WO2007141974A1 - Nonhuman model animal of hypertension - Google Patents

Nonhuman model animal of hypertension Download PDF

Info

Publication number
WO2007141974A1
WO2007141974A1 PCT/JP2007/059316 JP2007059316W WO2007141974A1 WO 2007141974 A1 WO2007141974 A1 WO 2007141974A1 JP 2007059316 W JP2007059316 W JP 2007059316W WO 2007141974 A1 WO2007141974 A1 WO 2007141974A1
Authority
WO
WIPO (PCT)
Prior art keywords
ramp1
gene
disorder
human animal
test substance
Prior art date
Application number
PCT/JP2007/059316
Other languages
French (fr)
Japanese (ja)
Inventor
Kazutake Tsujikawa
Hiroshi Yamamoto
Masaru Okabe
Hiroshi Okamoto
Katsutoshi Yayama
Original Assignee
Osaka University
Pca Intermed, Inc.
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 Osaka University, Pca Intermed, Inc. filed Critical Osaka University
Publication of WO2007141974A1 publication Critical patent/WO2007141974A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5082Supracellular entities, e.g. tissue, organisms
    • G01N33/5088Supracellular entities, e.g. tissue, organisms of vertebrates
    • 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
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • 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
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • 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
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • 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
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0375Animal model for cardiovascular diseases
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/30Vector systems comprising sequences for excision in presence of a recombinase, e.g. loxP or FRT

Definitions

  • a hypertensive model mouse useful as a model animal for hypertension-related diseases in which a specific gene is deleted and the function of the gene is completely lost, and prevention of hypertension-related diseases using the hypertension model mouse The present invention relates to a method for evaluating and Z or searching for a drug suitable for treatment.
  • RAMP1 belongs to the receptor activity-modifying protein (RAMP) family, and has a signal sequence at the N-terminus and a single transmembrane domain at the C-terminus.
  • RAMP receptor activity-modifying protein
  • RAMP1 forms a heterodimer by co-expression with calcitonin receptor-like receptor (CRLR) and functions as a receptor for calcitonin gene-related protein (CGR P), a neuropeptide that exhibits potent vasorelaxation. Is known.
  • JP 2005-503809 discloses a mutant mouse capable of expressing the human ⁇ CGRP receptor by a complex of endogenous CRLR and human ⁇ RAMP1, which is knocked-in by RAMP1 force S that has been humanized by amino acid mutation. Is disclosed.
  • This human ⁇ CGRP receptor has a function equivalent to that of the CGRP receptor antagonist shown in the wild type human CGRP receptor, and it is described that it can be used for screening assays for receptor-binding substances.
  • the animals lacking RA MP 1 are shown! /.
  • Patent 3639555 discloses a genetically modified mouse homozygous for the RAMP1 gene disruption.
  • a fragmented RAMP1 gene is a cell activity of the RAMP1 polypeptide encoded by the fragmented gene in cells that normally express the wild type of the RAMP1 gene. It means a RAMP gene that has been genetically modified to decrease.
  • the non-human mammal (mouse) genetically modified as described above expresses RAMP1 polypeptide, and the polypeptide activity is 50% or less of the wild type level.
  • AM adrenomedullin
  • Patent Document 1 Japanese Translation of Special Publication 2005-503809
  • Patent Document 2 Patent No. 3639555
  • Non-Patent Document 1 Science. 1997 Feb 14; 275 (5302): 964-967
  • Non-Patent Document 2 Nat Med. 1999 Apr; 5 (4): 434-8
  • Non-Patent Document 3 J. Exp. Med. 2002 Jan 21; 195: 151-160
  • An object of the present invention is to prevent a vascular disorder disease using a pathological condition (hypertension due to vasoconstrictive disorder) model non-human animal useful for the treatment of a vascular disorder disease, and the animal.
  • a pathological condition hypertension due to vasoconstrictive disorder
  • An object of the present invention is to provide a method for efficiently screening for Z or a therapeutic drug, and a drug obtained by the method.
  • Another object of the present invention is to provide a method for easily screening a functional food using a non-human animal model of hypertension, a method for evaluating the functional food, and a functional food obtained by the method.
  • the present inventors have found that the function of the RAMP1 gene is completely deficient due to partial or complete modification of the RAMP1 gene on the chromosome.
  • the present invention can be used as a high blood pressure model animal, and such a high blood pressure model animal has been found to be extremely useful for screening for drugs for the prevention and treatment of vascular disorder diseases and for evaluation of functional foods. completed.
  • the present invention provides a hypertensive model non-human animal characterized in that the function of the RAMP1 gene is completely deficient by modification of part or all of the RAMP1 gene on the chromosome.
  • the non-human animal of the present invention preferably has no selection marker gene inserted on the chromosome.
  • the hypertensive model non-human animal of the present invention has, for example, a region in which at least a part of the RAMP1 gene exon is sandwiched between LoxP sequences and a selectable marker gene is sandwiched between LoxP sequences on a chromosome.
  • ES cells obtained by allowing Cre enzyme to act on ES cells containing a region, or using ES cells containing a region where at least a part of the RAMP1 gene exon is sandwiched between LoxP sequences on the chromosome
  • the RAMPl-floxed mouse prepared in this way and the method of mating with a mouse capable of expressing the Cre enzyme can be produced by a deviation.
  • the expression of the RAMP1 gene may be conditionally suppressed.
  • the non-human animal is, for example, a rat or a mouse.
  • the present invention provides a blood vessel characterized by administering a test substance to the above-mentioned hypertension model non-human animal of the present invention or bringing a tissue, organ or cell derived from the animal into contact with the test substance.
  • a method for screening for drugs for the prevention and Z or treatment of disability diseases can be performed using Z or vasodilatory action as an index.
  • the vascular disorder diseases in the present invention include, for example, hypertension, vascular endothelial disorder, arteriosclerotic disease, and blood flow disorder diseases including memory disorder, dementia, peripheral blood flow disorder disease.
  • the present invention also provides a preventive and Z or therapeutic agent for a vascular disorder disease obtained by the drug screening method of the present invention.
  • the present invention provides a blood flow improving drug containing, as an active ingredient, a substance that enhances the expression or function of the RAMP1 gene.
  • the present invention also provides a prophylactic and Z or therapeutic agent for vascular disorder diseases containing a substance that enhances the expression or function of the RAMP1 gene as an active ingredient.
  • a test substance is administered to the above-described hypertension model non-human animal of the present invention, or There is provided a screening method for functional food, which comprises contacting a tissue, organ or cell derived from the animal with a test substance.
  • the method can be performed using blood pressure and Z or vasodilating action as indices.
  • the present invention provides a functional food obtained by the functional food screening method of the present invention.
  • the present invention is a function characterized in that the test substance is administered to the non-human hypertensive model non-human animal of the present invention, or a tissue, organ or cell derived from the animal is brought into contact with the test substance.
  • a method for evaluating sex foods is provided. The method can be performed using blood pressure and Z or vasodilating action as indices.
  • a model non-human animal that constantly shows hypertension due to vasodilatation failure is obtained by completely deleting the function of the RAMP1 gene by modifying the RAMP1 gene on the chromosome. Can do.
  • a drug useful for the prevention and treatment of vascular disorder disease can be efficiently screened, and the efficacy of the test substance can be easily evaluated.
  • Such non-hypertensive animals with hypertension can also be used for screening (selection) and evaluation of functional foods that exhibit a blood pressure lowering and blood vessel relaxation action to prevent or improve hypertension and blood flow disorders.
  • FIG. 1 is a schematic sectional view of a targeting vector used in Example 1.
  • FIG. 2 is a photograph of agarose gel electrophoresis showing the expression pattern of each organ of 8-week-old mice of RAMP1 + / +, RAMP1 +/ ⁇ and RAMP1 ⁇ / ⁇ obtained in Example 1.
  • FIG. 3 (i) is a graph showing blood pressure at 10 weeks of age for each of the RAMP1 + / +, RAMP1 +/-, and RAMP1-/-mice obtained in Example 1, and (mouth) is the heart rate. It is a graph to show.
  • FIG. 4 (i) is a graph showing blood pressure at 6-18 weeks of age for each of the RAMP1 + / +, RAMP1 +/-, and RAMP1-/-mice obtained in Example 1, and (mouth) is the same heart rate. It is a graph which shows a number.
  • FIG. 5 shows the vasoconstriction rate under the influence of three vasodilators on 12-week-old mice of RAMP1 + / +, RAMP1 +/-, and RAMP1-/-obtained in Example 1. The graph which shows is shown.
  • BEST MODE FOR CARRYING OUT THE INVENTION [0020]
  • RAMP1 belongs to the receptor activity-modifying protein (RAMP) family and has a signal sequence at the N-terminus and a single transmembrane domain at the C-terminus.
  • the nucleotide sequence of RAMP1 gene and the amino acid sequence of RAMP1 are known.
  • the nucleotide sequence of cDNA of human and mouse RAMP1 includes the sequences represented by GenBank Accession No. AJ001014 and No. BAA76617.
  • FIG. 3 shows wild-type (RAMP1 + / +), RAMP1 heterozygous (RAMP1 +/-), and RAMP1 homozygous (RAMP1-/-) mice measured in 6-week-old mice. It is a graph of blood pressure and (mouth) heart rate. As shown in Fig. 3 (ii), the heart rate is comparable, while blood pressure is compared to RAMP1 + / + mice and RAMP1 +/- mice as shown in Fig. 3 (mouth).
  • FIG. 4 is a graph of (i) blood pressure and (mouth) heart rate measured for each of the RAMPl + / +, RAMPl +/-, and RAMP1-/-mice every 3 weeks from 6 to 18 weeks of age. is there.
  • the heart rate is about the same up to 6-week-old power up to 18-week-old
  • blood pressure is about RAMP1-/-mice as shown in Fig. 4 (mouth).
  • RAMP1-deficient mice maintained the state of hypertension due to vasoconstriction from an early age. Therefore, it can be used as a model for hypertension and vasodilatation disorder.
  • the RAMP1 gene is known to have homologues such as human, rat, pig, and macaque monkeys, and these nucleotide sequences are disclosed in GenBank. Therefore, non-human animals other than mice can also be used as models for hypertension and vasodilatation disorders.
