 本発明は、上記研究成果に基づくものであって、以下を提供する。
(1)遺伝子改変マウスであって、
 一対のCreリコンビナーゼ認識配列、
 前記一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列、
 前記一対のCreリコンビナーゼ認識配列の下流に位置する、Transport Inhibitor Response 1(TIR1)ファミリータンパク質をコードするTIR1ファミリー遺伝子配列、及び
 前記一対のCreリコンビナーゼ認識配列の上流に位置し、前記一対のCreリコンビナーゼ認識配列間の組換え後に前記TIR1ファミリー遺伝子の発現を駆動することができるプロモーター配列
を含む、前記遺伝子改変マウス。
(2)前記TIR1ファミリータンパク質において、配列番号1で示すアミノ酸配列の74位に対応するPhe残基が、Gly残基、Ala残基、又はSer残基に置換されている、(1)に記載の遺伝子改変マウス。
(3)前記一対のCreリコンビナーゼ認識配列、前記転写終結polyA配列、前記TIR1ファミリー遺伝子配列、及び前記プロモーター配列をRosa26遺伝子座に含む、(1)又は(2)に記載の遺伝子改変マウス。
(4)デグロン配列が付加した標的タンパク質をコードする遺伝子、及び/又はCreリコンビナーゼをコードする遺伝子をさらに含む、(1)~(3)のいずれかに記載の遺伝子改変マウス。
(5)前記標的タンパク質をコードする遺伝子が、発生及び/又は生存に必須である、(4)に記載の遺伝子改変マウス。
(6)前記標的タンパク質が膜タンパク質である、(4)又は(5)に記載の遺伝子改変マウス。
(7)前記Creリコンビナーゼをコードする遺伝子が、部位特異的プロモーターの制御下に配置されている、(4)~(6)のいずれかに記載の遺伝子改変マウス。
(8)前記部位特異的プロモーターが発現を駆動する部位における、前記標的タンパク質をコードする遺伝子の機能が、発生及び/又は生存に必須である、(7)に記載の遺伝子改変マウス。
(9)(1)~(8)のいずれかに記載の遺伝子改変マウスに発生する、精子、受精卵、又は胚。
(10)(1)~(8)のいずれかに記載の遺伝子改変マウスに由来する細胞。
(11)標的タンパク質の分解をマウス生体内で誘導する方法であって、
 オーキシン又はオーキシンアナログを遺伝子改変マウスに投与して、デグロン配列が付加した標的タンパク質の分解を誘導する、分解誘導工程
を含み、
 前記遺伝子改変マウスは、TIR1ファミリー遺伝子発現システム、前記デグロン配列が付加した標的タンパク質をコードする遺伝子、及びCreリコンビナーゼをコードする遺伝子を含み、
 前記TIR1ファミリー遺伝子発現システムは、
  一対のCreリコンビナーゼ認識配列、
  前記一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列、
  前記一対のCreリコンビナーゼ認識配列の下流に位置する、Transport Inhibitor Response 1(TIR1)ファミリータンパク質をコードするTIR1ファミリー遺伝子配列、及び
  前記一対のCreリコンビナーゼ認識配列の上流に位置し、前記一対のCreリコンビナーゼ認識配列間の組換え前には、前記TIR1ファミリー遺伝子の発現を駆動することが実質的にはできないが、前記一対のCreリコンビナーゼ認識配列間の組換え後に前記TIR1ファミリー遺伝子の発現を駆動することができるプロモーター配列
を含む、前記方法。
(12)前記オーキシン又はオーキシンアナログの投与量が、0.1 mg/kg体重~50 mg/kg体重である、(11)に記載の方法。
(13)前記分解誘導工程後の前記遺伝子改変マウスの表現型を評価する評価工程をさらに含む、(11)又は(12)に記載の方法。
(14)前記分解誘導工程の3時間以上後に前記評価工程を行う、(13)に記載の方法。
(15)前記遺伝子改変マウスが胎仔又は離乳前の仔マウスであり、前記分解誘導工程においてオーキシン又はオーキシンアナログをその母親マウスに投与する、(11)~(14)のいずれかに記載の方法。
(16)前記遺伝子改変マウスが疾患モデルマウスであり、前記標的タンパク質が創薬標的候補タンパク質である、(11)~(15)のいずれかに記載の方法。
(17)前記創薬標的候補タンパク質をコードする遺伝子の全身ノックアウト又は条件付きノックアウトに基づく破壊は、前記疾患モデルマウスに対する治療又は予防効果を示さないか、又は(有害な)副作用を示す、(16)に記載の方法。
(18)前記創薬標的候補タンパク質とは異なる因子を標的とする薬剤を前記遺伝子改変マウスに投与する工程をさらに含み、
 前記評価工程が、前記創薬標的候補タンパク質と前記因子との両者を標的とした場合の表現型と、前記因子のみを標的とした場合の表現型とを比較することを含む、(16)又は(17)に記載の方法。
(19)標的タンパク質の分解をマウス由来の細胞において誘導する方法であって、
 (10)に記載の細胞を培養する、培養工程、
 オーキシン又はオーキシンアナログを前記細胞に添加して、デグロン配列が付加した標的タンパク質の分解を誘導する、分解誘導工程
を含む、前記方法。
 本明細書は本願の優先権の基礎となる日本国特許出願番号2023-036211号の開示内容を包含する。
The present invention is based on the above research results and provides the following.
(1) A genetically modified mouse,
A pair of Cre recombinase recognition sequences,
A transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences;
The genetically modified mouse comprises: a Transport Inhibitor Response 1 (TIR1) family gene sequence encoding a TIR1 family protein, which is located downstream of the pair of Cre recombinase recognition sequences; and a promoter sequence, which is located upstream of the pair of Cre recombinase recognition sequences and is capable of driving expression of the TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences.
(2) The genetically modified mouse described in (1), in which in the TIR1 family protein, the Phe residue corresponding to position 74 of the amino acid sequence shown in SEQ ID NO: 1 is replaced with a Gly residue, an Ala residue, or a Ser residue.
(3) A genetically modified mouse described in (1) or (2), which contains the pair of Cre recombinase recognition sequences, the transcription termination polyA sequence, the TIR1 family gene sequence, and the promoter sequence in the Rosa26 locus.
(4) The genetically modified mouse according to any one of (1) to (3), further comprising a gene encoding a target protein to which a degron sequence has been added and/or a gene encoding Cre recombinase.
(5) The genetically modified mouse described in (4), wherein the gene encoding the target protein is essential for development and/or survival.
(6) The genetically modified mouse according to (4) or (5), wherein the target protein is a membrane protein.
(7) The genetically modified mouse according to any one of (4) to (6), wherein the gene encoding the Cre recombinase is placed under the control of a site-specific promoter.
(8) The genetically modified mouse described in (7), wherein the function of the gene encoding the target protein at the site where the site-specific promoter drives expression is essential for development and/or survival.
(9) A sperm, a fertilized egg, or an embryo generated in a genetically modified mouse according to any one of (1) to (8).
(10) A cell derived from a genetically modified mouse according to any one of (1) to (8).
(11) A method for inducing degradation of a target protein in a mouse body, comprising:
A degradation induction step of administering an auxin or an auxin analog to a genetically modified mouse to induce degradation of a target protein to which a degron sequence has been added,
The genetically modified mouse comprises a TIR1 family gene expression system, a gene encoding a target protein to which the degron sequence has been added, and a gene encoding a Cre recombinase;
The TIR1 family gene expression system comprises:
A pair of Cre recombinase recognition sequences,
A transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences;
the method comprising: a TIR1 family gene sequence encoding a Transport Inhibitor Response 1 (TIR1) family protein, located downstream of the pair of Cre recombinase recognition sequences; and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences, which is substantially incapable of driving expression of the TIR1 family gene before recombination between the pair of Cre recombinase recognition sequences, but is capable of driving expression of the TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences.
(12) The method according to (11), wherein the amount of the auxin or auxin analog administered is 0.1 mg/kg body weight to 50 mg/kg body weight.
(13) The method according to (11) or (12), further comprising an evaluation step of evaluating the phenotype of the genetically modified mouse after the degradation induction step.
(14) The method according to (13), wherein the evaluation step is carried out at least 3 hours after the decomposition induction step.
(15) The method according to any one of (11) to (14), wherein the genetically modified mouse is a fetus or a preweaned mouse, and an auxin or an auxin analog is administered to the mother mouse in the degradation induction step.
(16) The method according to any one of (11) to (15), wherein the genetically modified mouse is a disease model mouse, and the target protein is a candidate protein for drug discovery target.
(17) The method according to (16), wherein disruption based on whole-body knockout or conditional knockout of a gene encoding the candidate drug discovery target protein has no therapeutic or preventive effect on the disease model mouse or exhibits (harmful) side effects.
(18) The method further comprises administering to the genetically modified mouse a drug targeting a factor other than the drug discovery target candidate protein;
The method according to (16) or (17), wherein the evaluation step comprises comparing a phenotype obtained when both the candidate drug discovery target protein and the factor are targeted with a phenotype obtained when only the factor is targeted.
(19) A method for inducing degradation of a target protein in a mouse-derived cell, comprising the steps of:
(10) A culturing step of culturing the cell according to (10);
The method described above, further comprising a degradation inducing step of adding an auxin or an auxin analog to the cells to induce degradation of the target protein to which the degron sequence has been added.
This specification includes the disclosure of Japanese Patent Application No. 2023-036211, which is the priority basis of this application.

 本発明によれば、マウス生体内において組織又は細胞種特異的にタンパク質の機能を高い時間分解能で制御することを可能とする遺伝子改変マウスが提供される。 The present invention provides a genetically modified mouse that allows for tissue- or cell-type-specific control of protein function with high temporal resolution in vivo.

Rosa26OsTIR1(F74G)マウス系統の作製方法を示す図である。図1Aは、CAGプロモーター(pCAG)、loxP-neor-STOP-loxP配列、OsTIR1(F74G) cDNA配列、及びires-EGFP配列を含むTarget VectorによるRosa26遺伝子座における相同組換えを示す。図1Bは、作製したRosa26OsTIR1(F74G)アレルを示す。A diagram showing the method for generating the Rosa26 OsTIR1(F74G) mouse line. Figure 1A shows homologous recombination at the Rosa26 locus using a Target Vector containing a CAG promoter (pCAG), loxP-neor-STOP-loxP sequence, OsTIR1(F74G) cDNA sequence, and ires-EGFP sequence. Figure 1B shows the generated Rosa26 OsTIR1(F74G) allele. CreリコンビナーゼによるloxP配列の部位特異的組換えによってloxP-neor-STOP-loxP配列が除去され、OsTIR1(F74G)タンパク質の発現が誘導されることを示す図である。FIG. 13 shows that the loxP-neor-STOP-loxP sequence is removed by site-specific recombination of the loxP sequence by Cre recombinase, thereby inducing expression of the OsTIR1(F74G) protein. マウス生体内におけるタンパク質分解の時系列解析を示す図である。図3Aは、Rosa26OsTIR1/+:Satb1Venus-mAID/+:Cd4Creマウスを作製するための交配を示す。図3Bは、5-ph-IAAを0.1 mg/bodyの用量で投与した後、抹消血T細胞においてSatb1-Venusの蛍光強度をフローサイトメトリーにより解析した結果を示す。図3Cは、5-ph-IAAを0.02 mg/bodyの用量で投与した後、抹消血T細胞においてSatb1-Venusの蛍光強度をフローサイトメトリーにより解析した結果を示す。This is a diagram showing a time series analysis of protein degradation in a mouse in vivo. FIG. 3A shows the crossbreeding for producing Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice. FIG. 3B shows the results of flow cytometry analysis of the fluorescence intensity of Satb1-Venus in peripheral blood T cells after administration of 5-ph-IAA at a dose of 0.1 mg/body. FIG. 3C shows the results of flow cytometry analysis of the fluorescence intensity of Satb1-Venus in peripheral blood T cells after administration of 5-ph-IAA at a dose of 0.02 mg/body. 図3B及び図3Cの結果をグラフで示す図である。FIG. 4 is a graph showing the results of FIG. 3B and FIG. 3C. 妊娠中又は授乳中の雌マウスに5-ph-IAAを投与し、胎仔又は新生仔マウスから摘出した胸腺のT細胞におけるSatb1-Venusの蛍光強度をフローサイトメトリーにより解析した結果を示す。5-ph-IAA was administered to pregnant or lactating female mice, and the fluorescence intensity of Satb1-Venus in T cells in the thymus excised from fetal or neonatal mice was analyzed by flow cytometry. 膜タンパク質であるPD-1タンパク質を標的としてin vitroでタンパク質分解を誘導した結果を示す図である。This figure shows the results of inducing protein degradation in vitro by targeting the membrane protein PD-1. 膜タンパク質であるPD-1タンパク質を標的としてマウス生体内でタンパク質分解を誘導した結果を示す図である。図7Aは、Rosa26OsTIR1/+:Pdcd1-mAID:Vav1Creマウスを作製するための交配を示す。図7Bは、5-ph-IAA又はPBSを腹腔内投与した12時間後にパイエル板の濾胞性T細胞におけるPD-1タンパク質の発現をフローサイトメトリーで解析した結果を示す。Figure 7 shows the results of inducing protein degradation in vivo in mice by targeting the membrane protein PD-1. Figure 7A shows the crossbreeding for generating Rosa26 OsTIR1/+ :Pdcd1-mAID:Vav1Cre mice. Figure 7B shows the results of flow cytometry analysis of PD-1 protein expression in follicular T cells in Peyer's patches 12 hours after intraperitoneal administration of 5-ph-IAA or PBS. MC38大腸癌が移植された全血球型細胞でOsTIR1を発現するPD1デグロンマウス(Rosa-OsTIR1/+; Pdcd1mAID/mAID;Vavcre)においてPD1の分解を誘導した結果を示す図である。図8Aは、実験の概要を示す。図8Bは、腫瘍浸潤細胞(TIL)におけるPD1の発現レベルを測定した結果を示す。図8Cは、PBS投与群及び5-ph-IAA投与群における腫瘍径の測定結果を示す。FIG. 8 shows the results of inducing PD1 degradation in PD1 degron mice (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; Vavcre) expressing OsTIR1 in whole blood cells transplanted with MC38 colon cancer. FIG. 8A shows an outline of the experiment. FIG. 8B shows the results of measuring the expression level of PD1 in tumor-infiltrating cells (TIL). FIG. 8C shows the results of measuring the tumor diameter in the PBS-administered group and the 5-ph-IAA-administered group. MC38大腸癌が移植されたCD8T細胞のみでOsTIR1を発現するPD1デグロンマウス(Rosa-OsTIR1/+;Pdcd1mAID/mAID;E8Icreマウス)においてPD1の分解を誘導した結果を示す図である。図9Aは、当該PD1デグロンマウスのPBS投与群及び5-ph-IAA投与群における腫瘍径の測定結果を示す。図9Bは、CD8T細胞のみでOsTIR1を発現するPD1デグロンマウス(Rosa-OsTIR1/+;Pdcd1mAID/mAID;E8Icreマウス)、又は全血球型細胞でOsTIR1を発現するPD1デグロンマウス(Rosa-OsTIR1/+; Pdcd1mAID/mAID;Vavcre)に5-ph-IAAを投与した場合の腫瘍径を比較した結果を示す。FIG. 9A shows the results of inducing PD1 degradation in PD1 degron mice (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; E8Icre mice) expressing OsTIR1 only in CD8 T cells transplanted with MC38 colon cancer. FIG. 9A shows the results of measuring the tumor diameter in the PBS-administered group and the 5-ph-IAA-administered group of the PD1 degron mice. FIG. 9B shows the results of comparing the tumor diameter when 5-ph-IAA was administered to PD1 degron mice expressing OsTIR1 only in CD8 T cells (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; E8Icre mice) and PD1 degron mice expressing OsTIR1 in all blood cells (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; Vavcre).