  • the hypertensive model non-human animal of the present invention (hereinafter sometimes referred to as "model animal") is completely deficient in the function of the RAMP1 gene due to partial or complete modification of the RAMP1 gene on the chromosome. It is characterized by that. “The gene function is completely deficient” means, for example, the function that wild-type RAMP1 originally has (for example, it constitutes the CGRP receptor). Function etc.) is completely lost. That is, the model animal of the present invention has no influence (action) due to RAMP1 protein in the individual or can be ignored, and does not include a dominant negative model or dominant active model of RAMP1.
  • a non-human animal in which the function of the RAMP1 gene on the chromosome is completely deleted is prepared according to a method known as a method for producing a knockout non-human animal using ES cells. can do.
  • the model animal of the present invention includes, for example, a configuration in which the entire region of the RAMP1 gene on the chromosome (for example, exons 1 to 3) has been removed; a partial modification of the RAMP1 gene on the chromosome, for example, Examples include a configuration in which the gene function is completely deleted by deletion, addition, or substitution in the base sequence. According to the former configuration, no transcription'translation product is generated in the individual, and it is useful as a model animal in which the influence of the RAMP1 gene is completely eliminated. In addition, the latter structure is not particularly limited as long as the gene function is completely deleted, and an appropriate site and range of the RAMP1 gene can be modified, and mRNA by partial transcription and translation can be used.
  • the model animal of the present invention preferably has a configuration in which at least exon 2 of the RAMP1 gene on the chromosome is removed. Exon 2 has a mature protein coding start site. Therefore, in the model animals from which exon 2 has been removed, the polypeptide fragment of RAMP1 that constitutes the CGRP receptor is not formed, and any influence on the CGRP receptor function associated with the production of the polypeptide fragment is completely eliminated. Therefore, it is suitable for a wide range of uses such as screening (including evaluation) of preventive and therapeutic agents for vascular disorder diseases including blood flow disorder diseases (including evaluation) and research of the mechanism of the disease itself.
  • the method for producing such a hypertensive model non-human animal is not particularly limited as long as it can completely lack the function of the RAMP1 gene on the chromosome, but for example, a method using the CreZLoxP system can be mentioned.
  • the CreZLoxP system is a system in which the Cre enzyme recognizes the LoxP sequence and causes specific recombination.
  • the advantage of this system is that, for example, the region where the DNA region can be deleted is limited to about 20 kb for homologous recombination In contrast to gene targeting, which is about 200 kb This is an easy method, and point mutations can be easily introduced, and conditional gene targeting is possible by the expression method of Cre enzyme.
  • a method of using the CreZLoxP system in the present invention for example, a method in which Cre enzyme is transiently expressed in ES cells and used in a step of removing a drug selection marker can be used.
  • ES cell RAMPl-floxed-marker ES cell
  • Cre enzyme is transiently expressed in ES cells and used in a step of removing a drug selection marker
  • ES cell RAMPl-floxed-marker ES cell
  • the former LoxP sequence sandwiches a sequence containing at least the mature protein coding start site of the RAMP1 gene on the chromosome (eg, eX0 n2)
  • the RAMP1 protein is completely defective due to the action of the Cre enzyme. Can be easily configured.
  • FIG. 1 is a schematic diagram showing an example of a method for producing ES cells obtained by allowing a Cre enzyme to act on the RAMPl-floxed-marker ES cells.
  • ES cells used for the production of a hypertensive model non-human animal of the present invention are (ii) against the RAMP1 gene consisting of exonl to 3 on the chromosome of the ES cell (ii) From the first selectable marker gene (thymidine kinase: TK) sandwiched between LoxP sequences, exon2 of the RAMP1 gene sandwiched between LoxP sequences, and the second selectable marker gene (neomycin: neo) sandwiched between LoxP sequences Using a targeting vector comprising the following structure, homologous recombination is caused, and TK activity is used as an index.
  • TK thymidine kinase
  • ES cells obtained in this way are completely deficient in the function of the RAMP1 gene on the chromosome and have no insertion of a selectable marker gene.
  • an ES cell-derived individual having the above-described configuration can be obtained according to a known method for producing a transgenic animal.
  • An example is a method of mating floxed mice with mice capable of expressing the Cre enzyme.
  • the RAMPl-floxed mouse is not particularly limited as long as it contains at least part of the RAMP1 gene exon sandwiched between LoxP sequences on the chromosome. For example, two RAMP1 gene exons are included. By homologous recombination in a form sandwiched between ⁇ sequences, the gene itself is damaged, and mice can be used.
  • the mouse capable of expressing the Cre enzyme can be prepared by a known method such as a method of introducing a Cre enzyme expression vector.
  • the RAMP1 gene can be obtained by using a mouse capable of expressing the Cre enzyme under the control of a yarn and tissue-specific or expression-inducible promoter (Cre enzyme conditional expression mouse). The conditional gene targeting can be easily performed.
  • the RAMP1 gene deficiency on the chromosome may be either constitutive or conditional [tissue-specific, time-specific (transient), etc.].
  • a hypertension model can be constructed under a desired condition.
  • a mouse that expresses the above Cre enzyme in a conditional manner. It can construct
  • the method for screening a prophylactic and Z or therapeutic agent for a vascular disorder disease of the present invention comprises administering a test substance to the above-mentioned hypertension model non-human animal of the present invention, or a tissue, organ derived from the animal, Or it is characterized by making a cell contact a test substance.
  • the former may be referred to as “screening method 1” and the latter as “screening method 2”.
  • test substance to be used in the screening method (1, 2) of the present invention may be any known or novel compound.
  • nucleic acids, carbohydrates, lipids, proteins, peptides, small organic molecules Synthetic or natural organic compounds such as compounds can be used.
  • the method of administering the test substance to the model animal is not particularly limited, and may be either oral or parenteral administration.
  • parenteral administration for example, systemic administration via the vein, artery, muscle, abdominal cavity, airway, etc., or local administration at or near the ischemic site can be used, but is not limited thereto.
  • parenteral administration more Preferably, it is locally administered to or near the blood vessel.
  • the dose of the test substance can be appropriately set according to the type of test substance, the administration method, the type, size, etc. of the model animal.
  • the evaluation in screening method 1 can be performed using, for example, the blood pressure value of the model animal as an index. Specifically, for example, the contraction after administering a specific amount of the test substance to the non-human animal model of hypertension of the present invention 3 times a day for 2 weeks (administration conditions such as frequency and days).
  • the blood pressure value for the period shows about 130% or less (preferably about 120% or less) of the average blood pressure value of the corresponding wild-type animal
  • the test substance is used for vascular disorder disease prevention and Z or treatment. It can be evaluated that it is useful as a medicine.
  • the model animal is a mouse
  • the hypertensive model mouse of the present invention showing an average blood pressure of about 140 mmHg as compared with a wild-type mouse having an average blood pressure of about lOOmmHg.
  • the blood pressure is 130 mmHg or less (preferably 120 mmHg)
  • it can be evaluated that the test substance is useful as a drug for the prevention and Z or treatment of vascular disorder diseases.
  • the method of bringing a test substance into contact with a tissue, organ, or cell derived from a model animal is not particularly limited, and the test substance is applied to a sample of tissue or organ collected from model animal power.
  • a known method such as a method of adding or culturing cells collected from a model animal in the presence of a test substance can be used.
  • As a method of introducing the test substance into the tissue or organ specimen the same method as the parenteral administration described above can be used.
  • the evaluation in the screening method 2 can be performed using, for example, a vasoconstriction rate as an index.
  • a vasoconstrictor is administered to a blood vessel sample collected from a non-human animal model of hypertension of the present invention, and then a CGRP and a test substance are cumulatively added, and the ratio is compared by ED.
  • test substance When a significant difference is observed when comparing 50 or by the area under the concentration curve (AUC), the test substance is considered to be useful as a drug for the prevention and Z or treatment of vascular disorders. Can be evaluated.
  • Hue - Refurin (10- 6 M) after the vessel was contracted by ⁇ Ka ⁇
  • a CGRP and test substance each final concentration lx 10- u ⁇ 3Xl0- 6 measures the tension variation when the cumulative added to a M, vasoconstriction refraction obtained by conversion is a ratio with ED optionally by cumulative added mosquito ⁇ analyte
  • the change in tension of the blood vessel specimen can be measured using a recorder (model 8K21; NEC) isometrically via, for example, an FD pickup and a carrier amplifier.
  • the vascular disease in the present invention includes hypertension and diseases (complications) caused by hypertension, such as vascular endothelial disorder, arteriosclerotic disease, and blood flow disorder.
  • hypertension and diseases (complications) caused by hypertension such as vascular endothelial disorder, arteriosclerotic disease, and blood flow disorder.
  • atherosclerotic diseases include cerebral infarction, cerebral hemorrhage, subarachnoid hemorrhage, or cerebrovascular disorder diseases; coronary artery diseases such as myocardial infarction and angina; aortic diseases such as aortic aneurysm and aortic dissection; It includes renal arterial diseases such as sclerosis and resulting renal failure; and peripheral arterial diseases such as obstructive arteriosclerosis.
  • blood flow disorders include memory disorders, dementia, and peripheral blood flow disorders.
  • the preventive and Z or therapeutic agent (hereinafter sometimes referred to as "drug") for vascular disorder disease of the present invention contains a test substance obtained by the screening method as an active ingredient.
  • drugs are administered orally or parenterally, for example, dissolved or suspended in a suitable vehicle.
  • parenteral administration for example, systemic administration via the vein, artery, muscle, abdominal cavity, airway, etc., or local administration to or near blood vessels can be used, but is not limited thereto.
  • oral administration is preferably performed, and can also be administered parenterally to the blood vessel or the vicinity thereof.
  • the drug can, for example, cause a substance that enhances the expression or function of the RAMP1 gene to stably reach the contracted blood vessel site or a cell in the vicinity thereof, permeate the cell membrane, and promote the release of the drug with lysosomal Z endosome force. It is also preferable that the delivery system is designed.
  • the drug dosage form may be either liquid or solid.
  • an effective amount of an inhibitor is dissolved, dispersed, or emulsified in a diluent or dispersion medium such as water or physiological saline.
  • a diluent or dispersion medium such as water or physiological saline.
  • These drugs can be added with known pharmaceutically acceptable additives.
  • the drug may be an encapsulated body in which an active ingredient is encapsulated in a ribosome, a sustained-release material, or the like, or a carrier supported on a carrier.
  • an active ingredient is encapsulated in a ribosome, a sustained-release material, or the like, or a carrier supported on a carrier.
  • the dose of the drug varies depending on the type of active ingredient, administration method, symptom, type of administration target, size, drug characteristics, etc., but is usually preferably about 0.5 to 50 mg per day for an adult. It is about 1 to 20 mg, and can be administered once a day or divided into multiple doses.
  • the blood flow improving agent of the present invention contains, as an active ingredient, a substance that enhances the expression or function of the RAMP1 gene by alleviating vasoconstriction.
  • expression means that protein is produced
  • substance that enhances expression means gene transcription, post-transcriptional regulation, translation into protein, and post-translational modification (tyrosine phosphorylation). It may act at any stage such as oxidation) or protein folding.
  • Substances that enhance function include, for example, RAMP1 protein; it acts on functional sites such as protein-protein interaction sites with CRLR in RAMP1 and receptor active sites such as CGRP receptors to increase signal transduction.
  • RAMP1 a substance that improves
  • “dominant active mutant” refers to a substance whose physiological activity has been improved by introducing a mutation into a protein. These mutants can indirectly enhance their function by a synergistic effect with the wild type.
  • Substances that enhance the expression of RAMP1 include, for example, transcription promoting factors, protein synthesis promoters, protein stabilizing enzymes, splicing factors that can promote mRNA cytoplasmic transition, mRNA stabilizing enzymes, and mRNA binding.
  • transcription promoting factors for example, transcription promoting factors, protein synthesis promoters, protein stabilizing enzymes, splicing factors that can promote mRNA cytoplasmic transition, mRNA stabilizing enzymes, and mRNA binding.
  • a substance capable of acting specifically on the target molecule is preferable in order to minimize side effects on other genes and proteins.
  • These functional nucleic acids and proteins may be produced in the administration subject after administration, and can be prepared by known methods using expression vectors, cells, etc. as necessary.
  • the dosage form, administration method, dosage and the like of these blood flow improving drugs are the same as those for the above drugs.
  • the preventive and Z or therapeutic agent for a vascular disorder disease of the present invention may also generate a RAMP1 gene.
  • a substance that enhances the current or function may be contained as an active ingredient. That is, the above blood flow improving drug can be used as a prophylactic or therapeutic drug for vascular disorder diseases.
  • the non-human animal model of hypertension of the present invention can also be used for screening and evaluation of functional foods.
  • functional food means the nutritional function (primary function) inherent in food, sensory functions such as taste fragrance (secondary function), and ecological defense and disease prevention. • Recovery ⁇ Food that has been designed and processed so that biological regulation can be clarified scientifically by focusing on the presence of biological regulation functions (third function) such as adjustment of physical rhythm. In other words, foods with tertiary functions are defined as functional foods.
  • test substances used in the screening method and evaluation method for functional foods of the present invention include vitamins, minerals, oligosaccharides, sugar alcohols, amino acids, peptides, proteins, animal extracts, lactic acid bacteria, yeasts, and the like. And microorganisms and extracts thereof, plants such as herbs and medicinal herbs and extracts thereof, glycosides, oils and fats, and the like.
  • the form of the test substance is not particularly limited, and liquids such as soft drinks, carbonated drinks, liquid seasonings such as dressings; solid seasonings such as table sugar; confectionery such as cookies; and instant foods A solid substance etc. are mentioned.
  • these functional foods may be in a pharmaceutical form such as tablets and capsules.
  • the functional food screening method and evaluation method of the present invention include the ability to administer a test substance to the above-described high blood pressure model non-human animal of the present invention, or a tissue, organ, or animal derived from the animal. It is characterized by contacting cells with a test substance. Preferably, a method of administering a test substance to a model animal is preferred.
  • the screening method for functional foods of the present invention can be carried out in accordance with the above-described prevention method for vascular disorder diseases and screening method for Z or therapeutic agent.
  • the evaluation method for functional food (including the evaluation in the screening method) can set a lower judgment criterion than the evaluation in the above-mentioned drug screening method (1, 2). More specifically, in the method for evaluating functional food, for example, in the aforementioned screening method 1 for drugs, the average blood pressure value of wild-type animals is about 135% or less (about 135 mmHg or less in the case of mouse blood pressure). A test substance can be evaluated as useful as a functional food. According to the above method, functional food that can alleviate hypertension and reduce the risk of vascular disorder disease can be easily obtained. Can be screened and evaluated.
  • the functional food evaluation method of the present invention can be used as a scientific basis for evaluating, for example, functional health foods including foods for specified health use and functional foods for nutrition.
  • the above method uses a decrease in blood pressure or a blood vessel relaxing action as an index, for example, the indication of “improvement of eating habits for people with higher blood pressure” in the food for specified health use is displayed on the obtained functional food. It may be possible to appeal the function directly to consumers.
  • the functional food of the present invention can be obtained by the screening method or the evaluation method described above. For this reason, it is extremely useful as a food that can promote the alleviation and improvement of hypertension and reduce or prevent the risk of diseases related to high blood pressure (eg, vascular disorder diseases).
  • diseases related to high blood pressure eg, vascular disorder diseases.
  • RAMP1 + / + means wild type
  • RAMP1 +/- means RAMP1 heterozygote
  • RAMP1-/- means RAMP1 homozygote
  • Example 1 (creation of a RAMP1 knockout mouse)
  • BAC clone GSll- " 2 01 (Kurashiki Spinning Co., Ltd.) found a genome containing exon of RAMP1 in a 9 kbp fragment excised with Ba mHl. By subcloning, we confirmed exon2 of RA MP1.
  • the LoxP sequence was inserted 200 bp upstream, and the LoxP-neomycin metagene (from pPNT) -LoxP cassette was inserted 2 kbp downstream 5. Upon of Exon2 5.
  • the thymid ine kinase gene (from pPNT) was inserted 7 bp into the targeting vector did.
  • Genome is extracted from cells and synthesized oligonucleotides PCR using the primers Primer456 (SEQ ID NO: 1) and Primer298 (SEQ ID NO: 2) as primers and 94 ° C-30 sec + (94 ° C-30 sec 68 ° C- lmin) X 30cycle + 72 ° C-2 min
  • primers Primer456 (SEQ ID NO: 1) and Primer298 (SEQ ID NO: 2) as primers and 94 ° C-30 sec + (94 ° C-30 sec 68 ° C- lmin) X 30cycle + 72 ° C-2 min
  • SEQ ID NO: 1 5,-TGTACCCTGT GCGGTTTGTC TGGGGGCGCG-3, (Primer456)
  • SEQ ID NO: 2 5,-AGGGGAGGAG TAGAAGGTGG CGCGAAGGGG-3 '(Primer298)
  • SEQ ID NO: 3 5,-GTCTTAACTG CACTGTGGGC-3' (Primer494)
  • the resulting RAMPl-floxed ES cells were transfected with 10 g of pCAGGS-Cre linearized with Notl using an erect mouth position using Gene Puller (Bio-Rad) (conditions were 250 ⁇ V and 500 ⁇ ⁇ F). From the next day, G418 (150 ⁇ g / mL) was added as a selective medium. One week after electroporation, colonized ES cells were picked up and passaged for genome extraction and freezing.
  • the clones with Cre were introduced using primers Primer494 (SEQ ID NO: 7) and Primer496 (SEQ ID NO: 8) as primers, and 94 ° C-30sec + (94 ° C-30sec 65 ° C- PCR was performed under the conditions of 30 sec 74 ° C-30 sec) X 40cycle + 72 ° C-2 min. Since the wild-type genome is more than 4kbp away, PCR does not run! / ⁇ , but when Creombi has recombinasion! /, A 500bp band can be confirmed. This confirmed the introduction of Cre recombinase in both clones. Genomic recombinasion was confirmed in all clones.
  • SEQ ID NO: 8 5,-TTGGAACATC ACCACGCACG-3, (Primer496)
  • RAMP1-floxed ES cells were injected into blastocysts of unfertilized eggs collected from C57BL / 6 mice (Japan SLC) and returned to pseudopregnant ICR mice.
  • the color of the litter obtained after 3 weeks was confirmed to be a chimeric mouse.
  • the genome extracted from the tail was subjected to PCR reaction using Primer456 and Primer298 as described above (94 ° C-30 sec using primers of synthetic oligonucleotide Primer456 (SEQ ID NO: 1) and Primer298 (SEQ ID NO: 2)). + (94 ° C-30 sec, 68 ° C-lmin) X PCR at 30 cycles + 72 ° C-2 min) and Southern blotting.
  • the obtained RAMP1 +/ ⁇ mice were mated to obtain RAMP1 ⁇ / ⁇ mice, and the same lump RAMP1 + / + mice and RAMP1 +/ ⁇ mice as controls. Dienotyping was performed on the genome extracted from the tail by a PCR reaction under the same conditions as in (3) above.
  • pCAGGS- Cre transgenic mice By mating pCAGGS- Cre transgenic mice and RAMPl-floxed mice that express Cre recombinase systemically, pCAGGS- Cre positive and RAMP1- floxed mice were obtained as offspring. By mating the mouse with a wild type mouse, a RAMP1 hetero-deficient mouse was born. The heterozygous mice obtained were mated to produce RAMP1-homozygous mice. In dienotyping, the genome extracted from the tail is made into a saddle shape, and synthetic oligonucleotides are used.
  • the RAMP1 ⁇ / ⁇ mice obtained in Example 1 were intravenously injected with a 15 mg / 0.5 ml injection prepared by dissolving the test substance in physiological saline, and the mean blood pressure was measured over time according to the method described later. taking measurement.
  • RAMP1 + / + mice and RAMP1 ⁇ / ⁇ mice at 6 weeks of age are injected with 0.5 ml of physiological saline, and blood pressure is similarly measured over time.
  • the blood pressure of RAMP1 ⁇ / ⁇ mice injected with the test substance is 120 mmHg or less, the test substance is useful as a prophylactic and Z or therapeutic agent for vascular disorder diseases.