1.遺伝子改変マウス
1-1.概要
 本発明の第1の態様は、遺伝子改変マウスである。本態様の遺伝子改変マウスは、一対のCreリコンビナーゼ認識配列間の組換え後にTIR1ファミリー遺伝子の発現を誘導することができる遺伝子発現システムを含む。本態様の遺伝子改変マウスでは、Creリコンビナーゼを組織又は細胞種特異的に発現させることによって、TIR1ファミリータンパク質の発現が誘導され、さらに任意の時点におけるオーキシン又はオーキシンアナログの投与によって、デグロン配列が付加した標的タンパク質の分解を一過的に誘導することができる。
1. Genetically Modified Mouse 1-1. Overview The first aspect of the present invention is a genetically modified mouse. The genetically modified mouse of this aspect includes a gene expression system capable of inducing expression of a TIR1 family gene after recombination between a pair of Cre recombinase recognition sequences. In the genetically modified mouse of this aspect, expression of a TIR1 family protein is induced by tissue- or cell-type-specific expression of Cre recombinase, and further, degradation of a target protein to which a degron sequence has been added can be transiently induced by administration of auxin or an auxin analog at any time point.

1-2.定義
 本明細書で頻用する以下の用語について定義する。
 本明細書において「遺伝子改変マウス(トランスジェニックマウス)」のマウス系統や遺伝的バックグラウンドは制限されず、例えばC57BL/6N系統、C57BL/6J系統、BALB/C系統、129系統等であってもよく、又は複数の系統を交配して得られる交雑系統であってもよい。また、遺伝子改変マウスは、例えばノックイン(KI)マウス、又はKIマウス以外のトランスジェニックマウス(例:外来遺伝子配列がゲノム上のランダムな位置に導入されたTgマウス)であってもよい。遺伝子改変マウスの作出方法は制限されず、例えば従来型の相同組換えに基づく遺伝子ターゲティングやCRISPR/Cas9システムを利用したゲノム編集法で作製した遺伝子改変マウスであってもよい。遺伝子改変マウスの発生段階や週齢/胎齢は問わず、例えば胎仔マウス、周産期マウス、新生仔マウス、離乳前マウス、若年成体マウス、又は成体マウスのいずれであってもよい。さらに遺伝子改変マウスは、雄性又は雌性のいずれであってもよい。また、遺伝子改変マウスは疾患モデルマウスであってよく、例えば遺伝子変異、遺伝子異常、遺伝子導入、薬剤処理、及び/又は外科的介入等により疾患を発症した又は疾患を発症する可能性のあるマウスであってもよい。
1-2. Definitions The following terms frequently used in this specification are defined below.
In the present specification, the mouse strain or genetic background of the "genetically modified mouse (transgenic mouse)" is not limited, and may be, for example, a C57BL/6N strain, a C57BL/6J strain, a BALB/C strain, a 129 strain, or the like, or may be a hybrid strain obtained by crossing a plurality of strains. The genetically modified mouse may be, for example, a knock-in (KI) mouse, or a transgenic mouse other than a KI mouse (e.g., a Tg mouse in which a foreign gene sequence is introduced at a random position on the genome). The method of producing the genetically modified mouse is not limited, and may be, for example, a genetically modified mouse produced by gene targeting based on conventional homologous recombination or a genome editing method using the CRISPR/Cas9 system. The genetically modified mouse may be at any developmental stage or week/fetal age, and may be, for example, a fetal mouse, a perinatal mouse, a neonatal mouse, a preweaning mouse, a young adult mouse, or an adult mouse. Furthermore, the genetically modified mouse may be either male or female. In addition, the genetically modified mouse may be a disease model mouse, for example, a mouse that has developed a disease or may develop a disease due to genetic mutation, genetic abnormality, gene introduction, drug treatment, and/or surgical intervention, etc.

 本明細書において「TIR1(Transport Inhibitor Response 1)ファミリータンパク質」とは、ユビキチン/プロテアソーム系のタンパク質分解において、E3ユビキチン化酵素複合体(SCF複合体)を形成するサブユニットの1つであるF-boxタンパク質である。植物由来のTIR1ファミリータンパク質は、成長ホルモンであるオーキシンの受容体として機能し、オーキシンとの結合に依存して、オーキシン情報伝達系の抑制因子Aux/IAAファミリータンパク質を認識して、標的タンパク質を分解することが知られている。本明細書においてTIR1ファミリータンパク質の由来植物種は制限されず、例えばシロイヌナズナ、イネ、ヒャクニチソウ、マツ、シダ、ヒメツリガネゴケ等であってもよい。具体例としては、例えば配列番号1で示すアミノ酸配列からなるイネ由来のTIR1タンパク質(以下、「OsTIR1タンパク質」という)が挙げられる。 In this specification, the term "TIR1 (Transport Inhibitor Response 1) family protein" refers to an F-box protein, which is one of the subunits that form the E3 ubiquitination complex (SCF complex) in the protein degradation of the ubiquitin/proteasome system. Plant-derived TIR1 family proteins function as receptors for the growth hormone auxin, and are known to recognize the Aux/IAA family proteins, which are inhibitors of the auxin signaling system, and degrade target proteins depending on the binding with auxin. In this specification, the plant species from which the TIR1 family proteins are derived is not limited, and may be, for example, Arabidopsis thaliana, rice, zinnia, pine, fern, Physcomitrella patens, etc. A specific example is the rice-derived TIR1 protein (hereinafter referred to as "OsTIR1 protein"), which consists of the amino acid sequence shown in SEQ ID NO: 1.

 本明細書において「TIR1ファミリー遺伝子配列」とは、TIR1ファミリータンパク質をコードする遺伝子の塩基配列又は核酸配列をいう。TIR1ファミリー遺伝子配列は、植物由来のTIR1ファミリータンパク質をコードする遺伝子であれば、その種類は限定されない。また、由来となる植物の種類も限定されず、例えば、シロイヌナズナ、イネ、ヒャクニチソウ、マツ、シダ、ヒメツリガネゴケ等に由来してもよい。TIR1ファミリー遺伝子配列の具体例としては、例えばTIR1遺伝子、AFB1遺伝子、AFB2遺伝子、AFB3遺伝子、FBX14遺伝子、及びAFB5遺伝子の塩基配列等が挙げられる。中でも、イネ由来のTIR1ファミリー遺伝子である、OsTIR1遺伝子は好ましく、係る遺伝子としては、NCBIアクセッション番号NM_001059194(GeneID:4335696)、Os04g0395600又はアクセッション番号EAY93933、OsI_15707の遺伝子が挙げられ、より具体的には配列番号1で示すアミノ酸配列からなるOsTIR1タンパク質をコードする、配列番号2で示す塩基配列からなるOsTIR1遺伝子配列が挙げられる。OsTIR1遺伝子配列等のTIR1ファミリー遺伝子配列は、マウス細胞等にコドン最適化した配列とすることもできる。 As used herein, "TIR1 family gene sequence" refers to the base sequence or nucleic acid sequence of a gene encoding a TIR1 family protein. There is no limitation on the type of TIR1 family gene sequence, so long as it is a gene encoding a plant-derived TIR1 family protein. There is also no limitation on the type of plant from which it is derived, and it may be derived, for example, from Arabidopsis thaliana, rice, zinnia, pine, fern, Physcomitrella patens, etc. Specific examples of TIR1 family gene sequences include the base sequences of the TIR1 gene, AFB1 gene, AFB2 gene, AFB3 gene, FBX14 gene, and AFB5 gene. Among them, the OsTIR1 gene, which is a TIR1 family gene derived from rice, is preferred, and examples of such genes include the genes with NCBI accession numbers NM_001059194 (GeneID: 4335696), Os04g0395600, or accession numbers EAY93933 and OsI_15707, and more specifically, the OsTIR1 gene sequence consisting of the nucleotide sequence shown in SEQ ID NO: 2, which encodes the OsTIR1 protein consisting of the amino acid sequence shown in SEQ ID NO: 1. The TIR1 family gene sequences such as the OsTIR1 gene sequence may also be codon-optimized for mouse cells, etc.

 本明細書において「オーキシンデグロンシステム」とは、植物ホルモンオーキシンに基づく植物のタンパク質分解系を非植物由来の真核細胞等の宿主細胞に応用したタンパク質分解制御システムをいう。具体的にはこのシステムは、上述のTIR1ファミリータンパク質がオーキシン又はオーキシンアナログとの結合依存的にデグロンで標識された標的タンパク質を宿主細胞のユビキチン/プロテアソーム分解系を利用して分解へと導くシステムである。 In this specification, the term "auxin degron system" refers to a protein degradation control system that applies the plant protein degradation system based on the plant hormone auxin to host cells such as eukaryotic cells of non-plant origin. Specifically, this system is a system in which the above-mentioned TIR1 family protein guides a target protein labeled with a degron to degradation using the ubiquitin/proteasome degradation system of the host cell in a manner dependent on binding with auxin or an auxin analog.

 本明細書において「オーキシン」とは、植物の成長を制御する一群の植物ホルモンをいう。本明細書においてオーキシンは、天然に存在するオーキシン及びオーキシンと類似した生理作用を示す非天然のオーキシン(合成オーキシン)のいずれも包含する。天然に存在するオーキシンとしては、インドール-3-酢酸(IAA)や4-クロロインドール-3-酢酸(4Cl-IAA)が知られている。合成オーキシンとしては、ナフタレン酢酸、ナフトキシ酢酸、フェニル酢酸、2,4-ジクロロフェノキシ酢酸(2,4-D)、及び2,4,5-トリクロロフェノキシ酢酸(2,4,5-T)等が知られている。天然オーキシンの1つであるインドール-3-酢酸(IAA)の構造を以下の式(I)に示す。 In this specification, "auxin" refers to a group of plant hormones that control plant growth. In this specification, auxin includes both naturally occurring auxins and non-natural auxins (synthetic auxins) that exhibit physiological actions similar to those of auxins. Known naturally occurring auxins include indole-3-acetic acid (IAA) and 4-chloroindole-3-acetic acid (4Cl-IAA). Known synthetic auxins include naphthaleneacetic acid, naphthoxyacetic acid, phenylacetic acid, 2,4-dichlorophenoxyacetic acid (2,4-D), and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T). The structure of indole-3-acetic acid (IAA), one of the natural auxins, is shown in the following formula (I).

Figure JPOXMLDOC01-appb-C000001
Figure JPOXMLDOC01-appb-C000001

 本明細書において「オーキシンアナログ」とは、上記いずれかのオーキシン、例えばインドール-3-酢酸(IAA)又は4-クロロインドール-3-酢酸(4Cl-IAA)等の天然オーキシンに類似した構造を有し、TIR1ファミリータンパク質と結合してデグロンで標識された標的タンパク質の分解を誘導することができる化合物であれば特に限定されない。オーキシンアナログの例は国際公開第2021/009993号に開示されており、例えば以下の一般式(II)で表される化合物又はそのエステル体、又は以下の一般式(III)で表される化合物であってもよい。 In this specification, the term "auxin analog" is not particularly limited as long as it has a structure similar to any of the above-mentioned auxins, such as natural auxins such as indole-3-acetic acid (IAA) or 4-chloroindole-3-acetic acid (4Cl-IAA), and can bind to a TIR1 family protein to induce degradation of a target protein labeled with a degron. Examples of auxin analogs are disclosed in WO 2021/009993, and may be, for example, a compound represented by the following general formula (II) or an ester thereof, or a compound represented by the following general formula (III).

Figure JPOXMLDOC01-appb-C000002
[式中、Rは、置換基を有していてもよく、環を構成する炭素原子の一部がヘテロ原子で置換されていてもよい環状の脂肪族炭化水素基、又は、置換基を有していてもよく、環を構成する炭素原子の一部がヘテロ原子で置換されていてもよい芳香族炭化水素基である。]
Figure JPOXMLDOC01-appb-C000002
[In the formula, R 1 is a cyclic aliphatic hydrocarbon group which may have a substituent and in which a portion of the carbon atoms constituting the ring may be substituted with a heteroatom, or an aromatic hydrocarbon group which may have a substituent and in which a portion of the carbon atoms constituting the ring may be substituted with a heteroatom.]

Figure JPOXMLDOC01-appb-C000003
[式中、Rは、水素原子、炭素数1~6のアルキル基、又はハロゲン原子を表し、Rは、水素原子、又は炭素数1~6のアルキル基を表す。但し、Rが、水素原子、又は炭素数1~6のアルキル基である場合、Rは、炭素数1~6のアルキル基である。]
Figure JPOXMLDOC01-appb-C000003
[In the formula, R 2 represents a hydrogen atom, an alkyl group having 1 to 6 carbon atoms, or a halogen atom, and R 3 represents a hydrogen atom or an alkyl group having 1 to 6 carbon atoms. However, when R 2 is a hydrogen atom or an alkyl group having 1 to 6 carbon atoms, R 3 is an alkyl group having 1 to 6 carbon atoms.]

 また、オーキシンアナログの具体例としては、以下の式(IV)で示す5-フェニル-インドール3-酢酸(以下、「5-ph-IAA」という)が挙げられる。 Another specific example of an auxin analog is 5-phenyl-indole-3-acetic acid (hereinafter referred to as "5-ph-IAA"), shown in the following formula (IV).

Figure JPOXMLDOC01-appb-C000004
Figure JPOXMLDOC01-appb-C000004

 本明細書において「複数個」とは、2以上の整数、例えば、2~10個、2~7個、2~5個、2~4個又は2~3個の整数をいう。また、本明細書において「数個」とは、例えば、2~10個、2~7個、2~5個、2~4個又は2~3個をいう。 In this specification, "multiple" refers to an integer of 2 or more, for example, 2 to 10, 2 to 7, 2 to 5, 2 to 4, or 2 to 3. In addition, in this specification, "several" refers to, for example, 2 to 10, 2 to 7, 2 to 5, 2 to 4, or 2 to 3.

 本明細書において「アミノ酸同一性(アミノ酸配列同一性)」とは、比較する2つのポリペプチドのアミノ酸配列において、アミノ酸残基の一致数が最大となるように、必要に応じて一方又は双方に適宜ギャップを挿入して整列化(アラインメント)したときに、全アミノ酸残基数における一致アミノ酸残基数の割合(%)をいう。「塩基同一性(塩基配列同一性)」も同様に決定することができる。 As used herein, "amino acid identity (amino acid sequence identity)" refers to the percentage of matching amino acid residues out of the total number of amino acid residues in the amino acid sequences of two polypeptides being compared, when aligned by inserting appropriate gaps into one or both, as necessary, to maximize the number of matching amino acid residues. "Base identity (base sequence identity)" can also be determined in a similar manner.