  • a 15 mg / 0.5 ml injection prepared by dissolving the test substance in physiological saline is intravenously injected, and the systolic blood pressure is measured over time according to the method described later.
  • RAMP1 + / + and RAMP1-/-mice at 6 weeks of age are injected with 0.5 ml of physiological saline and blood pressure is similarly measured over time.
  • the blood pressure of RAMP1-/-mouse administered with physiological saline is 140 mmHg
  • the blood pressure of RAMP1-/-mice injected with the test substance is 120 mmHg or less
  • the test substance is useful as a preventive and Z or therapeutic agent for vascular disorder diseases.
  • a RAMP1 mutant protein with enhanced RAMP1 function was prepared by converting the 74th amino acid sequence of RAMP1 into tributophan, and dissolved in physiological saline. Then, it is intravenously injected into the RAMP1 ⁇ / ⁇ mouse obtained in Example 1 using it as an injection solution of 15 mg / 0.5 ml, and the systolic blood pressure is measured over time according to the method described later. As a result, the blood pressure of RAMP1 ⁇ / ⁇ mice after injection showed 120 mmHg or less. Therefore, the RAMP1 mutein was used as a blood flow improving agent and as a vascular disorder disease. It is useful as a preventive and z or therapeutic agent for patients.
  • the solid food for mice was crushed, and the RAMP1 mutant protein prepared in Example 6 was mixed at a concentration of 15 mg / g and molded again into a solid form as a test substance, with an average blood pressure of 140 mmHg.
  • the heart rate and systolic blood pressure were measured by the following methods.
  • mice were held under anesthesia, and heart rate and systolic blood pressure were measured by tan- cuff removal using MK2000 (Muromachi Kikai Co., Ltd.). In order to acclimatize to the environment at the time of measurement, each individual was measured 30 times or more, then 10 measurements were performed as the main measurement, and the average value was taken as the value of that individual. The results are shown in Fig. 3. As shown in Figure 3, the mean blood pressure of wild-type mice and heterozygotes is about lOOmmHg, while the mean blood pressure of RAMP1 KO mice obtained above is about 140 mmHg, indicating clear hypertension. It was. In the same manner, changes in heart rate and blood pressure were measured for 6-18 week old mice. The results are shown in Fig. 4. As shown in Figure 4, heart rate did not change in any mouse. The blood pressure of the RAMP1 KO mice tended to gradually increase with the age of 6 weeks, which was already high.
  • the blood vessel was cut to a width of 3 mm to prepare a ring specimen. Insert a tungsten wire with a support rod made of acrylic resin into the blood vessel specimen, 95% 0

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Toxicology (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Environmental Sciences (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Analytical Chemistry (AREA)
  • Vascular Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Plant Pathology (AREA)

Abstract

It is intended to provide a nonhuman model animal of hypertension which is useful in treating a vascular disease, a method of efficiently screening a drug for preventing and/or treating a vascular disease with the use of the above animal, and a drug obtained by the above method. Namely, a nonhuman model animal of hypertension characterized by completely lacking the function of RAMP1 gene on the chromosome due to the partial or entire modification of the RAMP1 gene. This nonhuman model animal of hypertension can be constructed by, for example, either a method with the use of an ES cell obtained by treating an ES cell containing a region wherein at least a part of the exon of RAMP1 gene is sandwiched between LoxP sequences and another region wherein a selection marker gene is sandwiched between LoxP sequences on the chromosome with Cre enzyme, or a method with the use of mating an RAMP1-floxed mouse having been constructed by using an ES cell containing a region wherein at least a part of the exon of RAMP1 gene is sandwiched between LoxP sequences on the chromosome with a mouse capable of expressing a Cre enzyme.

Description

明 細 書  Specification
高血圧モデル非ヒト動物  Hypertension model non-human animal
技術分野  Technical field
[0001] 特定の遺伝子を欠失させて当該遺伝子の機能が完全に欠損されている高血圧関 連疾患のモデル動物として有用な高血圧モデルマウス、及び前記高血圧モデルマウ スを用いて高血圧関連疾患の予防'治療に適した薬剤を評価及び Z又は探索する 方法に関する。  [0001] A hypertensive model mouse useful as a model animal for hypertension-related diseases in which a specific gene is deleted and the function of the gene is completely lost, and prevention of hypertension-related diseases using the hypertension model mouse The present invention relates to a method for evaluating and Z or searching for a drug suitable for treatment.
背景技術  Background art
[0002] RAMP1は、受容体活性変更タンパク質 (RAMP)ファミリーに属し、 N末端にシグナ ル配列を、 C末端に 1回膜貫通ドメインを有するタンパク質である。 RAMP1は、カルシ トニン受容体様受容体 (CRLR)との同時発現によりへテロダイマーを形成し、強力な 血管弛緩作用を示す神経ペプチドであるカルシトニン遺伝子関連タンパク質 (CGR P)の受容体として機能することが知られて 、る。  [0002] RAMP1 belongs to the receptor activity-modifying protein (RAMP) family, and has a signal sequence at the N-terminus and a single transmembrane domain at the C-terminus. RAMP1 forms a heterodimer by co-expression with calcitonin receptor-like receptor (CRLR) and functions as a receptor for calcitonin gene-related protein (CGR P), a neuropeptide that exhibits potent vasorelaxation. Is known.
[0003] 特表 2005— 503809号公報には、アミノ酸変異によりヒト型化した RAMP1力 Sノック インされ、内在 CRLRとヒトイ匕 RAMP1との複合体によるヒトイ匕 CGRP受容体を発現可 能な変異マウスが開示されている。このヒトイ匕 CGRP受容体は CGRP受容体アンタ ゴニストが野生型ヒト CGRP受容体に示すのと同等の機能を有することから、受容体 結合性物質のスクリーニングアツセィなどに利用できることが記載されている力 RA MP 1を欠失させた動物は示されて 、な!/、。  [0003] JP 2005-503809 discloses a mutant mouse capable of expressing the human 匕 CGRP receptor by a complex of endogenous CRLR and human 匕 RAMP1, which is knocked-in by RAMP1 force S that has been humanized by amino acid mutation. Is disclosed. This human 力 CGRP receptor has a function equivalent to that of the CGRP receptor antagonist shown in the wild type human CGRP receptor, and it is described that it can be used for screening assays for receptor-binding substances. The animals lacking RA MP 1 are shown! /.
[0004] 特許第 3639555号には、 RAMP1遺伝子分断に関してホモ接合性の遺伝的に変 更されたマウスが開示されている。特許第 3639555号の記載によれば、分断された RAMP1遺伝子とは、 RAMP1遺伝子の野生型を正常に発現している細胞に於い て分断された遺伝子にコードされた RAMP1ポリペプチドの細胞活性が低下する様 に遺伝的に修飾されている RAMP遺伝子を意味している。また、上記のように遺伝 的に変更された非ヒト哺乳動物(マウス)は、 RAMP1ポリペプチドを発現し、該ポリべ プチド活性は野生型レベルの 50%以下であること、その理由として、 RAMP遺伝子 発現の低下(即ち、 RAMPmRNAレベルの低下により RAMPポリペプチドのレベル が低下する)の結果、及び Z又は分断 RAMP遺伝子が野生型 RAMPポリペプチド に比し低 、機能を持つ変異型ポリペプチドをコードして 、るためであることが記載さ れている。このようなマウスは、個体内で発現される RAMP1遺伝子のポリペプチドに よる、たとえ野生型より低活性であったとしてもその影響を無視することができず、疾 患モデルマウスとしての利用には不向きである。 [0004] Patent 3639555 discloses a genetically modified mouse homozygous for the RAMP1 gene disruption. According to the description of Japanese Patent No. 3639555, a fragmented RAMP1 gene is a cell activity of the RAMP1 polypeptide encoded by the fragmented gene in cells that normally express the wild type of the RAMP1 gene. It means a RAMP gene that has been genetically modified to decrease. In addition, the non-human mammal (mouse) genetically modified as described above expresses RAMP1 polypeptide, and the polypeptide activity is 50% or less of the wild type level. Decreased gene expression (ie, RAMP polypeptide levels due to decreased RAMP mRNA levels) And the Z or fragmented RAMP gene encodes a mutant polypeptide having a function lower than that of the wild-type RAMP polypeptide. Such mice cannot ignore the effects of the polypeptide of the RAMP1 gene expressed in individuals, even if it is less active than the wild type, and can be used as a disease model mouse. It is unsuitable.
[0005] 一方、アドレノメジユリン (AM)は、強力な血管弛緩性ペプチドとして、血圧や循環 動態との関連が強く示唆されている因子である。前記 AMの受容体は、 CRLRと RA MP2又は RAMP3との複合体で構成される点で、 CRLRと RAMP1との複合体で構 成される CGRP受容体と類似して ヽることから、 CGRP受容体との機能上の類似性 が示唆される力 CGRP受容体を特徴づける RAMP1と血圧との関連性はよくわかつ ていない。 [0005] On the other hand, adrenomedullin (AM) is a potent vasorelaxing peptide that has been strongly suggested to be associated with blood pressure and circulatory dynamics. Since the AM receptor is composed of a complex of CRLR and RAMP2 or RAMP3, it is similar to the CGRP receptor composed of a complex of CRLR and RAMP1, and thus CGRP receptor Forces suggesting functional similarity to the body The relationship between RAMP1 that characterizes the CGRP receptor and blood pressure is not well understood.
[0006] 特許文献 1:特表 2005— 503809号公報  [0006] Patent Document 1: Japanese Translation of Special Publication 2005-503809
特許文献 2 :特許第 3639555号  Patent Document 2: Patent No. 3639555
非特許文献 1 : Science. 1997 Feb 14;275(5302):964-967  Non-Patent Document 1: Science. 1997 Feb 14; 275 (5302): 964-967
非特許文献 2 : Nat Med. 1999 Apr;5(4) :434-8  Non-Patent Document 2: Nat Med. 1999 Apr; 5 (4): 434-8
非特許文献 3 : J. Exp. Med. 2002 Jan 21;195:151- 160  Non-Patent Document 3: J. Exp. Med. 2002 Jan 21; 195: 151-160
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0007] 本発明の目的は、血管障害性疾患の治療に有用な病態 (血管収縮性障害に起因 する高血圧)モデル非ヒト動物、及び当該動物を用いて血管障害性疾患の予防及び[0007] An object of the present invention is to prevent a vascular disorder disease using a pathological condition (hypertension due to vasoconstrictive disorder) model non-human animal useful for the treatment of a vascular disorder disease, and the animal.