 本明細書において「アミノ酸の置換」とは、天然のタンパク質を構成する20種類のアミノ酸間の置換をいう。アミノ酸の置換は、好ましくは電荷、側鎖、極性、芳香族性等の性質の類似する保存的アミノ酸群内での置換である。例えば、低極性側鎖を有する無電荷極性アミノ酸群(Gly, Asn, Gln, Ser, Thr, Cys, Tyr)、分枝鎖アミノ酸群(Leu, Val, Ile)、中性アミノ酸群(Gly, Ile, Val, Leu, Ala, Met, Pro)、親水性側鎖を有する中性アミノ酸群(Asn, Gln, Thr, Ser, Tyr, Cys)、酸性アミノ酸群(Asp, Glu)、塩基性アミノ酸群(Arg, Lys, His)、芳香族アミノ酸群(Phe, Tyr, Trp)内での置換が挙げられる。 As used herein, "amino acid substitution" refers to substitutions between the 20 types of amino acids that make up natural proteins. Amino acid substitutions are preferably within conservative amino acid groups that have similar properties such as charge, side chain, polarity, and aromaticity. Examples include substitutions within the uncharged polar amino acids with low polarity side chains (Gly, Asn, Gln, Ser, Thr, Cys, Tyr), branched-chain amino acids (Leu, Val, Ile), neutral amino acids (Gly, Ile, Val, Leu, Ala, Met, Pro), neutral amino acids with hydrophilic side chains (Asn, Gln, Thr, Ser, Tyr, Cys), acidic amino acids (Asp, Glu), basic amino acids (Arg, Lys, His), and aromatic amino acids (Phe, Tyr, Trp).

1-3.構成
 本発明の遺伝子改変マウスは、一対のCreリコンビナーゼ認識配列、一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列、一対のCreリコンビナーゼ認識配列の下流に位置するTIR1ファミリー遺伝子配列、及び一対のCreリコンビナーゼ認識配列の上流に位置するプロモーター配列を含む。
1-3. Configuration The genetically modified mouse of the present invention comprises a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences, a TIR1 family gene sequence located downstream of the pair of Cre recombinase recognition sequences, and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences.

 本明細書において、一対のCreリコンビナーゼ認識配列、一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列、一対のCreリコンビナーゼ認識配列の下流に位置するTIR1ファミリー遺伝子配列、及び一対のCreリコンビナーゼ認識配列の上流に位置するプロモーター配列を含み、一対のCreリコンビナーゼ認識配列間の組換え後にTIR1ファミリー遺伝子を発現することが可能であり、TIR1遺伝子及びその遺伝子発現に必要な発現制御領域からなる配列領域全体を「TIR1遺伝子発現システム」又は単に「遺伝子発現システム」と呼ぶ。したがって、本発明の遺伝子改変マウスは、TIR1遺伝子発現システムを含む。 In this specification, the entire sequence region comprising a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences, a TIR1 family gene sequence located downstream of the pair of Cre recombinase recognition sequences, and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences, which is capable of expressing a TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences, and which is composed of the TIR1 gene and an expression control region necessary for the gene expression, is referred to as a "TIR1 gene expression system" or simply a "gene expression system." Thus, the genetically modified mouse of the present invention comprises a TIR1 gene expression system.

 本発明の遺伝子改変マウスのTIR1遺伝子発現システムでは、プロモーター配列は一対のCreリコンビナーゼ認識配列の上流に位置し、TIR1ファミリー遺伝子配列は一対のCreリコンビナーゼ認識配列の下流に位置する。ここで「上流」及び「下流」は、プロモーターから誘導される転写の方向を基準として定義される。すなわち、プロモーターから転写される転写物において5'側が上流側に、3'側が下流側に対応する。 In the TIR1 gene expression system of the genetically modified mouse of the present invention, the promoter sequence is located upstream of a pair of Cre recombinase recognition sequences, and the TIR1 family gene sequence is located downstream of a pair of Cre recombinase recognition sequences. Here, "upstream" and "downstream" are defined based on the direction of transcription induced from the promoter. In other words, in the transcript transcribed from the promoter, the 5' side corresponds to the upstream side, and the 3' side corresponds to the downstream side.

 本発明の遺伝子改変マウスにおいて、TIR1遺伝子発現システムはゲノム中に含まれてもよく、或いはTIR1遺伝子発現システムを含む発現ベクター(例:プラスミドベクター)等のベクターが遺伝子改変マウスに導入されていてもよい。TIR1遺伝子発現システムがゲノム中に含まれる場合には、ゲノムの任意の位置に導入することが可能であるが、細胞種や発生段階に依らず幅広く発現させることが可能であり、他の内在性遺伝子の発現や機能に影響を与えにくい遺伝子座への導入が好ましく、例えばRosa26遺伝子座やβ-アクチン遺伝子座等が挙げられる。 In the genetically modified mouse of the present invention, the TIR1 gene expression system may be contained in the genome, or a vector such as an expression vector (e.g., a plasmid vector) containing the TIR1 gene expression system may be introduced into the genetically modified mouse. When the TIR1 gene expression system is contained in the genome, it can be introduced into any position in the genome, but it is preferable to introduce it into a gene locus that can be expressed widely regardless of cell type or developmental stage and that is unlikely to affect the expression or function of other endogenous genes, such as the Rosa26 gene locus or the β-actin gene locus.

 一実施形態において、遺伝子改変マウスは、一対のCreリコンビナーゼ認識配列、転写終結polyA配列、TIR1ファミリー遺伝子配列、及びプロモーター配列をRosa26遺伝子座に含む(すなわちTIR1遺伝子発現システムをRosa26遺伝子座に含む)。本明細書において「Rosa26遺伝子座」とは、Gt(ROSA)26Sorとも表記される、マウス第6染色体上のゲノム領域である。Rosa26遺伝子座においてTIR1遺伝子発現システムを導入する位置は、特に制限されない。例えば、TIR1遺伝子発現システムをRosa26遺伝子座のエクソン1とエクソン2の間のイントロン領域に導入することにより、安定な発現を得ることができる。 In one embodiment, the genetically modified mouse comprises a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence, a TIR1 family gene sequence, and a promoter sequence in the Rosa26 locus (i.e., comprises a TIR1 gene expression system in the Rosa26 locus). As used herein, the "Rosa26 locus" is a genomic region on mouse chromosome 6, also referred to as Gt(ROSA)26Sor. The position at which the TIR1 gene expression system is introduced in the Rosa26 locus is not particularly limited. For example, stable expression can be obtained by introducing the TIR1 gene expression system into the intron region between exon 1 and exon 2 of the Rosa26 locus.

 本発明の遺伝子改変マウスにおいて、TIR1ファミリー遺伝子配列は、任意のTIR1ファミリータンパク質をコードする塩基配列又は核酸配列である。例えば、シロイヌナズナ、イネ、ヒャクニチソウ、マツ、シダ、又はヒメツリガネゴケ等に由来する野生型又は変異型のTIR1ファミリータンパク質をコードする塩基配列又は核酸配列が挙げられる。イネ由来の野生型のTIR1ファミリータンパク質をコードする塩基配列又は核酸配列としては、配列番号1で示すアミノ酸配列からなるイネ由来のOsTIR1タンパク質をコードする塩基配列が挙げられ、例えば配列番号2で示す塩基配列からなるOsTIR1遺伝子配列が挙げられる。変異型のTIR1ファミリータンパク質の例としては、配列番号1で示すアミノ酸配列において1個又は数個(例:1~100個、1~50個、1~20個、1~10個、又は1~5個)のアミノ酸が欠失、挿入、置換若しくは付加されたアミノ酸配列、又は配列番号1で示すアミノ酸配列に対して70%以上、80%以上、90%以上、95%以上、98%以上、又は99%以上の同一性を有するアミノ酸配列からなる又はそれを含むタンパク質が挙げられる。このアミノ酸配列の同一性は、配列番号1で示すアミノ酸配列の全長について計算することもできるし、或いは後述するように配列番号1で示すアミノ酸配列の74位を除いて計算することもできる。野生型又は変異型のTIR1ファミリータンパク質は、野生型TIR1と同等以上の活性、すなわちオーキシン又はオーキシンアナログ存在下でデグロンを認識して標的タンパク質を分解に導く活性を有することが好ましい。 In the genetically modified mouse of the present invention, the TIR1 family gene sequence is a nucleotide sequence or nucleic acid sequence encoding any TIR1 family protein. Examples include nucleotide sequences or nucleic acid sequences encoding wild-type or mutant TIR1 family proteins derived from Arabidopsis thaliana, rice, zinnia, pine, fern, or Physcomitrella patens. An example of a nucleotide sequence or nucleic acid sequence encoding a wild-type TIR1 family protein derived from rice is a nucleotide sequence encoding the OsTIR1 protein derived from rice having the amino acid sequence shown in SEQ ID NO:1, for example an OsTIR1 gene sequence having the nucleotide sequence shown in SEQ ID NO:2. Examples of mutant TIR1 family proteins include an amino acid sequence in which one or several (e.g., 1 to 100, 1 to 50, 1 to 20, 1 to 10, or 1 to 5) amino acids have been deleted, inserted, substituted, or added in the amino acid sequence shown in SEQ ID NO: 1, or a protein consisting of or containing an amino acid sequence having 70% or more, 80% or more, 90% or more, 95% or more, 98% or more, or 99% or more identity to the amino acid sequence shown in SEQ ID NO: 1. This identity of the amino acid sequence can be calculated for the entire length of the amino acid sequence shown in SEQ ID NO: 1, or can be calculated for the amino acid sequence shown in SEQ ID NO: 1 excluding position 74, as described below. It is preferable that the wild-type or mutant TIR1 family protein has an activity equivalent to or greater than that of wild-type TIR1, i.e., the activity of recognizing a degron in the presence of auxin or an auxin analog and leading to the degradation of a target protein.

 一実施形態において、本発明の遺伝子改変マウス又はTIR1遺伝子発現システムにおけるTIR1ファミリータンパク質では、配列番号1で示すアミノ酸配列の74位に対応するPhe残基が、Gly残基、Ala残基、又はSer残基に置換されている(本明細書においてそれぞれ「F74G変異」、「F74A変異」、「F74S変異」等という)。配列番号1で示すアミノ酸配列の74位に対応するPhe残基が、Gly残基に置換されたイネ由来TIR1タンパク質としては、配列番号3で表されるアミノ酸配列からなるものが挙げられる。配列番号1で示すアミノ酸配列の74位に対応するPhe残基が、Ala残基に置換されたイネ由来TIR1タンパク質としては、配列番号4で表されるアミノ酸配列からなるものが挙げられる。本実施形態におけるTIR1ファミリータンパク質は、配列番号1で示すアミノ酸配列の74位を除いて、配列番号1で示すアミノ酸配列に対して70%以上、80%以上、90%以上、95%以上、98%以上、又は99%以上の同一性を有するアミノ酸配列からなる又はそれを含むものであってもよい。 In one embodiment, in the TIR1 family protein in the genetically modified mouse or TIR1 gene expression system of the present invention, the Phe residue corresponding to position 74 of the amino acid sequence shown in SEQ ID NO:1 is replaced with a Gly residue, an Ala residue, or a Ser residue (referred to as "F74G mutation", "F74A mutation", "F74S mutation", etc. in the present specification). An example of a rice-derived TIR1 protein in which the Phe residue corresponding to position 74 of the amino acid sequence shown in SEQ ID NO:1 is replaced with a Gly residue is an example of the amino acid sequence shown in SEQ ID NO:3. An example of a rice-derived TIR1 protein in which the Phe residue corresponding to position 74 of the amino acid sequence shown in SEQ ID NO:1 is replaced with an Ala residue is an example of the amino acid sequence shown in SEQ ID NO:4. The TIR1 family protein in this embodiment may be one that consists of or contains an amino acid sequence that has 70% or more, 80% or more, 90% or more, 95% or more, 98% or more, or 99% or more identity to the amino acid sequence shown in SEQ ID NO:1, except for position 74 of the amino acid sequence shown in SEQ ID NO:1.

 本明細書において「デグロン配列」とは、標的タンパク質に付加することによりオーキシンデグロンシステムによる分解を誘導することができるアミノ酸配列をいう。例えば植物由来Aux/IAAファミリータンパク質又はその断片はデグロン配列として機能するため、本発明の遺伝子改変マウス又はTIR1遺伝子発現システムにおいてデグロン配列として使用することができる。 As used herein, the term "degron sequence" refers to an amino acid sequence that can induce degradation by the auxin degron system when added to a target protein. For example, a plant-derived Aux/IAA family protein or a fragment thereof functions as a degron sequence and can therefore be used as a degron sequence in the genetically modified mouse or TIR1 gene expression system of the present invention.

 Aux/IAAファミリータンパク質をコードする遺伝子としては、植物由来のAux/IAAファミリー遺伝子であれば、その種類について特に制限されない。シロイヌナズナ由来のAux/IAAファミリー遺伝子の例としては、IAA1~IAA34遺伝子等が挙げられ、好ましくはIAA17遺伝子であってもよい。シロイヌナズナ由来のAux/IAAファミリー遺伝子の配列は、TAIR(The Arabidopsis Information Resource)に登録されており、必要に応じて入手可能である。デグロン配列として使用できる植物由来Aux/IAAファミリータンパク質の断片は、TIR1ファミリータンパク質/オーキシン又はオーキシンアナログ複合体と結合し、標的タンパク質を分解に導く活性を有することが好ましく、例えば、後述のmAIDの全長又は部分配列であってもよい。 The gene encoding the Aux/IAA family protein is not particularly limited in type, so long as it is a plant-derived Aux/IAA family gene. Examples of Arabidopsis-derived Aux/IAA family genes include the IAA1 to IAA34 genes, and preferably the IAA17 gene. The sequences of Arabidopsis-derived Aux/IAA family genes are registered in TAIR (The Arabidopsis Information Resource) and are available as needed. A fragment of a plant-derived Aux/IAA family protein that can be used as a degron sequence preferably has an activity of binding to a TIR1 family protein/auxin or auxin analog complex and leading to the degradation of a target protein, and may be, for example, a full-length or partial sequence of mAID, which will be described later.

 本明細書において「mAID(Mini-auxin-inducible degron)」とは、Aux/IAAファミリータンパク質の一つであるシロイヌナズナIAA17の部分配列からなるデグロン配列を意味する。mAIDは、Aux/IAAファミリータンパク質のドメインII領域のN末端側及びC末端側に少なくとも2個ずつのLys残基を含む領域からなる配列、又は該配列を2個以上連結してなる配列であり得る。mAIDの具体例としては配列番号5で示すアミノ酸配列が挙げられる。 In this specification, "mAID (Mini-auxin-inducible degron)" refers to a degron sequence consisting of a partial sequence of Arabidopsis thaliana IAA17, which is one of the Aux/IAA family proteins. mAID can be a sequence consisting of a region containing at least two Lys residues on the N-terminal and C-terminal sides of the domain II region of an Aux/IAA family protein, or a sequence consisting of two or more such sequences linked together. A specific example of an mAID is the amino acid sequence shown in SEQ ID NO:5.