Z又は治療用薬剤を効率よくスクリーニングする方法、該方法により得られる薬剤を 提供することにある。 An object of the present invention is to provide a method for efficiently screening for Z or a therapeutic drug, and a drug obtained by the method.
本発明の他の目的は、高血圧モデル非ヒト動物を用いて機能性食品を簡易にスク リーニングする方法及び評価する方法、及び該方法により得られる機能性食品を提 供することにある。  Another object of the present invention is to provide a method for easily screening a functional food using a non-human animal model of hypertension, a method for evaluating the functional food, and a functional food obtained by the method.
課題を解決するための手段  Means for solving the problem
[0008] 本発明者らは、上記目的を達成するため鋭意検討した結果、染色体上の RAMP1 遺伝子の一部又は全部の改変により、当該 RAMP1遺伝子の機能が完全に欠損して いる動物を高血圧モデル動物として利用できること、このような高血圧モデル動物は 、血管障害性疾患の予防や治療用の薬剤のスクリーニングや、機能性食品の評価に 極めて有用であることを見出し、本発明を完成した。 [0008] As a result of intensive investigations to achieve the above object, the present inventors have found that the function of the RAMP1 gene is completely deficient due to partial or complete modification of the RAMP1 gene on the chromosome. The present invention can be used as a high blood pressure model animal, and such a high blood pressure model animal has been found to be extremely useful for screening for drugs for the prevention and treatment of vascular disorder diseases and for evaluation of functional foods. completed.
[0009] すなわち、本発明は、染色体上の RAMP1遺伝子の一部又は全部の改変により、当 該 RAMP1遺伝子の機能が完全に欠損していることを特徴とする高血圧モデル非ヒト 動物を提供する。本発明の非ヒト動物は、染色体上に選択マーカー遺伝子が挿入さ れていないことが好ましい。  [0009] That is, the present invention provides a hypertensive model non-human animal characterized in that the function of the RAMP1 gene is completely deficient by modification of part or all of the RAMP1 gene on the chromosome. The non-human animal of the present invention preferably has no selection marker gene inserted on the chromosome.
[0010] 本発明の高血圧モデル非ヒト動物は、例えば、染色体上に、 RAMP1遺伝子のェキ ソンの少なくとも一部が LoxP配列に挟まれた領域と、選択マーカー遺伝子が LoxP配 列に挟まれた領域とを含む ES細胞に、 Cre酵素を作用させて得られる ES細胞を用 いる方法、又は染色体上に、 RAMP1遺伝子のェキソンの少なくとも一部が LoxP配列 に挟まれた領域を含む ES細胞を用いて作製された RAMPl-floxedマウスと、 Cre酵素 を発現可能なマウスとの交配による方法の 、ずれかで作製することができる。本発明 において、 RAMP1遺伝子の発現がコンデイショナルに抑制されていてもよい。前記非 ヒト動物は、例えばラット又はマウスである。  [0010] The hypertensive model non-human animal of the present invention has, for example, a region in which at least a part of the RAMP1 gene exon is sandwiched between LoxP sequences and a selectable marker gene is sandwiched between LoxP sequences on a chromosome. Using ES cells obtained by allowing Cre enzyme to act on ES cells containing a region, or using ES cells containing a region where at least a part of the RAMP1 gene exon is sandwiched between LoxP sequences on the chromosome The RAMPl-floxed mouse prepared in this way and the method of mating with a mouse capable of expressing the Cre enzyme can be produced by a deviation. In the present invention, the expression of the RAMP1 gene may be conditionally suppressed. The non-human animal is, for example, a rat or a mouse.
[0011] 本発明は、上記本発明の高血圧モデル非ヒト動物に被検物質を投与するか、又は 該動物由来の組織、器官、若しくは細胞を被検物質と接触させることを特徴とする血 管障害性疾患の予防及び Z又は治療用薬剤のスクリーニング方法を提供する。前 記方法は、及び Z又は血管拡張作用を指標として行うことができる。本発明における 血管障害性疾患には、例えば高血圧、血管内皮障害、動脈硬化性疾患、及び記憶 障害,認知症,末梢血流障害性疾患を含む血流障害性疾患が含まれる。  [0011] The present invention provides a blood vessel characterized by administering a test substance to the above-mentioned hypertension model non-human animal of the present invention or bringing a tissue, organ or cell derived from the animal into contact with the test substance. Disclosed is a method for screening for drugs for the prevention and Z or treatment of disability diseases. The above method can be performed using Z or vasodilatory action as an index. The vascular disorder diseases in the present invention include, for example, hypertension, vascular endothelial disorder, arteriosclerotic disease, and blood flow disorder diseases including memory disorder, dementia, peripheral blood flow disorder disease.
[0012] 本発明は、また、上記本発明の薬剤スクリーニング方法より得られる血管障害性疾 患の予防及び Z又は治療薬を提供する。  [0012] The present invention also provides a preventive and Z or therapeutic agent for a vascular disorder disease obtained by the drug screening method of the present invention.
[0013] さらに、本発明は、 RAMP1遺伝子の発現又は機能を亢進する物質を有効成分とし て含有する血流改善薬を提供する。  [0013] Furthermore, the present invention provides a blood flow improving drug containing, as an active ingredient, a substance that enhances the expression or function of the RAMP1 gene.
[0014] また、本発明は、 RAMP1遺伝子の発現又は機能を亢進する物質を有効成分として 含有する血管障害性疾患の予防及び Z又は治療薬を提供する。 [0014] The present invention also provides a prophylactic and Z or therapeutic agent for vascular disorder diseases containing a substance that enhances the expression or function of the RAMP1 gene as an active ingredient.
[0015] 本発明は、上記本発明の高血圧モデル非ヒト動物に被検物質を投与するか、又は 該動物由来の組織、器官、若しくは細胞を被検物質と接触させることを特徴とする機 能性食品のスクリーニング方法を提供する。前記方法は、血圧及び Z又は血管拡張 作用を指標として行うことができる。 [0015] In the present invention, a test substance is administered to the above-described hypertension model non-human animal of the present invention, or There is provided a screening method for functional food, which comprises contacting a tissue, organ or cell derived from the animal with a test substance. The method can be performed using blood pressure and Z or vasodilating action as indices.
[0016] 本発明は、上記本発明の機能性食品のスクリーニング方法により得られる機能性食 品を提供する。  [0016] The present invention provides a functional food obtained by the functional food screening method of the present invention.
[0017] さらに、本発明は、上記本発明の高血圧モデル非ヒト動物に被検物質を投与する 力 又は該動物由来の組織、器官、若しくは細胞を被検物質と接触させることを特徴 とする機能性食品の評価方法を提供する。前記方法は、血圧及び Z又は血管拡張 作用を指標として行うことができる。  [0017] Further, the present invention is a function characterized in that the test substance is administered to the non-human hypertensive model non-human animal of the present invention, or a tissue, organ or cell derived from the animal is brought into contact with the test substance. A method for evaluating sex foods is provided. The method can be performed using blood pressure and Z or vasodilating action as indices.
発明の効果  The invention's effect
[0018] 本発明によれば 染色体上の RAMP1遺伝子の改変により、当該 RAMP1遺伝子の 機能を完全に欠損させることにより、血管拡張不全に起因する恒常的に高血圧を示 すモデル非ヒト動物を得ることができる。本発明の高血圧モデル非ヒト動物を用いるこ とにより、血管障害性疾患の予防及や治療に有用な薬剤を効率よくスクリーニングで き、また被検物質の薬効を簡易に評価することができる。このような高血圧モデル非ヒ ト動物は、また、血圧の低下や血管弛緩作用を発揮し、高血圧や血流障害を予防あ るいは改善する機能性食品のスクリーニング (選別)や評価に利用できる。  [0018] According to the present invention, a model non-human animal that constantly shows hypertension due to vasodilatation failure is obtained by completely deleting the function of the RAMP1 gene by modifying the RAMP1 gene on the chromosome. Can do. By using the hypertensive model non-human animal of the present invention, a drug useful for the prevention and treatment of vascular disorder disease can be efficiently screened, and the efficacy of the test substance can be easily evaluated. Such non-hypertensive animals with hypertension can also be used for screening (selection) and evaluation of functional foods that exhibit a blood pressure lowering and blood vessel relaxation action to prevent or improve hypertension and blood flow disorders.
図面の簡単な説明  Brief Description of Drawings
[0019] [図 1]実施例 1に用いたターゲテイングベクターの概略断面図である。 FIG. 1 is a schematic sectional view of a targeting vector used in Example 1.
[図 2]実施例 1で得た RAMP1 +/+、 RAMP1 +/-、 RAMP1 -/-の各マウス 8週齢の臓器 別発現パターンを示すァガロースゲル電気泳動の写真である。  FIG. 2 is a photograph of agarose gel electrophoresis showing the expression pattern of each organ of 8-week-old mice of RAMP1 + / +, RAMP1 +/− and RAMP1 − / − obtained in Example 1.
[図 3] (ィ)は実施例 1で得た RAMP1 +/+、 RAMP1 +/-、 RAMP1 -/-の各マウス 10週 齢における血圧を示すグラフであり、(口)は同心拍数を示すグラフである。  [Fig. 3] (i) is a graph showing blood pressure at 10 weeks of age for each of the RAMP1 + / +, RAMP1 +/-, and RAMP1-/-mice obtained in Example 1, and (mouth) is the heart rate. It is a graph to show.
[図 4] (ィ)は実施例 1で得た RAMP1 +/+、 RAMP1 +/-、 RAMP1 -/-の各マウス 6〜18 週齢における血圧を示すグラフであり、(口)は同心拍数を示すグラフである。  [Fig. 4] (i) is a graph showing blood pressure at 6-18 weeks of age for each of the RAMP1 + / +, RAMP1 +/-, and RAMP1-/-mice obtained in Example 1, and (mouth) is the same heart rate. It is a graph which shows a number.