 本明細書において「Creリコンビナーゼ認識配列」とは、Creリコンビナーゼによって認識される配列であって、2つの配列間での組換えを誘導できる配列をいう。Creリコンビナーゼ認識配列の具体的な塩基配列は当該技術分野において知られており、例えばloxP配列(ATAACTTCGTATAGCATACATTATACGAAGTTAT、配列番号6);lox511配列(ATAACTTCGTATAGTATACATTATACGAAGTTAT、配列番号7);lox2272配列(ATAACTTCGTATAGGATACTTTATACGAAGTTAT、配列番号8);loxFAS配列(ATAACTTCGTATATACCTTTCTATACGAAGTTAT、配列番号9)等が挙げられる。本発明の遺伝子改変マウス又はTIR1遺伝子発現システムでは、一対のCreリコンビナーゼ認識配列はTIR1ファミリー遺伝子配列とプロモーター配列との間に配置され、一対のCreリコンビナーゼ認識配列の間には転写終結polyA配列が配置される。一対のCreリコンビナーゼ認識配列は、原則として同じ向きに配置される。一対のCreリコンビナーゼ認識配列が同じ向きで存在するとき、Creリコンビナーゼはこの2つの配列を認識してその間で部位特異的組換えを引き起こす結果、両配列で挟まれた配列が切り出されて除去されるためである。 As used herein, the term "Cre recombinase recognition sequence" refers to a sequence that is recognized by Cre recombinase and can induce recombination between two sequences. Specific base sequences of Cre recombinase recognition sequences are known in the art, and examples include loxP sequences (ATAACTTCGTATAGCATACATTATACGAAGTTAT, SEQ ID NO: 6); lox511 sequences (ATAACTTCGTATAGTATACATTATACGAAGTTAT, SEQ ID NO: 7); lox2272 sequences (ATAACTTCGTATAGGATACTTTATACGAAGTTAT, SEQ ID NO: 8); and loxFAS sequences (ATAACTTCGTATATACCTTTCTATACGAAGTTAT, SEQ ID NO: 9). In the genetically modified mouse or TIR1 gene expression system of the present invention, a pair of Cre recombinase recognition sequences is located between the TIR1 family gene sequence and the promoter sequence, and a transcription termination polyA sequence is located between the pair of Cre recombinase recognition sequences. A pair of Cre recombinase recognition sequences are generally arranged in the same direction. When a pair of Cre recombinase recognition sequences are present in the same direction, Cre recombinase recognizes the two sequences and induces site-specific recombination between them, resulting in the excision and removal of the sequence sandwiched between the two sequences.

 本明細書において「転写終結polyA配列」とは、転写の終結を引き起こすことができる塩基配列をいう。転写終結polyA配列は、ポリアデニル化シグナル又は転写ターミネーター等とも呼ばれ、転写終結及び転写産物の3'末端のポリアデニル化を誘導することができる。本発明の遺伝子改変マウス又はTIR1遺伝子発現システムにおいては、転写終結polyA配列は、一対のCreリコンビナーゼ認識配列間に配置され、その配列上で転写を終結させることができる。転写終結polyA配列の種類は特に制限されず、任意の転写終結polyA配列を使用することができるが、例えば、ヒト成長ホルモン(HGH)遺伝子、SV40遺伝子、ウサギβ-グロビン遺伝子、ウシ成長ホルモン(BGH)遺伝子、又はホスホグリセリン酸キナーゼ(PGK)遺伝子に由来する転写終結polyA配列を使用してもよく、真核細胞中で機能し得る公知の任意の転写終結polyA配列を使用してもよい。本発明の遺伝子改変マウス又はTIR1遺伝子発現システムにおいて一対のCreリコンビナーゼ認識配列間には、1つ又は複数の転写終結polyA配列を配置することができる。例えば、2個以上又は3個以上の転写終結polyA配列を配置する場合、その下流に配置された遺伝子のleakyな発現を抑制できるため有利である。 As used herein, the term "transcription termination polyA sequence" refers to a base sequence capable of causing the termination of transcription. The transcription termination polyA sequence is also called a polyadenylation signal or transcription terminator, and can induce the termination of transcription and the polyadenylation of the 3' end of the transcription product. In the genetically modified mouse or TIR1 gene expression system of the present invention, the transcription termination polyA sequence is arranged between a pair of Cre recombinase recognition sequences, and transcription can be terminated on that sequence. The type of transcription termination polyA sequence is not particularly limited, and any transcription termination polyA sequence can be used. For example, a transcription termination polyA sequence derived from the human growth hormone (HGH) gene, the SV40 gene, the rabbit β-globin gene, the bovine growth hormone (BGH) gene, or the phosphoglycerate kinase (PGK) gene may be used, or any known transcription termination polyA sequence that can function in eukaryotic cells may be used. In the genetically modified mouse or TIR1 gene expression system of the present invention, one or more transcription termination polyA sequences can be arranged between a pair of Cre recombinase recognition sequences. For example, placing two or more transcription termination polyA sequences is advantageous because it is possible to suppress leaky expression of genes placed downstream.

 本発明の遺伝子改変マウス又はTIR1遺伝子発現システムにおいて、「プロモーター配列」は、一対のCreリコンビナーゼ認識配列の上流側に配置され、一対のCreリコンビナーゼ認識配列間の組換え後にその下流に位置するTIR1ファミリー遺伝子の発現を駆動することができるプロモーターである。一対のCreリコンビナーゼ認識配列間の組換え前には、上記プロモーター配列から誘導される転写は、上述の転写終結polyA配列上で停止するため、その下流に位置するTIR1ファミリー遺伝子配列は転写されないか又は実質的に転写されない。プロモーター配列の種類は、オーキシンデグロンシステムを機能させる目的の細胞種で遺伝子の発現を駆動できるものであれば特に制限されず、外因性のプロモーター又は内在性のプロモーターのいずれであってもよく、またユビキタスプロモーター(全身性プロモーター)又は部位特異的プロモーターであってもよく、さらに構成的活性型プロモーター、発現誘導型プロモーター、又は時期特異的活性型プロモーターであってもよい。外因性のプロモーターはマウス等の哺乳動物由来のプロモーターや哺乳動物以外に由来するプロモーターであってもよく、例えばCAGプロモーター、CMVプロモーター、SV40プロモーター、EF1aプロモーター、又はRSVプロモーター等を使用することができる。内在性のプロモーターは、TIR1遺伝子発現システムが導入されるゲノム中の遺伝子座の内在性プロモーターであってもよい。例えば、TIR1遺伝子発現システムをRosa26遺伝子座に導入する場合、Rosa26遺伝子の内在性プロモーターにより、TIR1ファミリー遺伝子の発現を駆動してもよい。 In the genetically modified mouse or TIR1 gene expression system of the present invention, the "promoter sequence" is a promoter that is located upstream of a pair of Cre recombinase recognition sequences and can drive the expression of the TIR1 family gene located downstream thereof after recombination between the pair of Cre recombinase recognition sequences. Before recombination between the pair of Cre recombinase recognition sequences, transcription induced from the promoter sequence is terminated on the above-mentioned transcription termination polyA sequence, so that the TIR1 family gene sequence located downstream thereof is not transcribed or is not substantially transcribed. The type of promoter sequence is not particularly limited as long as it can drive the expression of genes in the target cell type in which the auxin degron system is to function, and may be either an exogenous promoter or an endogenous promoter, a ubiquitous promoter (systemic promoter) or a site-specific promoter, or may be a constitutively active promoter, an expression-inducible promoter, or a time-specific active promoter. The exogenous promoter may be a promoter derived from a mammal such as a mouse, or a promoter derived from a non-mammalian organism, and for example, a CAG promoter, a CMV promoter, an SV40 promoter, an EF1a promoter, or an RSV promoter can be used. The endogenous promoter may be an endogenous promoter of a locus in the genome into which the TIR1 gene expression system is introduced. For example, when the TIR1 gene expression system is introduced into the Rosa26 gene locus, the expression of the TIR1 family gene may be driven by the endogenous promoter of the Rosa26 gene.

 本発明の遺伝子改変マウス又はTIR1遺伝子発現システムでは、上記のプロモーター配列は、その下流に位置するTIR1ファミリー遺伝子に、一対のCreリコンビナーゼ認識配列間の組換え後に作動可能に連結されていることが好ましい。本明細書において「作動可能に連結されている」とは、上記のプロモーター配列とTIR1ファミリータンパク質をコードするTIR1ファミリー遺伝子配列との間の機能的連結を意味する。この機能的連結とは、一対のCreリコンビナーゼ認識配列間の組換え後においてTIR1ファミリー遺伝子配列の転写が可能となるような連結をいう。 In the genetically modified mouse or TIR1 gene expression system of the present invention, the above promoter sequence is preferably operably linked to the TIR1 family gene located downstream thereof after recombination between a pair of Cre recombinase recognition sequences. As used herein, "operably linked" refers to a functional link between the above promoter sequence and a TIR1 family gene sequence that encodes a TIR1 family protein. This functional link refers to a link that allows transcription of the TIR1 family gene sequence after recombination between a pair of Cre recombinase recognition sequences.

 一実施形態において、本発明の遺伝子改変マウスは、デグロン配列が付加した標的タンパク質をコードする遺伝子、及び/又はCreリコンビナーゼをコードする遺伝子をさらに含む。 In one embodiment, the genetically modified mouse of the present invention further comprises a gene encoding a target protein to which a degron sequence has been added and/or a gene encoding a Cre recombinase.

 上記の実施形態の遺伝子改変マウスにおいて、デグロン配列が付加される標的タンパク質の種類に制限はない。例えば、外因性のタンパク質であってもよく、或いは内在性のタンパク質であってもよい。標的タンパク質は例えば、疾患原因タンパク質等の創薬標的タンパク質であってもよい。 In the genetically modified mouse of the above embodiment, there is no limit to the type of target protein to which the degron sequence is added. For example, it may be an exogenous protein or an endogenous protein. The target protein may be, for example, a drug discovery target protein such as a disease-causing protein.

 また、標的タンパク質にデグロン配列を付加する位置は限定されず、例えば標的タンパク質のN末端側及び/又はC末端側であってもよく、標的タンパク質のアミノ酸配列の途中への挿入であってもよい。またデグロン配列を付加する数は1個又は複数個であってもよい。デグロン配列が付加した標的タンパク質をコードする遺伝子は、デグロン配列をコードする塩基配列をノックイン等により導入した内在性遺伝子であってもよい。 In addition, the position at which the degron sequence is added to the target protein is not limited, and may be, for example, the N-terminus and/or C-terminus of the target protein, or may be inserted into the middle of the amino acid sequence of the target protein. In addition, the number of degron sequences added may be one or more. The gene encoding the target protein to which the degron sequence has been added may be an endogenous gene into which a base sequence encoding the degron sequence has been introduced by knock-in or the like.

 さらなる実施形態において、標的タンパク質をコードする遺伝子は、発生及び/又は生存に必須である。ここで発生に必須な遺伝子とは、例えばその遺伝子を全身ノックアウトにより破壊した場合に個体が発生異常を示す遺伝子である。また、ここで生存に必須な遺伝子とは、例えばその遺伝子を全身ノックアウトにより破壊した場合に少なくとも一部の個体が致死性を示す遺伝子である。このような遺伝子は従来の全身ノックアウトでは解析することが困難であったが、本発明の遺伝子改変マウスを用いた一過的な機能抑制では解析可能となる。 In a further embodiment, the gene encoding the target protein is essential for development and/or survival. A gene essential for development here is, for example, a gene that causes developmental abnormalities in an individual when the gene is disrupted by whole-body knockout. A gene essential for survival here is, for example, a gene that causes lethality in at least some individuals when the gene is disrupted by whole-body knockout. Such genes were difficult to analyze using conventional whole-body knockout methods, but can be analyzed by transient functional inhibition using the genetically modified mouse of the present invention.

 さらなる実施形態において、標的タンパク質は膜タンパク質である。膜タンパク質は、例えば内在性膜タンパク質、表在性膜タンパク質、又は脂質アンカー型タンパク質であってもよい。 In a further embodiment, the target protein is a membrane protein. The membrane protein may be, for example, an integral membrane protein, a peripheral membrane protein, or a lipid-anchored protein.

 また、上述の実施形態において、Creリコンビナーゼの種類は限定せず、例えば大腸菌P1ファージ由来のCreリコンビナーゼ又はその改変体であり得る。Creリコンビナーゼの改変体の例としては、Cre-ERT及びCre-ERT2等のタモキシフェン誘発性Creリコンビナーゼを挙げることができる。 In the above-mentioned embodiment, the type of Cre recombinase is not limited, and may be, for example, a Cre recombinase derived from Escherichia coli P1 phage or a modified form thereof. Examples of modified Cre recombinase include tamoxifen-inducible Cre recombinases such as Cre-ERT and Cre-ERT2.

 さらなる実施形態において、Creリコンビナーゼをコードする遺伝子は、組織特異的又は細胞種特異的プロモーター等の部位特異的プロモーターの制御下に配置されている。当該技術分野では様々なCreマウスが利用可能であり、目的の組織や細胞種に応じてCreマウスを選択して交配させることによって、部位特異的プロモーターの制御下に配置されているCreリコンビナーゼをコードする遺伝子を遺伝子改変マウスに導入することが可能であり、便利である。 In a further embodiment, the gene encoding the Cre recombinase is placed under the control of a site-specific promoter, such as a tissue-specific or cell type-specific promoter. Various Cre mice are available in the art, and by selecting and mating Cre mice according to the tissue or cell type of interest, it is possible and convenient to introduce the gene encoding the Cre recombinase placed under the control of a site-specific promoter into a genetically modified mouse.

 さらなる実施形態において、部位特異的プロモーターが発現を駆動する部位における標的タンパク質をコードする遺伝子の機能が、発生及び/又は生存に必須である。本実施形態における発生に必須な遺伝子とは、例えばその遺伝子を同じ部位特異的プロモーターの制御下でCreリコンビナーゼを発現させることに基づく条件付きノックアウトにより破壊した場合に個体が発生異常を示す遺伝子である。また、本実施形態における生存に必須な遺伝子とは、例えばその遺伝子を同じ部位特異的プロモーターの制御下でCreリコンビナーゼを発現させることに基づく条件付きノックアウトにより破壊した場合に少なくとも一部の個体が致死性を示す遺伝子である。このような遺伝子は従来の条件付きノックアウトでは解析することが困難であったが、本発明の遺伝子改変マウスを用いた一過的な機能抑制では解析可能となる。 In a further embodiment, the function of the gene encoding the target protein at the site where the expression is driven by the site-specific promoter is essential for development and/or survival. In this embodiment, a gene essential for development is, for example, a gene that causes developmental abnormalities in an individual when the gene is disrupted by conditional knockout based on the expression of Cre recombinase under the control of the same site-specific promoter. In this embodiment, a gene essential for survival is, for example, a gene that causes lethality in at least some individuals when the gene is disrupted by conditional knockout based on the expression of Cre recombinase under the control of the same site-specific promoter. Although such genes were difficult to analyze using conventional conditional knockout, they can be analyzed by transient functional inhibition using the genetically modified mouse of the present invention.