[図 5] (ィ)は実施例 1で得た RAMP1 +/+、 RAMP1 +/-、 RAMP1 -/-の 12週齢マウスに 対して 3種の血管拡張因子の影響下での血管収縮率を示すグラフを示している。 発明を実施するための最良の形態 [0020] RAMP1は、受容体活性変更タンパク質 (RAMP)ファミリーに属し、 N末端にシグナ ル配列を、 C末端に 1回膜貫通ドメインを有する蛋白質である。 RAMP1遺伝子の塩基 配列及び RAMP1のアミノ酸配列は公知であり、例えばヒト及びマウス RAMP1の cDN Aの塩基配列は、 GenBank Accession No. AJ001014及び No. BAA76617で表される 配列等が挙げられる。 [Fig. 5] (i) shows the vasoconstriction rate under the influence of three vasodilators on 12-week-old mice of RAMP1 + / +, RAMP1 +/-, and RAMP1-/-obtained in Example 1. The graph which shows is shown. BEST MODE FOR CARRYING OUT THE INVENTION [0020] RAMP1 belongs to the receptor activity-modifying protein (RAMP) family and has a signal sequence at the N-terminus and a single transmembrane domain at the C-terminus. The nucleotide sequence of RAMP1 gene and the amino acid sequence of RAMP1 are known. For example, the nucleotide sequence of cDNA of human and mouse RAMP1 includes the sequences represented by GenBank Accession No. AJ001014 and No. BAA76617.
[0021] 本発明者らは、 染色体上の RAMP1遺伝子の一部又は全部の改変により、当該 R AMP1遺伝子の機能が完全に欠損させた RAMP1完全欠損型マウスを作製し、その表 現形について詳細な解析を行った。図 3は、野生型(RAMP1+/+)マウス及び RAMP1 ヘテロ接合体 (RAMP1+/-)マウス、 RAMP1ホモ接合体(RAMP1- /- )マウスにつ ヽて 、 6週齢マウスで測定した (ィ)血圧と(口)心拍数のグラフである。図 3 (ィ)に示される ように、心拍数は同程度であるのに対し、血圧については、図 3 (口)に示されるように 、 RAMP1+/+マウスや RAMP1+/-マウスと比較して、 RAMP1- /-マウスは 6週齢という 極めて早い段階ですでに高い。図 4は、前記 RAMPl+/+、 RAMPl+/-、 RAMP1-/-の 各マウスについて、 6週齢から 18週齢までの 3週ごとに測定した (ィ)血圧と(口)心拍 数のグラフである。図 4 (ィ)に示されるように、心拍数は 6週齢力 18週齢まで同程度 であるのに対し、血圧については、図 4 (口)に示されるように、 RAMP1-/-マウスは 6 週齢という極めて早い段階ですでに高い値を示し、週齢とともに徐々に高くなる傾向 にあった。またこの高血圧症は CGRPによる血管拡張作用が完全に抑制されたことに 起因することも明らかになった.このように、 RAMP1欠損マウスは、生後早い段階から 血管収縮による高血圧の状態が維持されて 、ることから、高血圧及び血管拡張障害 モデルとして利用することが可能である。また、 RAMP1遺伝子には、ヒト、ラット、ブタ、 ァカゲザル等のホモログの存在が知られており、これらの塩基配列は GenBankに公 開されている。従って、マウス以外の非ヒト動物についても同様に高血圧及び血管拡 張障害モデルとして利用可能である。  [0021] The present inventors have produced a RAMP1 completely deficient mouse in which the function of the RAMP1 gene has been completely deleted by modifying a part or all of the RAMP1 gene on the chromosome, and the expression form is detailed. Analysis was performed. Figure 3 shows wild-type (RAMP1 + / +), RAMP1 heterozygous (RAMP1 +/-), and RAMP1 homozygous (RAMP1-/-) mice measured in 6-week-old mice. It is a graph of blood pressure and (mouth) heart rate. As shown in Fig. 3 (ii), the heart rate is comparable, while blood pressure is compared to RAMP1 + / + mice and RAMP1 +/- mice as shown in Fig. 3 (mouth). RAMP1-/-mice are already very high at 6 weeks of age. Fig. 4 is a graph of (i) blood pressure and (mouth) heart rate measured for each of the RAMPl + / +, RAMPl +/-, and RAMP1-/-mice every 3 weeks from 6 to 18 weeks of age. is there. As shown in Fig. 4 (i), the heart rate is about the same up to 6-week-old power up to 18-week-old, whereas blood pressure is about RAMP1-/-mice as shown in Fig. 4 (mouth). Was already very high at 6 weeks of age and tended to increase gradually with age. It was also revealed that this hypertension was caused by the complete suppression of the vasodilatory effect of CGRP. Thus, RAMP1-deficient mice maintained the state of hypertension due to vasoconstriction from an early age. Therefore, it can be used as a model for hypertension and vasodilatation disorder. In addition, the RAMP1 gene is known to have homologues such as human, rat, pig, and macaque monkeys, and these nucleotide sequences are disclosed in GenBank. Therefore, non-human animals other than mice can also be used as models for hypertension and vasodilatation disorders.
[0022] 本発明の高血圧モデル非ヒト動物(以下、「モデル動物」と称する場合がある)は、 染色体上の RAMP1遺伝子の一部又は全部の改変により、当該 RAMP1遺伝子の機 能が完全に欠損して 、ることを特徴として 、る。「遺伝子の機能が完全に欠損して ヽ る」とは、例えば、野生型 RAMP1が本来有する機能 (例えば CGRP受容体を構成する 機能等)が完全に失われていることを意味している。すなわち、本発明のモデル動物 は、個体内における RAMP1タンパクによる影響 (作用)が全くないか無視できる程度 であって、 RAMP1のドミナントネガティブモデルやドミナントアクティブモデルを含むも のではない。 [0022] The hypertensive model non-human animal of the present invention (hereinafter sometimes referred to as "model animal") is completely deficient in the function of the RAMP1 gene due to partial or complete modification of the RAMP1 gene on the chromosome. It is characterized by that. “The gene function is completely deficient” means, for example, the function that wild-type RAMP1 originally has (for example, it constitutes the CGRP receptor). Function etc.) is completely lost. That is, the model animal of the present invention has no influence (action) due to RAMP1 protein in the individual or can be ignored, and does not include a dominant negative model or dominant active model of RAMP1.
[0023] 本発明にお 、て、染色体上の RAMP1遺伝子の機能が完全に欠損された非ヒト動物 は、 ES細胞を用いたノックアウト非ヒト動物を作製する方法として公知の方法に従つ て作製することができる。  [0023] In the present invention, a non-human animal in which the function of the RAMP1 gene on the chromosome is completely deleted is prepared according to a method known as a method for producing a knockout non-human animal using ES cells. can do.
[0024] 本発明のモデル動物の構成としては、例えば、染色体上の RAMP1遺伝子の全領 域 (例えばェキソン 1〜3)が除去された構成;染色体上の RAMP1遺伝子の一部の改 変、例えば塩基配列に欠失、付加、置換により、遺伝子の機能が完全に欠損された 構成等が挙げられる。前者の構成によれば、個体内に転写'翻訳産物が一切生成さ れず、 RAMP1遺伝子による影響が完全に除去されたモデル動物として有用である。 また、後者の構成としては、遺伝子の機能が完全に欠損されれば特に限定されず、 R AMP1遺伝子の適宜な部位、範囲に改変を施すことができ、部分的な転写'翻訳によ る mRNAやポリペプチド断片が生成されていてもよい。なかでも、本発明のモデル動 物としては、染色体上の RAMP1遺伝子の少なくともェキソン 2が除去された構成であ ることが好ましい。前記ェキソン 2には成熟蛋白質コード開始部位が配置されている。 従って、ェキソン 2が除去されたモデル動物においては、 CGRP受容体を構成する RA MP1のポリペプチド断片が形成されず、該ポリペプチド断片の生成に伴う CGRP受容 体機能などへの影響を一切排除することができるため、血流障害性疾患を含む血管 障害性疾患の予防薬や治療薬等のスクリーニング (評価を含む)同疾患自体のメカ -ズムの研究用途等の広範な用途に好適である。  [0024] The model animal of the present invention includes, for example, a configuration in which the entire region of the RAMP1 gene on the chromosome (for example, exons 1 to 3) has been removed; a partial modification of the RAMP1 gene on the chromosome, for example, Examples include a configuration in which the gene function is completely deleted by deletion, addition, or substitution in the base sequence. According to the former configuration, no transcription'translation product is generated in the individual, and it is useful as a model animal in which the influence of the RAMP1 gene is completely eliminated. In addition, the latter structure is not particularly limited as long as the gene function is completely deleted, and an appropriate site and range of the RAMP1 gene can be modified, and mRNA by partial transcription and translation can be used. Or a polypeptide fragment may be produced. Among them, the model animal of the present invention preferably has a configuration in which at least exon 2 of the RAMP1 gene on the chromosome is removed. Exon 2 has a mature protein coding start site. Therefore, in the model animals from which exon 2 has been removed, the polypeptide fragment of RAMP1 that constitutes the CGRP receptor is not formed, and any influence on the CGRP receptor function associated with the production of the polypeptide fragment is completely eliminated. Therefore, it is suitable for a wide range of uses such as screening (including evaluation) of preventive and therapeutic agents for vascular disorder diseases including blood flow disorder diseases (including evaluation) and research of the mechanism of the disease itself.
[0025] このような高血圧モデル非ヒト動物の作製方法としては、染色体上の RAMP1遺伝子 の機能を完全に欠損することができれば特に限定されな 、が、例えば CreZLoxPシ ステムを利用する方法が挙げられる。 CreZLoxPシステムは、 Cre酵素が LoxP配列を 認識して特異的組換えを起こすシステムであり、該システムの利点としては、例えば、 DNA領域の欠失可能域が相同組換えは 20kb程度が限度であるのに対して 200kb 程度と大きぐジーンターゲティングにおいて染色体上力 選択マーカー遺伝子の除 去が容易な手法であり、点突然変異を容易に導入でき、さらに Cre酵素の発現方法 によってコンデイショナルジーンターゲテイングが可能であること等が挙げられる。 [0025] The method for producing such a hypertensive model non-human animal is not particularly limited as long as it can completely lack the function of the RAMP1 gene on the chromosome, but for example, a method using the CreZLoxP system can be mentioned. . The CreZLoxP system is a system in which the Cre enzyme recognizes the LoxP sequence and causes specific recombination. The advantage of this system is that, for example, the region where the DNA region can be deleted is limited to about 20 kb for homologous recombination In contrast to gene targeting, which is about 200 kb This is an easy method, and point mutations can be easily introduced, and conditional gene targeting is possible by the expression method of Cre enzyme.