1-4.効果
 本発明の遺伝子改変マウスでは、部位特異的プロモーターによりCreリコンビナーゼの発現が誘導される組織や細胞種において特異的に一対のCreリコンビナーゼ認識配列間の組換えが引き起こされ、それにより一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列がTIR1遺伝子発現システムから除去されて、Creリコンビナーゼ認識配列の下流に配置されたTIR1ファミリー遺伝子の発現が誘導されて、組織又は細胞種特異的にTIR1ファミリータンパク質が発現する。このマウスにオーキシン又はオーキシンアナログを投与することによって、TIR1ファミリータンパク質がオーキシン又はオーキシンアナログと結合し、デグロンで標識された標的タンパク質を宿主細胞のユビキチン/プロテアソーム分解系を利用して分解へと導くことができる。
1-4. Effect In the genetically modified mouse of the present invention, recombination between a pair of Cre recombinase recognition sequences is specifically induced in tissues or cell types where the expression of Cre recombinase is induced by a site-specific promoter, and as a result, the transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences is removed from the TIR1 gene expression system, and the expression of the TIR1 family gene located downstream of the Cre recombinase recognition sequence is induced, resulting in tissue- or cell-type-specific expression of the TIR1 family protein. By administering auxin or an auxin analog to this mouse, the TIR1 family protein binds to the auxin or auxin analog, and the target protein labeled with a degron can be degraded using the ubiquitin/proteasome degradation system of the host cell.

 一方、部位特異的プロモーターによりCreリコンビナーゼの発現が誘導されない組織や細胞種においては、転写終結polyA配列が除去されず、TIR1ファミリータンパク質が実質的に発現しないため、オーキシン又はオーキシンアナログを投与した場合でも標的タンパク質の分解が誘導されない。また、オーキシン又はオーキシンアナログを投与しない場合には、デグロンで標識された標的タンパク質の分解は誘導されない。したがって、本発明の遺伝子改変マウスでは、組織又は細胞種特異的に、極めて高い時間分解能で標的タンパク質の分解を誘導することができる。 On the other hand, in tissues or cell types in which the expression of Cre recombinase is not induced by a site-specific promoter, the transcription termination polyA sequence is not removed and TIR1 family proteins are not substantially expressed, so that even when auxin or an auxin analog is administered, degradation of the target protein is not induced. Furthermore, when auxin or an auxin analog is not administered, degradation of the target protein labeled with a degron is not induced. Therefore, in the genetically modified mouse of the present invention, degradation of the target protein can be induced in a tissue- or cell-type-specific manner with extremely high temporal resolution.

 本発明によれば、本発明の遺伝子改変マウスに発生する、精子、受精卵、又は胚もまた提供される。精子、受精卵、又は胚は、それぞれ凍結精子、凍結受精卵、又は凍結胚であってもよい。また、本発明の遺伝子改変マウスに由来する細胞、例えば本発明の遺伝子改変マウスに由来する後述のマウス細胞も提供される。 The present invention also provides sperm, fertilized eggs, or embryos that develop in the genetically modified mouse of the present invention. The sperm, fertilized eggs, or embryos may be frozen sperm, frozen fertilized eggs, or frozen embryos, respectively. Also provided are cells derived from the genetically modified mouse of the present invention, for example, mouse cells described below that are derived from the genetically modified mouse of the present invention.

2.標的タンパク質分解誘導方法
2-1.概要
 本発明の第2の態様は、標的タンパク質分解方法である。本態様の標的タンパク質分解方法は、オーキシン又はオーキシンアナログを遺伝子改変マウスに投与するか又はマウス細胞に添加することによって、デグロン配列が付加した標的タンパク質の分解を誘導することができる。本態様の方法では、オーキシン又はオーキシンアナログを母親マウスに投与することによって、胎仔又は離乳前の仔マウスにおいて標的タンパク質の分解を誘導することができる。また、疾患モデルマウスにおいて創薬標的候補タンパク質の分解を誘導することもできる。
2. Method for inducing degradation of a target protein 2-1. Overview The second aspect of the present invention is a method for degrading a target protein. The method for degrading a target protein of this aspect can induce degradation of a target protein to which a degron sequence has been added by administering auxin or an auxin analog to a genetically modified mouse or adding it to a mouse cell. In the method of this aspect, degradation of a target protein can be induced in a fetus or a mouse pup before weaning by administering auxin or an auxin analog to a mother mouse. Degradation of a candidate protein as a target for drug discovery can also be induced in a disease model mouse.

2-2.方法
 本発明の標的タンパク質分解方法は、遺伝子改変マウス(マウス個体)又は遺伝子改変マウスの由来するマウス細胞を対象として実施することができる。本発明の標的タンパク質分解方法の具体的な構成は、対象がマウス個体か或いはマウス細胞かによって異なる。遺伝子改変マウスを対象とする場合には、本発明の標的タンパク質分解方法は、オーキシン又はオーキシンアナログを遺伝子改変マウスに投与して、デグロン配列が付加した標的タンパク質の分解を誘導する、分解誘導工程を必須工程として含み、分解誘導工程後の遺伝子改変マウスの表現型を評価する評価工程を選択工程として含む。マウス細胞を対象とする場合には、本発明の標的タンパク質分解方法は、マウス細胞を培養する培養工程、及びオーキシン又はオーキシンアナログをマウス細胞に添加して、デグロン配列が付加した標的タンパク質の分解を誘導する、分解誘導工程を必須工程として含み、分解誘導工程後のマウス細胞の表現型を評価する評価工程を選択工程として含む。
2-2. Method The target protein degradation method of the present invention can be carried out on a genetically modified mouse (mouse individual) or mouse cells derived from a genetically modified mouse. The specific configuration of the target protein degradation method of the present invention differs depending on whether the target is a mouse individual or mouse cells. When a genetically modified mouse is used as a target, the target protein degradation method of the present invention includes, as an essential step, a degradation induction step in which auxin or an auxin analog is administered to the genetically modified mouse to induce degradation of a target protein to which a degron sequence has been added, and includes, as a selection step, an evaluation step in which the phenotype of the genetically modified mouse after the degradation induction step is evaluated. When a mouse cell is used as a target, the target protein degradation method of the present invention includes, as an essential step, a culture step in which mouse cells are cultured, and a degradation induction step in which auxin or an auxin analog is added to the mouse cell to induce degradation of a target protein to which a degron sequence has been added, and includes, as a selection step, an evaluation step in which the phenotype of the mouse cell after the degradation induction step is evaluated.

(分解誘導工程)
 本発明の標的タンパク質分解方法において「分解誘導工程」とは、遺伝子改変マウスを対象とする場合には、オーキシン又はオーキシンアナログを遺伝子改変マウスに投与して、デグロン配列が付加した標的タンパク質の分解を誘導する工程であり、マウス細胞を対象とする場合には、オーキシン又はオーキシンアナログを前記マウス細胞に添加して、デグロン配列が付加した標的タンパク質の分解を誘導する工程である。
(Decomposition induction process)
In the target protein degradation method of the present invention, the "degradation inducing step" refers, when a genetically modified mouse is the target, to a step of administering auxin or an auxin analogue to the genetically modified mouse to induce degradation of the target protein to which a degron sequence has been added, and, when a mouse cell is the target, to a step of adding auxin or an auxin analogue to the mouse cell to induce degradation of the target protein to which a degron sequence has been added.

 本態様において、遺伝子改変マウス又はマウス細胞は、TIR1ファミリー遺伝子発現システム、前記デグロン配列が付加した標的タンパク質をコードする遺伝子、及びCreリコンビナーゼをコードする遺伝子を含む。ここで、TIR1ファミリー遺伝子発現システムは、一対のCreリコンビナーゼ認識配列、前記一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列、前記一対のCreリコンビナーゼ認識配列の下流に位置する、Transport Inhibitor Response 1(TIR1)ファミリータンパク質をコードするTIR1ファミリー遺伝子配列、及び前記一対のCreリコンビナーゼ認識配列の上流に位置し、前記一対のCreリコンビナーゼ認識配列間の組換え後に前記TIR1ファミリー遺伝子の発現を駆動することができるプロモーター配列を含む。一対のCreリコンビナーゼ認識配列、転写終結polyA配列、TIR1ファミリー遺伝子配列、及びプロモーター配列の構成は第1態様で詳細に説明しているため、ここでの説明は省略する。 In this embodiment, the genetically modified mouse or mouse cell comprises a TIR1 family gene expression system, a gene encoding a target protein to which the degron sequence has been added, and a gene encoding a Cre recombinase. Here, the TIR1 family gene expression system comprises a pair of Cre recombinase recognition sequences, a transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences, a TIR1 family gene sequence encoding a Transport Inhibitor Response 1 (TIR1) family protein located downstream of the pair of Cre recombinase recognition sequences, and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences and capable of driving expression of the TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences. The configurations of the pair of Cre recombinase recognition sequences, the transcription termination polyA sequence, the TIR1 family gene sequence, and the promoter sequence are described in detail in the first embodiment, and therefore will not be described here.

 本発明の標的タンパク質分解方法において、遺伝子改変マウスの発生段階や週齢/胎齢は問わない。例えば、胎仔マウス、周産期マウス、新生仔マウス、離乳前マウス、若年成体マウス、又は成体マウスのいずれであってもよい。 In the method for degrading a target protein of the present invention, the developmental stage or week/fetal age of the genetically modified mouse does not matter. For example, the genetically modified mouse may be a fetal mouse, a perinatal mouse, a newborn mouse, a pre-weaning mouse, a young adult mouse, or an adult mouse.

 遺伝子改変マウスへの投与方法は、標的タンパク質の分解を誘導する組織や細胞種へのオーキシン又はオーキシンアナログの送達が可能な投与方法であれば、限定しない。例えば、静脈内投与(尾静脈注射)、腹腔内投与、又は皮下投与等を挙げることができる。胎仔マウスや離乳前のマウスであれば、分解を誘導する対象となるそのマウスに直接投与する以外に、その母親マウスにオーキシン又はオーキシンアナログを投与することにより、胎盤を通過する血漿や、授乳される乳を介して間接的に投与することができる。例えば、胎仔マウスであれば妊娠マウスへの投与、離乳前の仔マウスであれば授乳中の雌マウスにオーキシン又はオーキシンアナログを投与することにより、胎仔マウス又は仔マウスへの間接的な投与が可能である。  The method of administration to genetically modified mice is not limited as long as it is capable of delivering auxin or auxin analogues to tissues or cell types that induce degradation of the target protein. Examples include intravenous administration (tail vein injection), intraperitoneal administration, and subcutaneous administration. In the case of fetal mice or preweaned mice, in addition to administering directly to the mouse in which degradation is to be induced, auxin or auxin analogues can be administered indirectly to the mother mouse via the plasma that crosses the placenta or the milk that is fed. For example, in the case of fetal mice, auxin or auxin analogues can be administered to pregnant mice, and in the case of preweaned pups, auxin or auxin analogues can be administered indirectly to fetal mice or pups by administering them to lactating female mice.

 遺伝子改変マウスへのオーキシン又はオーキシンアナログの投与回数は制限されない。例えば単回投与又は複数回投与であってもよく、例えば1回、2回、3回、4回、又はそれ以上であってもよい。また複数回投与は周期的な投与、例えば1日毎、2日毎、3日毎、又は1週間毎の繰り返し投与であってもよい。マウス細胞へのオーキシン又はオーキシンアナログの添加回数も同様である。 The number of times the auxin or auxin analogue is administered to the genetically modified mouse is not limited. For example, it may be a single administration or multiple administrations, for example, once, twice, three times, four times, or more. The multiple administrations may also be periodic administrations, for example repeated administrations every day, every two days, every three days, or once a week. The same applies to the number of times the auxin or auxin analogue is added to the mouse cells.

 遺伝子改変マウスを対象とする場合のオーキシン又はオーキシンアナログの投与量は、標的タンパク質の分解を誘導可能であり、かつ遺伝子改変マウスに有害な影響を及ぼさない範囲であれば特に制限されず、例えば遺伝子改変マウスの体重に基づいて0.001 mg/kg体重~10 g/kg体重、0.01 mg/kg体重~1 g/kg体重、又は0.1 mg/kg体重~50 mg/kg体重であってもよく、好ましくは0.5 mg/kg体重~10 mg/kg体重又は1 mg/kg体重~5 mg/kg体重であり得る。一回の投与量は特に制限されないが、例えば0.02 mg~200 g、0.2 mg~20 g、又は2 mg~1 gであってもよく、好ましくは10 mg~200 mg又は20 mg~100 mgであり得る。投与量は、例えば所望する分解の程度に応じて調節することが可能であり、高い分解レベルを目的とする投与量としては5 mg/kg体重以上又は10 mg/kg体重以上、低い分解レベルを目的とする投与量としては2 mg/kg体重以下又は1 mg/kg体重以下が例示される。マウス細胞へのオーキシン又はオーキシンアナログの添加量は、標的タンパク質の分解を誘導可能であり、かつマウス細胞に有害な影響を及ぼさない範囲であれば特に制限されず、例えば0.001 mg/L~10 g/L、0.01 mg/L~1 g/L、又は0.1 mg/L~50 mg/Lであってもよい。 When a genetically modified mouse is used as the subject, the dosage of the auxin or auxin analogue is not particularly limited as long as it is capable of inducing degradation of the target protein and does not have a deleterious effect on the genetically modified mouse, and may be, for example, 0.001 mg/kg to 10 g/kg body weight, 0.01 mg/kg to 1 g/kg body weight, or 0.1 mg/kg to 50 mg/kg body weight based on the body weight of the genetically modified mouse, and preferably 0.5 mg/kg to 10 mg/kg body weight or 1 mg/kg to 5 mg/kg body weight. The dosage per administration is not particularly limited, and may be, for example, 0.02 mg to 200 g, 0.2 mg to 20 g, or 2 mg to 1 g, and preferably 10 mg to 200 mg or 20 mg to 100 mg. The dosage can be adjusted depending on, for example, the desired degree of degradation; for example, a dosage aimed at a high level of degradation is 5 mg/kg body weight or more or 10 mg/kg body weight or more, and for example, a dosage aimed at a low level of degradation is 2 mg/kg body weight or less or 1 mg/kg body weight or less. The amount of auxin or auxin analogue added to mouse cells is not particularly limited as long as it is capable of inducing degradation of the target protein and does not have a deleterious effect on the mouse cells, and may be, for example, 0.001 mg/L to 10 g/L, 0.01 mg/L to 1 g/L, or 0.1 mg/L to 50 mg/L.

(評価工程)
 本発明の標的タンパク質分解方法において「評価工程」とは、遺伝子改変マウスを対象とする場合には、分解誘導工程後の前記遺伝子改変マウスの表現型を評価する工程であり、マウス細胞を対象とする場合には、分解誘導工程後のマウス細胞の表現型を評価する工程である。
(Evaluation process)
In the target protein degradation method of the present invention, the "evaluation step" refers to a step of evaluating the phenotype of a genetically modified mouse after the degradation induction step, when a genetically modified mouse is the target, and a step of evaluating the phenotype of mouse cells after the degradation induction step, when mouse cells are the target.

 本明細書において「遺伝子改変マウスの表現型」は、マウス個体が示す観察可能な特徴、又はマウス個体から採取した器官、組織、細胞、体液等が示す観察可能な特徴であれば特に制限されない。個体レベルの表現型であれば、全身及び身体部位の外観又は形状;体重、体脂肪率、心電図、握力等の筋力;又は歩行等の運動、感覚機能、学習、情報処理、食餌、排泄等(行動表現型)であってもよい。器官又は組織の表現型は制限されず、例えば皮膚、心臓、肝臓、腸等の臓器、脳等の中枢神経系、末梢神経系において観察可能な特徴であってよく、任意の器官又は組織の細胞において観察可能な特徴であってもよく、血液、尿等の体液において観察可能な特徴(例えば、体液中におけるタンパク質、脂質、核酸等の生体分子)であってもよい。 In this specification, the "phenotype of a genetically modified mouse" is not particularly limited as long as it is an observable characteristic exhibited by an individual mouse, or an observable characteristic exhibited by an organ, tissue, cell, body fluid, etc. taken from an individual mouse. The individual-level phenotype may be the appearance or shape of the whole body and body parts; weight, body fat percentage, electrocardiogram, muscle strength such as grip strength; or movement such as walking, sensory function, learning, information processing, feeding, excretion, etc. (behavioral phenotype). The phenotype of an organ or tissue is not limited, and may be, for example, a characteristic observable in an organ such as the skin, heart, liver, intestine, or the central nervous system or peripheral nervous system such as the brain, a characteristic observable in the cells of any organ or tissue, or a characteristic observable in a body fluid such as blood or urine (for example, biomolecules such as proteins, lipids, and nucleic acids in a body fluid).

 本明細書において「マウス細胞の表現型」は、細胞全体、細胞内部(例:細胞質、核、ミトコンドリア、小胞体、ゴルジ体等の細胞小器官)又は細胞表層(例:細胞膜、細胞表面)を問わず細胞において観察可能な特徴であれば制限されないが、例えば、細胞の増殖能やマーカーの検出量;遺伝子(例:mRNA、miRNA等の核酸)やタンパク質の発現量又は活性;又は抗原や代謝産物の量等であってもよい。マウス細胞の細胞種は限定されず、体細胞又は生殖細胞であってもよく、また幹細胞、前駆細胞、未分化細胞、分化細胞、又は株化細胞(例:初代培養細胞、がん細胞)であってもよい。分化細胞の一例としては、線維芽細胞、上皮細胞、肝細胞、血液細胞、免疫細胞、間葉系細胞、神経細胞、筋肉細胞等を挙げることができる。 In this specification, the "phenotype of mouse cells" is not limited to any characteristic that can be observed in a cell, whether it is the whole cell, the inside of the cell (e.g., organelles such as the cytoplasm, nucleus, mitochondria, endoplasmic reticulum, and Golgi apparatus), or the cell surface (e.g., cell membrane and cell surface), but may be, for example, the proliferation ability of a cell or the detected amount of a marker; the expression level or activity of a gene (e.g., nucleic acid such as mRNA or miRNA) or a protein; or the amount of an antigen or a metabolite. The type of mouse cells is not limited, and may be somatic cells or germ cells, or may be stem cells, precursor cells, undifferentiated cells, differentiated cells, or established cell lines (e.g., primary culture cells, cancer cells). Examples of differentiated cells include fibroblasts, epithelial cells, hepatocytes, blood cells, immune cells, mesenchymal cells, nerve cells, and muscle cells.

 表現型の具体的な評価方法は、特に制限されず、評価対象とする表現型の種類に応じて選択することができる。評価方法や評価手段の具体例としては、血清生化学検査、免疫検査、心電図測定、体重測定、体脂肪測定、臓器重量測定、行動観察、形態観察、Y迷路、握力試験、オープンフィールド試験、フローサイトメトリー、ノザンブロッティング、in situハイブリダイゼーション、マイクロアレイ、RNA seq、RT-PCR等のPCR、ELISA、免疫組織化学、ウェスタンブロッティング等を挙げることができる。 The specific method of evaluating a phenotype is not particularly limited and can be selected according to the type of phenotype to be evaluated. Specific examples of evaluation methods and means include serum biochemistry tests, immunological tests, electrocardiograms, body weight measurements, body fat measurements, organ weight measurements, behavioral observations, morphological observations, Y-mazes, grip strength tests, open field tests, flow cytometry, northern blotting, in situ hybridization, microarrays, RNA seq, PCR such as RT-PCR, ELISA, immunohistochemistry, and western blotting.

 評価工程は分解誘導工程後の任意の時点で行うことができる。例えば、分解誘導工程後の1分後~1か月後、30分後~1週間後、1時間後~3日後、2時間後~1日後、3時間後~12時間後、5時間後~10時間後、7時間後~8時間後に評価工程を行ってもよく、3時間以上後に行うことが好ましい。なお、評価工程は、分解誘導工程後に標的タンパク質が分解されている期間において行ってもよく、又は標的タンパク質が分解されて元の発現レベルが回復した後に行ってもよい。目的とする表現型に応じて評価工程を行う時点を選択すればよい。 The evaluation step can be carried out at any time after the degradation induction step. For example, the evaluation step may be carried out 1 minute to 1 month, 30 minutes to 1 week, 1 hour to 3 days, 2 hours to 1 day, 3 hours to 12 hours, 5 hours to 10 hours, 7 hours to 8 hours after the degradation induction step, and preferably 3 hours or more later. The evaluation step may be carried out during the period in which the target protein is degraded after the degradation induction step, or may be carried out after the target protein has been degraded and the original expression level has been restored. The time point for carrying out the evaluation step may be selected depending on the desired phenotype.

 本態様の標的タンパク質分解方法の一実施形態において、標的タンパク質は、創薬標的候補タンパク質である。本明細書において「創薬標的候補タンパク質」とは、創薬標的となるタンパク質を探索する際に候補となるタンパク質をいう。具体的には、その分解を誘導することによって疾患又は疾患モデルに対する治療効果又は予防効果や副作用の低減や回避といった目的の効果が得られることが期待される候補タンパク質である。本実施形態では、遺伝子改変マウスは疾患モデルマウスであってもよい。創薬標的候補タンパク質の具体的な種類は制限されない。例えば、創薬標的候補タンパク質をコードする遺伝子の全身ノックアウト又は条件付きノックアウトに基づく破壊が、前記疾患モデルマウスに対する治療又は予防効果を示さないか、又は(有害な)副作用(例:致死性)を示したため、これまでの創薬において有益な効果が見逃されていたタンパク質であってもよい。 In one embodiment of the target protein degradation method of this aspect, the target protein is a candidate drug discovery target protein. In this specification, the term "candidate drug discovery target protein" refers to a protein that is a candidate when searching for a protein that is a target for drug discovery. Specifically, it is a candidate protein whose degradation is expected to produce a desired effect, such as a therapeutic or preventive effect on a disease or disease model, or a reduction or avoidance of side effects. In this embodiment, the genetically modified mouse may be a disease model mouse. The specific type of the candidate drug discovery target protein is not limited. For example, it may be a protein whose beneficial effect has been overlooked in previous drug discovery because destruction based on whole-body knockout or conditional knockout of the gene encoding the candidate drug discovery target protein did not show a therapeutic or preventive effect on the disease model mouse or showed (harmful) side effects (e.g., lethality).

 本明細書において「疾患」は制限されず、例えばがん、肝炎、感染症等であってもよい。また、本明細書において「がん」の種類は限定しないが、例えば、腺がん、扁平上皮がん、小細胞がん及び大細胞がん等が挙げられる。具体的ながんの種類としては、例えば、悪性黒色腫、甲状腺がん、肺がん、乳がん、食道がん、胃がん、大腸がん、小腸がん、前立腺がん、卵巣がん、腎がん、肝がん、膵がん、胆道がん、脳腫瘍、頭頸部がん、中皮腫、骨肉腫、血液がん、リンパ腫、骨髄腫等が挙げられる。 In this specification, the term "disease" is not limited, and may be, for example, cancer, hepatitis, infectious disease, etc. Furthermore, the type of "cancer" is not limited, and examples include adenocarcinoma, squamous cell carcinoma, small cell carcinoma, and large cell carcinoma. Specific types of cancer include, for example, malignant melanoma, thyroid cancer, lung cancer, breast cancer, esophageal cancer, gastric cancer, colon cancer, small intestine cancer, prostate cancer, ovarian cancer, kidney cancer, liver cancer, pancreatic cancer, biliary tract cancer, brain tumor, head and neck cancer, mesothelioma, osteosarcoma, blood cancer, lymphoma, myeloma, etc.

 さらなる実施形態では、本態様の標的タンパク質分解方法は、創薬標的候補タンパク質とは異なる因子を標的とする薬剤を前記遺伝子改変マウスに投与する工程をさらに含む。本実施形態において「創薬標的候補タンパク質とは異なる因子」とは、創薬標的候補タンパク質と組み合わせて標的とすることによって、疾患又は疾患モデルに対する治療効果又は予防効果や副作用の低減や回避といった目的の効果を増強させることが期待される因子であり、例えば創薬標的候補タンパク質とは異なるタンパク質や核酸等の任意の生体物質である。創薬標的候補タンパク質とは異なる因子を標的とする薬剤は、制限されず、低分子化合物、抗体医薬、siRNAやアンチセンス核酸等の核酸医薬であってよく、その投与はオーキシン又はオーキシンアナログの投与と同時であってもなくてもよい。本実施形態では、評価工程は、創薬標的候補タンパク質と前記因子との両者を標的とした場合の表現型と、前記因子のみを標的とした場合の表現型とを比較することができる。この比較によって、創薬標的候補タンパク質とは異なる因子を標的とする薬剤の投与と組み合わせて創薬標的候補タンパク質の分解を誘導することにより、疾患に対する治療効果又は予防効果や副作用の低減や回避といった目的の効果を増強させる効果の程度や有無を判定することができる。 In a further embodiment, the target protein degradation method of this aspect further includes a step of administering to the genetically modified mouse a drug targeting a factor other than the candidate drug target protein. In this embodiment, the "factor other than the candidate drug target protein" is a factor that is expected to enhance the desired effect, such as a therapeutic or preventive effect on a disease or disease model, or a reduction or avoidance of side effects, by targeting it in combination with the candidate drug target protein, and is, for example, any biological substance such as a protein or nucleic acid other than the candidate drug target protein. The drug targeting a factor other than the candidate drug target protein is not limited and may be a low molecular weight compound, an antibody drug, or a nucleic acid drug such as siRNA or antisense nucleic acid, and may be administered simultaneously with or without the administration of auxin or auxin analog. In this embodiment, the evaluation step can compare the phenotype when both the candidate drug target protein and the factor are targeted with the phenotype when only the factor is targeted. This comparison makes it possible to determine the degree and presence of an effect of enhancing the desired effect, such as a therapeutic or preventive effect against a disease or a reduction or avoidance of side effects, by inducing the degradation of a candidate drug target protein in combination with the administration of a drug that targets a factor different from the candidate drug target protein.

2-3.効果
 本発明の標的タンパク質分解方法では、標的タンパク質の分解をマウス生体内又はマウス細胞において極めて高い時間分解能で制御することができるため、一過的な分解とその後の迅速な回復を達成することができる。
2-3. Effects The method for degrading a target protein of the present invention can control degradation of a target protein in a living mouse body or in mouse cells with extremely high time resolution, thereby achieving transient degradation and subsequent rapid recovery.

 従来の全身ノックアウトや条件付きノックアウトでは、候補遺伝子を破壊することにより長期に亘って候補タンパク質の機能が阻害される。この場合のマウスの表現型と、実際に開発した治療薬を動物に投与して一過的にタンパク質の機能を制御する場合の表現型とは必ずしも一致しない場合があり、実際に開発した治療薬を投与して初めて予想外の副作用が明らかになる場合もある。このような場合、治療開発に要した膨大な労力やコストの投資が無駄になってしまう。これに対して本発明の標的タンパク質分解方法では、一過的にタンパク質の機能を制御することによる効果を観察できるため、開発予定の治療薬が奏する実際の効果を創薬候補タンパク質の探索過程においてより正確に事前検証することが可能となり、無駄な投資コストを削減し、創薬の初期段階を効率化することができる。 In conventional whole-body knockout and conditional knockout, the function of the candidate protein is inhibited for a long period of time by destroying the candidate gene. The phenotype of the mouse in this case may not necessarily match the phenotype when the developed therapeutic drug is actually administered to the animal to transiently control the function of the protein, and unexpected side effects may only become apparent after the developed therapeutic drug is actually administered. In such cases, the enormous investment of labor and cost required for therapeutic development is wasted. In contrast, the targeted protein degradation method of the present invention makes it possible to observe the effects of transiently controlling the function of a protein, making it possible to more accurately verify in advance the actual effects of the therapeutic drug to be developed in the process of searching for candidate proteins for drug discovery, reducing unnecessary investment costs and streamlining the initial stages of drug discovery.

 また、全身ノックアウトや条件付きノックアウトによる長期的な発現抑制では(補償機構の誘導等によって)認められないが、一過的な分解制御によって初めて認められる効果があり得ることも十分に想定される。したがって、従来の全身ノックアウトや条件付きノックアウトでは治療効果等の効果が得られなかった候補タンパク質を本発明の方法の標的とすることにより、これまでの創薬初期段階で見落とされていた有用な候補タンパク質を特定することもできる。 It is also entirely conceivable that there may be effects that cannot be seen with long-term expression suppression using whole-body knockout or conditional knockout (due to the induction of compensatory mechanisms, etc.), but that can only be seen through transient degradation control. Therefore, by targeting candidate proteins for which no therapeutic or other effects could be obtained using conventional whole-body knockout or conditional knockout with the method of the present invention, it is also possible to identify useful candidate proteins that have been overlooked in the early stages of drug discovery.

<実施例1:Rosa26OsTIR1(F74G)マウス系統の樹立>
(目的)
 OsTIR1(F74G)タンパク質の細胞種特異的な発現を可能とするRosa26OsTIR1(F74G)マウス系統を作製する。
Example 1: Establishment of Rosa26 OsTIR1(F74G) mouse line
(the purpose)
We will generate the Rosa26 OsTIR1(F74G) mouse line, which allows cell type-specific expression of the OsTIR1(F74G ) protein.

(方法と結果)
 イネ由来のTransport Inhibitor Response 1(TIR1)ファミリータンパク質であるOsTIR1タンパク質のアミノ酸配列(配列番号1)において74位のPhe残基がGly残基に置換されたOsTIR1(F74G)タンパク質をコードするcDNA配列(配列番号10)及びires-EGFP配列を、CAGプロモーター(pCAG)及びloxP-neor-STOP-loxP配列(配列番号11)の下流に配置したTarget Vectorを作製した(図1A)。このTarget VectorをES細胞に導入し、Rosa26遺伝子のExon 1とExon 2との間のイントロン領域で相同組換えを起こしたESクローンを同定した(図1B)。このESクローンからキメラマウスを作製し、このキメラマウスから目的のRosa26OsTIR1(F74G)アレルを有するF1ファウンダーマウスを得ることによって、Rosa26OsTIR1(F74G)マウス系統を樹立した。
(Methods and Results)
We constructed a target vector in which the cDNA sequence (SEQ ID NO: 10) encoding the OsTIR1(F74G) protein, a rice-derived Transport Inhibitor Response 1 (TIR1) family protein in which the Phe residue at position 74 in the amino acid sequence of the OsTIR1 protein (SEQ ID NO: 1) was replaced with a Gly residue, and the ires-EGFP sequence was placed downstream of a CAG promoter (pCAG) and a loxP-neor-STOP-loxP sequence (SEQ ID NO: 11) (Fig. 1A). We introduced this target vector into ES cells and identified ES clones that had undergone homologous recombination in the intron region between Exon 1 and Exon 2 of the Rosa26 gene (Fig. 1B). We generated chimeric mice from these ES clones, and by obtaining F1 founder mice carrying the desired Rosa26 OsTIR1(F74G) allele from these chimeric mice, we established the Rosa26 OsTIR1(F74G) mouse line.