[0026] 本発明における CreZLoxPシステムの利用方法としては、例えば、 ES細胞で Cre酵 素を一過性に発現させて薬剤選択マーカーを除く工程に利用する方法を用いること ができ、具体的には、染色体上に、 RAMP1遺伝子のェキソンの少なくとも一部が Lox P配列に挟まれた領域と、選択マーカー遺伝子が LoxP配列に挟まれた領域とを含む ES細胞(RAMPl-floxed- markerES細胞)に、 Cre酵素を作用させて得られる ES細胞 を用いる方法が挙げられる。なかでも、前者の LoxP配列が染色体上の RAMP1遺伝 子の少なくとも成熟蛋白質コード開始部位を含む配列(例えば eX0n2等)を挟んで 、 る場合には、 Cre酵素の作用により RAMP1蛋白質の完全欠損型を容易に構成するこ とができ好ましい。 [0026] As a method of using the CreZLoxP system in the present invention, for example, a method in which Cre enzyme is transiently expressed in ES cells and used in a step of removing a drug selection marker can be used. Specifically, In an ES cell (RAMPl-floxed-marker ES cell) containing a region where at least a part of the exon of the RAMP1 gene is sandwiched between LoxP sequences and a region where a selectable marker gene is sandwiched between LoxP sequences on the chromosome, A method using ES cells obtained by the action of Cre enzyme can be mentioned. In particular, when the former LoxP sequence sandwiches a sequence containing at least the mature protein coding start site of the RAMP1 gene on the chromosome (eg, eX0 n2), the RAMP1 protein is completely defective due to the action of the Cre enzyme. Can be easily configured.
[0027] 図 1は、前記 RAMPl-floxed- markerES細胞に、 Cre酵素を作用させて得られる ES 細胞の作製方法の一例を示す模式図である。本発明の高血圧モデル非ヒト動物の 作製に用いる ES細胞は、例えば、図 1に示されるように、(i)ES細胞の染色体上にお ける exonl〜3からなる RAMP1遺伝子に対し、(ii)LoxP配列に挟まれた第 1の選択マー カー遺伝子(チミジンキナーゼ: TK)、 LoxP配列に挟まれた RAMP1遺伝子の exon2、 及び LoxP配列に挟まれた第 2の選択マーカー遺伝子 (ネオマイシン: neo)からなる構 成を含むターゲテイングベクターを用いて相同組換えを起こさせ、 TK活性を指標とし て、(iii)染色体上に、 LoxP配列に挟まれた RAMP1遺伝子の exon2及び LoxP配列に 挟まれた neo遺伝子の配列が組み込まれた RAMPl-floxed- markerES細胞を選択し、 これに Cre酵素を作用させて特異的組換えを起こさせ、(iv)染色体上の exon2と neo遺 伝子とを除去することにより得ることができる。なお、前記 (iv)に残る一つの LoxP配列 は、標的遺伝子の発現に影響しないことが確認されている。図 1の方法によれば、染 色体上から RAMP1遺伝子の exon2が除去されるため、 RAMP1遺伝子の一部又は全 部の転写 Z翻訳産物の生成が完全に抑制される。こうして得られる ES細胞は、染色 体上の RAMP1遺伝子の機能が完全に欠損され、且つ選択マーカー遺伝子の挿入 がない。次いで、トランスジヱニック動物を作製する公知の方法に従い、上記構成を 有する ES細胞由来の個体を得ることができる。 [0028] 本発明における CreZLoxPシステムの利用方法の他の例として、染色体上に、 RA MP1遺伝子のェキソンの少なくとも一部が LoxP配列に挟まれた領域を含む ES細胞 を用いて作製された RAMPl-floxedマウスと、 Cre酵素を発現可能なマウスとの交配に よる方法が挙げられる。前記 RAMPl-floxedマウスとしては、染色体上に、 RAMP1遺 伝子のェキソンの少なくとも一部が LoxP配列で挟まれた領域を含んでいれば特に限 定されないが、例えば、 RAMP1遺伝子のェクソンを 2つの ΙοχΡ配列で挟む形で相同 組換えさせ、それ自体では遺伝子が損なわれて 、な 、マウスなどを用いることができ る。前記 Cre酵素を発現可能なマウスとしては、例えば Cre酵素発現ベクターを導入 する方法等の公知の方法で作製することができる。なかでも、 Cre酵素を発現可能な マウスとして、糸且織特異的又は発現誘導可能なプロモーター制御下に Cre酵素を発 現可能なマウス(Cre酵素コンデイショナル発現マウス)等を用いることにより、 RAMP1 遺伝子のコンデイショナルジーンターゲティングを容易に行うことができる。 [0027] FIG. 1 is a schematic diagram showing an example of a method for producing ES cells obtained by allowing a Cre enzyme to act on the RAMPl-floxed-marker ES cells. For example, as shown in FIG. 1, ES cells used for the production of a hypertensive model non-human animal of the present invention are (ii) against the RAMP1 gene consisting of exonl to 3 on the chromosome of the ES cell (ii) From the first selectable marker gene (thymidine kinase: TK) sandwiched between LoxP sequences, exon2 of the RAMP1 gene sandwiched between LoxP sequences, and the second selectable marker gene (neomycin: neo) sandwiched between LoxP sequences Using a targeting vector comprising the following structure, homologous recombination is caused, and TK activity is used as an index. (Iii) On the chromosome, the RAMP1 gene sandwiched between the exon2 and LoxP sequences sandwiched between the LoxP sequences and neo Select RAMPl-floxed-marker ES cells into which the gene sequence has been integrated, and then act on the Cre enzyme to cause specific recombination, and (iv) remove exon2 and neo genes on the chromosome Can be obtained. It has been confirmed that the one LoxP sequence remaining in (iv) does not affect the expression of the target gene. According to the method of FIG. 1, since exon2 of the RAMP1 gene is removed from the chromosome, the generation of a transcription Z translation product of part or all of the RAMP1 gene is completely suppressed. The ES cells obtained in this way are completely deficient in the function of the RAMP1 gene on the chromosome and have no insertion of a selectable marker gene. Subsequently, an ES cell-derived individual having the above-described configuration can be obtained according to a known method for producing a transgenic animal. [0028] As another example of the method of using the CreZLoxP system in the present invention, RAMPl- prepared using ES cells containing a region on the chromosome where at least a part of the exon of the RAMP1 gene is sandwiched between LoxP sequences. An example is a method of mating floxed mice with mice capable of expressing the Cre enzyme. The RAMPl-floxed mouse is not particularly limited as long as it contains at least part of the RAMP1 gene exon sandwiched between LoxP sequences on the chromosome. For example, two RAMP1 gene exons are included. By homologous recombination in a form sandwiched between ΙοχΡ sequences, the gene itself is damaged, and mice can be used. The mouse capable of expressing the Cre enzyme can be prepared by a known method such as a method of introducing a Cre enzyme expression vector. In particular, as a mouse capable of expressing the Cre enzyme, the RAMP1 gene can be obtained by using a mouse capable of expressing the Cre enzyme under the control of a yarn and tissue-specific or expression-inducible promoter (Cre enzyme conditional expression mouse). The conditional gene targeting can be easily performed.
[0029] 前記染色体上の RAMP1遺伝子の欠損は、恒常的、コンデイショナル [組織特異的、 時期特異的(一過性)等]のいずれであってもよい。なかでも、 RAMP1遺伝子の発現 力 Sコンデイショナルに抑制されて 、る場合は、高血圧モデルを所望の条件下で構築 することができる点で好ましぐ例えば、上記 Cre酵素をコンデイショナルに発現する マウスを用いる方法等の公知の方法により構築することができる。  [0029] The RAMP1 gene deficiency on the chromosome may be either constitutive or conditional [tissue-specific, time-specific (transient), etc.]. In particular, if the expression level of RAMP1 gene is suppressed S-conditionally, it is preferable in that a hypertension model can be constructed under a desired condition. For example, a mouse that expresses the above Cre enzyme in a conditional manner. It can construct | assemble by well-known methods, such as the method of using.
[0030] 本発明の血管障害性疾患の予防及び Z又は治療薬のスクリーニング方法は、上記 本発明の高血圧モデル非ヒト動物に被検物質を投与するか、又は該動物由来の組 織、器官、若しくは細胞を被検物質と接触させることを特徴としている。以下、前者を「 スクリーニング方法 1」、後者を「スクリーニング方法 2」と称する場合がある。  [0030] The method for screening a prophylactic and Z or therapeutic agent for a vascular disorder disease of the present invention comprises administering a test substance to the above-mentioned hypertension model non-human animal of the present invention, or a tissue, organ derived from the animal, Or it is characterized by making a cell contact a test substance. Hereinafter, the former may be referred to as “screening method 1” and the latter as “screening method 2”.
[0031] 本発明のスクリーニング方法(1、 2)に供される被検物質は、公知又は新規化合物 の何れであってもよぐ例えば、核酸、糖質、脂質、蛋白質、ペプチド、有機低分子化 合物等の合成又は天然由来の有機化合物を用いることができる。  [0031] The test substance to be used in the screening method (1, 2) of the present invention may be any known or novel compound. For example, nucleic acids, carbohydrates, lipids, proteins, peptides, small organic molecules Synthetic or natural organic compounds such as compounds can be used.
[0032] スクリーニング方法 1にお 、て、被検物質をモデル動物へ投与する方法は、特に限 定されず、経口及び非経口投与の何れであってもよい。非経口投与には、例えば、 静脈、動脈、筋肉、腹腔、気道等を経路とする全身投与、又は虚血部位又はその近 傍への局所投与を利用できるが、これらに限定されない。好ましくは非経口投与、より 好ましくは血管又はその近傍へ局所投与される。被検物質の投与量は、被検物質の 種類、投与方法、モデル動物の種類、大きさ等に応じて適宜設定できる。 [0032] In screening method 1, the method of administering the test substance to the model animal is not particularly limited, and may be either oral or parenteral administration. For parenteral administration, for example, systemic administration via the vein, artery, muscle, abdominal cavity, airway, etc., or local administration at or near the ischemic site can be used, but is not limited thereto. Preferably parenteral administration, more Preferably, it is locally administered to or near the blood vessel. The dose of the test substance can be appropriately set according to the type of test substance, the administration method, the type, size, etc. of the model animal.
[0033] スクリーニング方法 1における評価は、例えばモデル動物の血圧値を指標に行うこ とができる。具体的には、例えば、本発明の高血圧モデル非ヒト動物に対し、特定量 の被験物質を、 1日 3回を 2週間(回数、日数等の投与条件)の条件下で投与した後 の収縮期血圧値が、対応する野生型動物の平均血圧値の 130%以下程度 (好ましく は 120%以下程度)を示したときに、当該被検物質が血管障害性疾患予防及び Z又 は治療用の薬剤として有用であると評価することができる。モデル動物がマウスの例 では、例えば、平均血圧約 lOOmmHgの野生型マウスと比較して平均血圧約 140mmH gを示す本発明の高血圧モデルマウスに、被験物質を 1日 3回 2週間投与した後の血 圧が 130mmHg以下 (好ましくは 120mmHg)を示したときに、当該被験物質が血管障害 性疾患予防及び Z又は治療用の薬剤として有用であると評価することができる。  [0033] The evaluation in screening method 1 can be performed using, for example, the blood pressure value of the model animal as an index. Specifically, for example, the contraction after administering a specific amount of the test substance to the non-human animal model of hypertension of the present invention 3 times a day for 2 weeks (administration conditions such as frequency and days). When the blood pressure value for the period shows about 130% or less (preferably about 120% or less) of the average blood pressure value of the corresponding wild-type animal, the test substance is used for vascular disorder disease prevention and Z or treatment. It can be evaluated that it is useful as a medicine. In the case where the model animal is a mouse, for example, after the test substance is administered 3 times a day for 2 weeks to the hypertensive model mouse of the present invention showing an average blood pressure of about 140 mmHg as compared with a wild-type mouse having an average blood pressure of about lOOmmHg. When the blood pressure is 130 mmHg or less (preferably 120 mmHg), it can be evaluated that the test substance is useful as a drug for the prevention and Z or treatment of vascular disorder diseases.
[0034] スクリーニング方法 2において、被検物質をモデル動物由来の組織、器官、若しく は細胞と接触させる方法としては、特に限定されず、モデル動物力 採取した組織や 器官の標本に被験物質を投入する方法、モデル動物から採取した細胞を被験物質 存在下で培養する方法等の公知の方法を利用できる。前記組織や器官の標本に被 験物質を投入する方法としては、上述した非経口投与と同様の方法を用いることがで きる。  In screening method 2, the method of bringing a test substance into contact with a tissue, organ, or cell derived from a model animal is not particularly limited, and the test substance is applied to a sample of tissue or organ collected from model animal power. A known method such as a method of adding or culturing cells collected from a model animal in the presence of a test substance can be used. As a method of introducing the test substance into the tissue or organ specimen, the same method as the parenteral administration described above can be used.
[0035] スクリーニング方法 2における評価は、例えば、血管収縮率を指標に行うことができ る。具体的には、例えば、本発明の高血圧モデル非ヒト動物から採取した血管標本 に対し、血管収縮剤を投与した後、 a CGRP及び被験物質を累積添加して ED で比  [0035] The evaluation in the screening method 2 can be performed using, for example, a vasoconstriction rate as an index. Specifically, for example, a vasoconstrictor is administered to a blood vessel sample collected from a non-human animal model of hypertension of the present invention, and then a CGRP and a test substance are cumulatively added, and the ratio is compared by ED.
50 較した場合,あるいは濃度曲線下面積 (AUC)で比較した場合に有意差が認められ たときに、当該被検物質が血管障害性疾患予防及び Z又は治療用の薬剤として有 用であると評価することができる。モデル動物がマウスの例では、例えば、血管標本 を作成し、フエ-レフリン(10— 6M)添カ卩により血管を収縮させた後、 a CGRP及び被験 物質を各終濃度 lx 10— u〜3xl0— 6Mとなるように累積添加したときの張力変化を測定 し、換算して得られた血管収縮率が、被検物質の累積添カ卩により場合により ED で比 When a significant difference is observed when comparing 50 or by the area under the concentration curve (AUC), the test substance is considered to be useful as a drug for the prevention and Z or treatment of vascular disorders. Can be evaluated. In the example of an animal model mouse, for example, create a blood vessel specimens Hue - Refurin (10- 6 M) after the vessel was contracted by添Ka卩, a CGRP and test substance each final concentration lx 10- u ~ 3Xl0- 6 measures the tension variation when the cumulative added to a M, vasoconstriction refraction obtained by conversion is a ratio with ED optionally by cumulative added mosquito卩analyte
50 較した場合,あるいは AUCで比較した場合に有意差が認められたときに、その被検 物質が血管障害性疾患防及び Z又は治療用の薬剤として有用であると評価すること ができる。前記血管標本の張力変化は、例えば FDピックアップとキヤリヤーアンプを 介して等尺性にレコーダー(model 8K21; NEC)を用いて測定できる。 If there is a significant difference when comparing 50 or AUC, the test It can be evaluated that the substance is useful as a drug for prevention and Z or treatment of vascular disorder diseases. The change in tension of the blood vessel specimen can be measured using a recorder (model 8K21; NEC) isometrically via, for example, an FD pickup and a carrier amplifier.
[0036] 本発明のスクリーニング方法により、血管障害性疾患の予防や治療に有用な薬剤 を得ることができる。本発明における血管疾患には、高血圧、及び高血圧が誘因とな る疾病 (合併症)、例えば血管内皮障害、動脈硬化性疾患、及び血流障害性疾患等 が含まれる。動脈硬化性疾患には、例えば脳梗塞、脳出血、くも膜下出血、あるいは 脳血管障害系疾患;心筋梗塞や狭心症などの冠動脈系疾患;大動脈瘤、大動脈解 離等の大動脈系疾患;腎動脈硬化症やそれによる腎不全等の腎動脈系疾患;及び 閉塞性動脈硬化症等の末梢動脈系疾患等が含まれる。血流障害性疾患には,例え ば記憶障害,認知症,末梢血流障害性疾患等が含まれる。 [0036] By the screening method of the present invention, a drug useful for the prevention or treatment of vascular disorder disease can be obtained. The vascular disease in the present invention includes hypertension and diseases (complications) caused by hypertension, such as vascular endothelial disorder, arteriosclerotic disease, and blood flow disorder. Examples of atherosclerotic diseases include cerebral infarction, cerebral hemorrhage, subarachnoid hemorrhage, or cerebrovascular disorder diseases; coronary artery diseases such as myocardial infarction and angina; aortic diseases such as aortic aneurysm and aortic dissection; It includes renal arterial diseases such as sclerosis and resulting renal failure; and peripheral arterial diseases such as obstructive arteriosclerosis. Examples of blood flow disorders include memory disorders, dementia, and peripheral blood flow disorders.
[0037] 本発明の血管障害性疾患の予防及び Z又は治療薬 (以下、「薬剤」と称する場合 がある)としては、上記スクリーニング方法により得られる被検物質を有効成分として 含んでいる。このような薬剤は、例えば、適宜なビヒクルに溶解又は懸濁して、経口又 は非経口投与される。非経口投与には、例えば、静脈、動脈、筋肉、腹腔、気道等を 経路とする全身投与、又は血管又はその近傍への局所投与を利用できるが、これら に限定されない。疾患の症状に応じて、経口投与が好ましく行われ、また血管又はそ の近傍へ非経口的に局所投与することもできる。前記薬剤は、例えば、 RAMP1遺伝 子の発現又は機能を亢進する物質を収縮血管部位又はその近傍の細胞に安定に 到達させ、細胞膜を透過し、リソソーム Zエンドソーム力 の薬剤の遊離を促進しうる ドラッグデリバリーシステムの設計がされて 、ることも好ま 、。  [0037] The preventive and Z or therapeutic agent (hereinafter sometimes referred to as "drug") for vascular disorder disease of the present invention contains a test substance obtained by the screening method as an active ingredient. Such drugs are administered orally or parenterally, for example, dissolved or suspended in a suitable vehicle. For parenteral administration, for example, systemic administration via the vein, artery, muscle, abdominal cavity, airway, etc., or local administration to or near blood vessels can be used, but is not limited thereto. Depending on the symptoms of the disease, oral administration is preferably performed, and can also be administered parenterally to the blood vessel or the vicinity thereof. The drug can, for example, cause a substance that enhances the expression or function of the RAMP1 gene to stably reach the contracted blood vessel site or a cell in the vicinity thereof, permeate the cell membrane, and promote the release of the drug with lysosomal Z endosome force. It is also preferable that the delivery system is designed.
[0038] 前記薬剤の剤形としては、液剤、固形剤の何れであってもよぐ例えば、水、生理食 塩水等の希釈液又は分散媒に有効量の阻害剤を溶解、分散、乳化させた液剤;有 効量の阻害剤を粉末、顆粒状等で含む DDS製剤、サッシェ剤、錠剤等の固形剤など が挙げられる。これらの薬剤には、医薬上許容される公知の添加剤を添加することが できる。  [0038] The drug dosage form may be either liquid or solid. For example, an effective amount of an inhibitor is dissolved, dispersed, or emulsified in a diluent or dispersion medium such as water or physiological saline. Liquid preparations; DDS preparations containing an effective amount of an inhibitor in the form of powder, granules, solid preparations such as sachets and tablets. These drugs can be added with known pharmaceutically acceptable additives.
[0039] 薬剤は、また、有効成分がリボソームや徐放性材料等に封入された封入体や担体 に担持された担持体などであってもよい。リボソーム等に封入することにより、有効成 分をヌクレアーゼゃプロテアーゼによる分解力も保護し、リボソーム膜が細胞表面と 結合してエンドサイト一シスにより細胞内に到達しやすくなる点で有利である。コラー ゲン等の徐放性材料に封入することにより、有効成分の長期持続性