 作製したRosa26OsTIR1(F74G)アレルからのOsTIR1(F74G)タンパク質の発現には、CreリコンビナーゼによるloxP配列の部位特異的組換えによってloxP-neor-STOP-loxP配列が除去されることが必要であり、特定の組織又は細胞種特異的にCreリコンビナーゼを発現するトランスジェニック(Tg)マウスを選定して掛け合わせることにより、組織又は細胞種特異的にOsTIR1(F74G)タンパク質を発現させることができる(図2)。 Expression of the OsTIR1 (F74G) protein from the Rosa26 OsTIR1 (F74G) allele requires the removal of the loxP-neor-STOP-loxP sequence by site-specific recombination of the loxP sequence using Cre recombinase. By selecting and crossing transgenic (Tg) mice expressing Cre recombinase in a tissue- or cell-specific manner, OsTIR1(F74G) protein can be expressed in a tissue- or cell-specific manner (Fig. 2).

<実施例2:マウス生体内におけるタンパク質分解の時系列解析>
(目的)
 mini AIDデグロン配列(mAID)を融合させた標的タンパク質を発現するマウスを作製し、このマウスに実施例1で作製したRosa26OsTIR1(F74G)マウス、及びCreリコンビナーゼを発現するTgマウスを交配させる。交配により得られたマウスにオーキシンアナログを投与して標的タンパク質の分解を誘導し、生体内においてタンパク質分解の時間変化を解析する。
Example 2: Time series analysis of protein degradation in vivo in mice
(the purpose)
Mice expressing a target protein fused with a mini AID degron sequence (mAID) are generated, and these mice are crossed with the Rosa26 OsTIR1(F74G) mice generated in Example 1 and Tg mice expressing Cre recombinase. An auxin analog is administered to the mice obtained by crossing to induce degradation of the target protein, and the time course of protein degradation in vivo is analyzed.

(方法と結果)
 ゲノム編集法を用いてVenus蛍光タンパク質及びmini AIDデグロン配列(mAID)をコードする塩基配列をSatb1遺伝子に導入することによって、Satb1核タンパク質のN末端側にVenus蛍光タンパク質及びmini AIDデグロン配列(mAID)を融合させた融合タンパク質を発現するSatb1Venus-mAIDマウスを作製した(図3A)。
(Methods and Results)
By using genome editing to introduce the base sequence encoding the Venus fluorescent protein and the mini AID degron sequence (mAID) into the Satb1 gene, we generated Satb1 Venus-mAID mice that express a fusion protein in which the Venus fluorescent protein and the mini AID degron sequence (mAID) are fused to the N-terminus of the Satb1 nuclear protein (Fig. 3A).

 次に、実施例1で作製したRosa26OsTIR1(F74G)マウス、Satb1Venus-mAIDマウス、及びT細胞特異的にCreリコンビナーゼを発現するCd4Cre Tgマウス(4~8週齢の雄又は雌;ワシントン大学Chris Wilson博士より供与)を交配させることによって、Rosa26OsTIR1/+:Satb1Venus-mAID/+:Cd4Creマウスを作製した(図3A)。 Next, the Rosa26 OsTIR1(F74G) mice prepared in Example 1, Satb1 Venus-mAID mice, and Cd4Cre Tg mice (4-8 week-old male or female; provided by Dr. Chris Wilson, University of Washington) that express Cre recombinase specifically in T cells were crossed to generate Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice (Figure 3A).

 Rosa26OsTIR1/+:Satb1Venus-mAID/+:Cd4Creマウスに対して、0.1 mg/body又は0.02 mg/bodyの用量で5-フェニル-インドール3-酢酸(以下、「5-ph-IAA」と略記する)を腹腔内投与(intra-peritoneal administration;i.p.)した。投与から0~72時間の各時点でマウスから経時的に採血し、抹消血中のT細胞でのSatb1-Venusの蛍光強度をフローサイトメトリーにより解析した。 5-Phenyl-indole-3-acetic acid (5-ph-IAA) was administered intraperitoneally (ip) to Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice at a dose of 0.1 mg/body or 0.02 mg/body. Blood was collected from the mice at each time point from 0 to 72 hours after administration, and the fluorescence intensity of Satb1-Venus in T cells in the peripheral blood was analyzed by flow cytometry.

 結果を図3及び図4に示す。0.1 mg/bodyの用量で投与した場合、Satb1-Venusの発現は、5-ph-IAAの投与から2~3時間後に低下し始めて6~8時間後に消失し、32~48時間後に回復し始めて72時間後にほぼ完全な回復を示した(図3B、図4)。0.02 mg/bodyの用量で投与した場合には、Satb1-Venusの発現は、5-ph-IAAの投与から2~3時間後に低下し始めて5~8時間後に大幅な低下を示し、24時間後以降は回復した(図3C、図4)。 The results are shown in Figures 3 and 4. When administered at a dose of 0.1 mg/body, Satb1-Venus expression began to decrease 2-3 hours after administration of 5-ph-IAA, disappeared 6-8 hours later, began to recover 32-48 hours later, and showed almost complete recovery after 72 hours (Figures 3B and 4). When administered at a dose of 0.02 mg/body, Satb1-Venus expression began to decrease 2-3 hours after administration of 5-ph-IAA, showed a significant decrease 5-8 hours later, and recovered after 24 hours (Figures 3C and 4).

 以上の結果から、Rosa26OsTIR1/+:Satb1Venus-mAID/+:Cd4Creマウスへの5-ph-IAA投与によって、マウス生体内における標的タンパク質の一過的な分解及びその後の迅速な回復が達成された。さらに、5-ph-IAAの投与量を調節することによって、タンパク質分解の程度を制御可能であることが示された。 These results suggest that administration of 5-ph-IAA to Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice resulted in transient degradation of the target protein in vivo and rapid recovery afterwards. Furthermore, it was shown that the degree of protein degradation could be controlled by adjusting the dose of 5-ph-IAA.

<実施例3:胎仔マウス及び新生仔マウスにおけるタンパク質分解の誘導>
(目的)
 胎仔マウス及び新生仔マウスにおける標的タンパク質分解の効果を検証するために、妊娠中又は授乳中の雌マウスに5-ph-IAAを投与し、胎仔又は新生仔マウスにおいて標的タンパク質の分解を解析する。
Example 3: Induction of protein degradation in fetal and neonatal mice
(the purpose)
To verify the effect of target protein degradation in fetal and neonatal mice, 5-ph-IAA will be administered to pregnant or lactating female mice, and the degradation of target proteins will be analyzed in fetal or neonatal mice.

(方法と結果)
 実施例2で作製したRosa26OsTIR1/+:Satb1Venus-mAID/+:Cd4Creマウスについて、妊娠中(妊娠17.5日目)のマウス又は出産後2日目の授乳中の雌マウスに0.1 mg/bodyの用量で5-ph-IAAを腹腔内投与した。投与から24時間後、雌マウスの胎仔マウス又は雌マウスが授乳した新生仔マウスから胸腺を摘出し、T細胞でのSatb1-Venusの蛍光強度をフローサイトメトリーにより解析した。
(Methods and Results)
5-ph-IAA was intraperitoneally administered at a dose of 0.1 mg/body to pregnant mice (17.5 days of pregnancy) or lactating female mice on the second day after birth for the Rosa26 OsTIR1/+ :Satb1 Venus-mAID/+ :Cd4Cre mice prepared in Example 2. 24 hours after administration, the thymus was removed from fetal mice or newborn mice lactated by female mice, and the fluorescence intensity of Satb1-Venus in T cells was analyzed by flow cytometry.

 結果を図5に示す。Satb1-Venusの蛍光強度をフローサイトメトリーにより計測した結果、胎仔及び新生仔マウスの両方でSatb1-Venusの発現が消失していることが明らかになった。この結果から、母親マウスへの5-ph-IAA投与により、胎仔及び新生仔マウスにおいてタンパク質分解を誘導できることが実証された。 The results are shown in Figure 5. Measurement of the fluorescence intensity of Satb1-Venus by flow cytometry revealed that expression of Satb1-Venus was lost in both fetal and neonatal mice. This result demonstrated that administration of 5-ph-IAA to maternal mice can induce protein degradation in fetal and neonatal mice.

<実施例4:膜タンパク質を標的とする分解>
(目的)
 膜タンパク質を標的としたタンパク質分解の効果を培養細胞及びマウス生体内で検証する。
Example 4: Targeted degradation of membrane proteins
(the purpose)
The effect of targeting membrane proteins for protein degradation will be verified in cultured cells and in vivo in mice.

(方法と結果)
(1)培養細胞でのPD-1タンパク質の分解
 PD-1タンパク質のC末端側にmAIDデグロン配列を融合させた融合タンパク質及びOsTIR1(F74G)タンパク質を発現させたT細胞株(Jurkat T細胞)を作製し、1μMの5-ph-IAA又はDMSOを培養液中に添加した。添加から0.5時間後、1時間後、及び4時間後、T細胞株におけるPD-1タンパク質の発現レベルをフローサイトメトリーにより解析した。
(Methods and Results)
(1) Degradation of PD-1 protein in cultured cells We generated T cell lines (Jurkat T cells) expressing a fusion protein in which the mAID degron sequence was fused to the C-terminus of PD-1 protein and OsTIR1(F74G) protein, and added 1 μM 5-ph-IAA or DMSO to the culture medium. 0.5, 1, and 4 hours after addition, the expression levels of PD-1 protein in the T cell lines were analyzed by flow cytometry.

 結果を図6に示す。5-ph-IAAを添加したT細胞株は、DMSOを添加した対照細胞と比較して、PD-1タンパク質の発現が減少した。 The results are shown in Figure 6. T cell lines treated with 5-ph-IAA showed reduced expression of PD-1 protein compared to control cells treated with DMSO.

(2)生体内でのPD-1タンパク質の分解
 ゲノム編集法を用いてmini AIDデグロン配列(mAID)をコードする塩基配列をPD-1遺伝子に導入することによって、PD-1タンパク質のC末端側にmini AIDデグロン配列(mAID)を融合させた融合タンパク質を発現するPdcd1-mAIDマウスを作製した(図7A)。
(2) Degradation of PD-1 protein in vivo By using genome editing to introduce a base sequence encoding the mini AID degron sequence (mAID) into the PD-1 gene, we generated Pdcd1-mAID mice expressing a fusion protein in which the mini AID degron sequence (mAID) is fused to the C-terminus of the PD-1 protein (Figure 7A).

 次に、実施例1で作製したRosa26OsTIR1(F74G)マウス、Pdcd1-mAIDマウス、及び血球系細胞特異的にCreリコンビナーゼを発現するVav1Cre Tgマウス(4~8週齢の雄又は雌;ジャクソン研究所より購入)を交配させることによって、Rosa26OsTIR1/+:Pdcd1-mAID:Vav1Creマウスを作製した(図7A)。 Next, Rosa26 OsTIR1(F74G) mice prepared in Example 1, Pdcd1-mAID mice, and Vav1Cre Tg mice (4-8 week-old male or female; purchased from Jackson Laboratory) that express Cre recombinase specifically in blood cells were crossed to generate Rosa26 OsTIR1/+ :Pdcd1-mAID:Vav1Cre mice (Figure 7A).

 Rosa26OsTIR1/+:Pdcd1-mAID: Vav1Creマウスに対して、0.1 mg/bodyの用量で5-ph-IAA又はPBSを腹腔内投与した。投与から12時間後にパイエル板から濾胞性T細胞を調製し、PD-1タンパク質の発現をフローサイトメトリーで計測した。 Rosa26 OsTIR1/+ :Pdcd1-mAID: Vav1Cre mice were intraperitoneally administered 5-ph-IAA or PBS at a dose of 0.1 mg/body. Twelve hours after administration, follicular T cells were prepared from Peyer's patches, and PD-1 protein expression was measured by flow cytometry.

 結果を図7Bに示す。PBSを投与した対照マウスと比較して、5-ph-IAAを投与したマウスでは、PD-1タンパク質の発現が消失した。 The results are shown in Figure 7B. Compared to control mice treated with PBS, PD-1 protein expression was lost in mice treated with 5-ph-IAA.

<実施例5:PD1タンパク質を標的とする分解に基づく抗腫瘍効果の検証>
(目的)
 実施例4で作出したマウスに大腸癌を移殖し、5-ph-IAAの投与によってPD1の分解を誘導することによって、抗腫瘍免疫効果が得られることを検証する。また、PD1の分解誘導に基づく抗腫瘍免疫応答の増強に重要な血液細胞の種類を特定する。
Example 5: Verification of antitumor effect based on targeted degradation of PD1 protein
(the purpose)
We will transplant colon cancer into the mice created in Example 4, and verify that antitumor immune effects can be obtained by inducing PD1 degradation through administration of 5-ph-IAA. We will also identify the types of blood cells that are important in enhancing antitumor immune responses based on the induction of PD1 degradation.

(方法と結果)
 実施例4で作出したPD1デグロンマウス(Rosa-OsTIR1/+; Pdcd1mAID/mAID;Vavcre)にMC38大腸癌を移植した。移植の10日後からPBS又は5-ph-IAAを2日に1回の頻度で連続投与し(図8A)、PBS投与群と5-ph-IAA投与群における腫瘍径を比較した。
(Methods and Results)
MC38 colon cancer was transplanted into the PD1 degron mice (Rosa-OsTIR1/+; Pdcd1 mAID/mAID ; Vavcre) generated in Example 4. From 10 days after transplantation, PBS or 5-ph-IAA was administered continuously once every two days ( FIG. 8A ), and the tumor diameters in the PBS-administered group and the 5-ph-IAA-administered group were compared.

 腫瘍径の測定結果を図8Cに示す。5-ph-IAA投与群では腫瘍径の顕著な縮小が観察された。これは、過去の文献で示されている抗PD1抗体投与の効果と同様の効果である。また、腫瘍浸潤細胞(TIL)におけるPD1の発現レベルを測定したところ、PD1の発現が低下していることも確認された(図8B)。 The results of tumor size measurements are shown in Figure 8C. A significant reduction in tumor size was observed in the 5-ph-IAA administration group. This is similar to the effect of anti-PD1 antibody administration shown in previous literature. In addition, when the expression level of PD1 in tumor-infiltrating cells (TIL) was measured, it was confirmed that PD1 expression was reduced (Figure 8B).

 次に、PD1の分解誘導に基づく抗腫瘍免疫応答の増強に重要な細胞種を特定するために、異なるCreトランスジェニックマウスを使用してOsTIR1を発現する細胞種を検討した。具体的には、全血球型細胞でOsTIR1を発現するRosa-OsTIR1/+;Pdcd1mAID/mAID;Vavcreマウス、及びCD8T細胞のみでOsTIR1を発現するRosa-OsTIR1/+;Pdcd1mAID/mAID;E8Icreマウスに上記と同様の方法でMC38大腸癌を移殖し、PBS又は5-ph-IAAを連続投与して、腫瘍径を測定した。 Next, to identify the cell types important for enhancing antitumor immune responses based on PD1 degradation induction, we examined the cell types expressing OsTIR1 using different Cre transgenic mice. Specifically, MC38 colon cancer was transplanted into Rosa-OsTIR1/+;Pdcd1 mAID/mAID ;Vavcre mice expressing OsTIR1 in all blood cells, and Rosa-OsTIR1/+;Pdcd1 mAID/mAID ;E8Icre mice expressing OsTIR1 only in CD8 T cells, using the same method as above. PBS or 5-ph-IAA was administered continuously, and the tumor diameter was measured.

 結果を図9に示す。CD8T細胞のみでOsTIR1を発現するRosa-OsTIR1/+;Pdcd1mAID/mAID;E8Icreマウスにおいても、5-ph-IAAの投与によって腫瘍径の顕著な縮小が観察された(図9A)。この効果は、全血球型細胞でOsTIR1を発現するRosa-OsTIR1/+;Pdcd1mAID/mAID;Vavcreマウスにおいて観察された効果と同等だった(図9B)。以上の結果から、CD8T細胞のみでのPD1分解が抗腫瘍免疫応答増強には十分であることが示された。この結果は、Creトランスジェニックマウスを選択することによってOsTIR1を発現する細胞種を特定できるという本発明のPD1デグロンマウスの利点を示している。 The results are shown in Figure 9. In Rosa-OsTIR1/+;Pdcd1 mAID/mAID ;E8Icre mice expressing OsTIR1 only in CD8 T cells, a significant reduction in tumor size was observed by administration of 5-ph-IAA (Figure 9A). This effect was equivalent to that observed in Rosa-OsTIR1/+;Pdcd1 mAID/mAID ;Vavcre mice expressing OsTIR1 in all blood cells (Figure 9B). These results indicate that PD1 degradation only in CD8 T cells is sufficient to enhance antitumor immune responses. This result indicates the advantage of the PD1 degron mouse of the present invention, that the cell type expressing OsTIR1 can be identified by selecting Cre transgenic mice.

 以上の結果から、膜タンパク質を標的とした分解を誘導できることが示された。
 本明細書で引用した全ての刊行物、特許及び特許出願はそのまま引用により本明細書に組み入れられるものとする。
These results demonstrate that it is possible to induce the targeted degradation of membrane proteins.
All publications, patents, and patent applications cited herein are hereby incorporated by reference in their entirety.

Claims (18)

 遺伝子改変マウスであって、
 一対のCreリコンビナーゼ認識配列、
 前記一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列、
 前記一対のCreリコンビナーゼ認識配列の下流に位置する、Transport Inhibitor Response 1(TIR1)ファミリータンパク質をコードするTIR1ファミリー遺伝子配列、及び
 前記一対のCreリコンビナーゼ認識配列の上流に位置し、前記一対のCreリコンビナーゼ認識配列間の組換え後に前記TIR1ファミリー遺伝子の発現を駆動することができるプロモーター配列
を含む、前記遺伝子改変マウス。
A genetically modified mouse, comprising:
A pair of Cre recombinase recognition sequences,
A transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences;
The genetically modified mouse comprises: a Transport Inhibitor Response 1 (TIR1) family gene sequence encoding a TIR1 family protein, which is located downstream of the pair of Cre recombinase recognition sequences; and a promoter sequence, which is located upstream of the pair of Cre recombinase recognition sequences and is capable of driving expression of the TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences.
 前記TIR1ファミリータンパク質において、配列番号1で示すアミノ酸配列の74位に対応するPhe残基が、Gly残基、Ala残基、又はSer残基に置換されている、請求項1に記載の遺伝子改変マウス。 The genetically modified mouse according to claim 1, wherein the Phe residue corresponding to position 74 of the amino acid sequence shown in SEQ ID NO:1 in the TIR1 family protein is replaced with a Gly residue, an Ala residue, or a Ser residue.  前記一対のCreリコンビナーゼ認識配列、前記転写終結polyA配列、前記TIR1ファミリー遺伝子配列、及び前記プロモーター配列をRosa26遺伝子座に含む、請求項1又は2に記載の遺伝子改変マウス。 The genetically modified mouse according to claim 1 or 2, comprising the pair of Cre recombinase recognition sequences, the transcription termination polyA sequence, the TIR1 family gene sequence, and the promoter sequence in the Rosa26 locus.  デグロン配列が付加した標的タンパク質をコードする遺伝子、及び/又はCreリコンビナーゼをコードする遺伝子をさらに含む、請求項1に記載の遺伝子改変マウス。 The genetically modified mouse of claim 1, further comprising a gene encoding a target protein to which a degron sequence has been added and/or a gene encoding a Cre recombinase.  前記標的タンパク質が膜タンパク質である、請求項4に記載の遺伝子改変マウス。 The genetically modified mouse of claim 4, wherein the target protein is a membrane protein.  前記Creリコンビナーゼをコードする遺伝子が、部位特異的プロモーターの制御下に配置されている、請求項4又は5に記載の遺伝子改変マウス。 The genetically modified mouse according to claim 4 or 5, wherein the gene encoding the Cre recombinase is placed under the control of a site-specific promoter.  前記部位特異的プロモーターが発現を駆動する部位における、前記標的タンパク質をコードする遺伝子の機能が、発生及び/又は生存に必須である、請求項6に記載の遺伝子改変マウス。 The genetically modified mouse according to claim 6, wherein the function of the gene encoding the target protein at the site where the site-specific promoter drives expression is essential for development and/or survival.  請求項1又は2に記載の遺伝子改変マウスに発生する、精子、受精卵、又は胚。  A sperm, a fertilized egg, or an embryo generated in the genetically modified mouse according to claim 1 or 2.  請求項1又は2に記載の遺伝子改変マウスに由来する細胞。 Cells derived from the genetically modified mouse according to claim 1 or 2.  標的タンパク質の分解をマウス生体内で誘導する方法であって、
 オーキシン又はオーキシンアナログを遺伝子改変マウスに投与して、デグロン配列が付加した標的タンパク質の分解を誘導する、分解誘導工程
を含み、
 前記遺伝子改変マウスは、TIR1ファミリー遺伝子発現システム、前記デグロン配列が付加した標的タンパク質をコードする遺伝子、及びCreリコンビナーゼをコードする遺伝子を含み、
 前記TIR1ファミリー遺伝子発現システムは、
  一対のCreリコンビナーゼ認識配列、
  前記一対のCreリコンビナーゼ認識配列間に位置する転写終結polyA配列、
  前記一対のCreリコンビナーゼ認識配列の下流に位置する、Transport Inhibitor Response 1(TIR1)ファミリータンパク質をコードするTIR1ファミリー遺伝子配列、及び
  前記一対のCreリコンビナーゼ認識配列の上流に位置し、前記一対のCreリコンビナーゼ認識配列間の組換え後に前記TIR1ファミリー遺伝子の発現を駆動することができるプロモーター配列
を含む、前記方法。
A method for inducing degradation of a target protein in a mouse body, comprising:
A degradation induction step of administering an auxin or an auxin analog to a genetically modified mouse to induce degradation of a target protein to which a degron sequence has been added,
The genetically modified mouse comprises a TIR1 family gene expression system, a gene encoding a target protein to which the degron sequence has been added, and a gene encoding a Cre recombinase;
The TIR1 family gene expression system comprises:
A pair of Cre recombinase recognition sequences,
A transcription termination polyA sequence located between the pair of Cre recombinase recognition sequences;
the method comprising: a Transport Inhibitor Response 1 (TIR1) family gene sequence encoding a TIR1 family protein, located downstream of the pair of Cre recombinase recognition sequences; and a promoter sequence located upstream of the pair of Cre recombinase recognition sequences, capable of driving expression of the TIR1 family gene after recombination between the pair of Cre recombinase recognition sequences.
 前記オーキシン又はオーキシンアナログの投与量が、0.1 mg/kg体重~50 mg/kg体重である、請求項10に記載の方法。 The method of claim 10, wherein the dosage of the auxin or auxin analog is between 0.1 mg/kg body weight and 50 mg/kg body weight.  前記分解誘導工程後の前記遺伝子改変マウスの表現型を評価する評価工程をさらに含む、請求項10又は11に記載の方法。 The method according to claim 10 or 11, further comprising an evaluation step of evaluating the phenotype of the genetically modified mouse after the degradation induction step.  前記分解誘導工程の3時間以上後に前記評価工程を行う、請求項12に記載の方法。 The method according to claim 12, wherein the evaluation step is carried out at least 3 hours after the decomposition induction step.  前記遺伝子改変マウスが胎仔又は離乳前の仔マウスであり、前記分解誘導工程においてオーキシン又はオーキシンアナログをその母親マウスに投与する、請求項10又は11に記載の方法。 The method according to claim 10 or 11, wherein the genetically modified mouse is a fetus or a preweaned mouse, and the auxin or auxin analog is administered to the mother mouse in the degradation induction step.  前記遺伝子改変マウスが疾患モデルマウスであり、前記標的タンパク質が創薬標的候補タンパク質である、請求項10に記載の方法。 The method according to claim 10, wherein the genetically modified mouse is a disease model mouse, and the target protein is a candidate protein for drug discovery.  前記創薬標的候補タンパク質をコードする遺伝子の全身ノックアウト又は条件付きノックアウトに基づく破壊は、前記疾患モデルマウスに対する治療又は予防効果を示さないか、又は副作用を示す、請求項15に記載の方法。 The method according to claim 15, wherein the disruption based on a whole-body knockout or conditional knockout of the gene encoding the candidate drug discovery target protein does not show a therapeutic or preventive effect on the disease model mouse, or shows side effects.  前記創薬標的候補タンパク質とは異なる因子を標的とする薬剤を前記遺伝子改変マウスに投与する工程をさらに含み、
 前記評価工程が、前記創薬標的候補タンパク質と前記因子との両者を標的とした場合の表現型と、前記因子のみを標的とした場合の表現型とを比較することを含む、請求項15又は16に記載の方法。
The method further comprises administering to the genetically modified mouse a drug that targets a factor other than the drug discovery target candidate protein;
The method according to claim 15 or 16, wherein the evaluation step comprises comparing a phenotype obtained when both the candidate drug target protein and the factor are targeted with a phenotype obtained when only the factor is targeted.
 標的タンパク質の分解をマウス由来の細胞において誘導する方法であって、
 請求項9に記載の細胞を培養する、培養工程、
 オーキシン又はオーキシンアナログを前記細胞に添加して、デグロン配列が付加した標的タンパク質の分解を誘導する、分解誘導工程
を含む、前記方法。
1. A method for inducing degradation of a target protein in a cell derived from a mouse, comprising:
A culturing step of culturing the cell according to claim 9 ;
The method described above, further comprising a degradation inducing step of adding an auxin or an auxin analog to the cells to induce degradation of the target protein to which the degron sequence has been added.
PCT/JP2024/008753 2023-03-09 2024-03-07 Genetically modified mouse Pending WO2024185847A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2023-036211 2023-03-09
JP2023036211 2023-03-09

Publications (1)

Publication Number Publication Date
WO2024185847A1 true WO2024185847A1 (en) 2024-09-12

Family

ID=92675310

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2024/008753 Pending WO2024185847A1 (en) 2023-03-09 2024-03-07 Genetically modified mouse

Country Status (1)

Country Link
WO (1) WO2024185847A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015507932A (en) * 2012-02-15 2015-03-16 マイス ウィズ ホーンズ リミテッド ライアビリティ カンパニー Transgenic animals with customizable traits
WO2021009993A1 (en) * 2019-07-16 2021-01-21 大学共同利用機関法人情報・システム研究機構 Nonhuman animal and use for same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015507932A (en) * 2012-02-15 2015-03-16 マイス ウィズ ホーンズ リミテッド ライアビリティ カンパニー Transgenic animals with customizable traits
WO2021009993A1 (en) * 2019-07-16 2021-01-21 大学共同利用機関法人情報・システム研究機構 Nonhuman animal and use for same

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KANEMAKI MASATO: "Protein Degradation Control by Use of a Small Molecule: From Chemical Degraders to Degron Technologies", KAGAKU TO SEIBUTSU, vol. 58, no. 7, 1 July 2020 (2020-07-01), pages 411 - 415, XP093208118, DOI: 10.1271/kagakutoseibutsu.58.411 *
KIM JAE-HONG, KWON OSUNG, BHUSAL ANUP, LEE JIYOUN, HWANG EUN MI, RYU HOON, PARK JAE-YONG, SUK KYOUNGHO: "Neuroinflammation Induced by Transgenic Expression of Lipocalin-2 in Astrocytes", FRONTIERS IN CELLULAR NEUROSCIENCE, FRONTIERS RESEARCH FOUNDATION, CH, vol. 16, 23 February 2022 (2022-02-23), CH , XP093208110, ISSN: 1662-5102, DOI: 10.3389/fncel.2022.839118 *

Similar Documents

Publication Publication Date Title
CN107287204B (en) Construction method and application of PD-1 gene modified humanized animal model
US20200022342A1 (en) Genetically Modified Non-Human Animal With Human Or Chimeric PD-L1
WO2018041118A1 (en) Genetically modified non-human animal with human or chimeric pd-l1
US20200029538A1 (en) Genome editing method
JP2005519631A (en) Targeted chromosomal mutagenesis using zinc finger nuclease
JP2009219499A (en) Gene disruption, composition and method relating thereto
JP2025003538A (en) Genetically modified non-human animals having human or chimeric MHC protein complexes
JP2023104002A (en) exon humanized mouse
CN103635588B (en) Method for screening substances with body weight regulating effect
CN114134183B (en) Construction method and application of SIGLEC15 gene humanized animal model
JP6600694B2 (en) Non-human animals exhibiting upper and lower motor neuron function and sensory decay
WO2024185847A1 (en) Genetically modified mouse
CN117467701B (en) A method for constructing a transgenic lung adenocarcinoma mouse model overexpressing MR-1
JP2019092391A (en) Production method of gene modified animal using germ cell defect animal
KR102124236B1 (en) PARK2 knock-out porcine Models for Parkinson' s disease and the Use thereof
CN115010800B (en) Construction method and application of PVRIG gene humanized non-human animal
KR101055376B1 (en) CIIA transgenic mice resistant to degenerative neurological disease
JP5240756B2 (en) Cartilage disease model non-human animal
JP2002142610A (en) ALS model rat
JPH10509313A (en) Transgenic organisms and methods of using the same
JP2023175832A (en) Methods for producing fusion proteins, nucleic acids, cells and animals
JP6078383B2 (en) Transgenic non-human mammal
WO2019161805A1 (en) Hr knockout non-human animal
KR20160088997A (en) Composition for Preparing an Immuno-Deficient Zebrafish Model and Use Thereof
WO2008062904A1 (en) Method of enabling stable expression of transgene

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: 24767213

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE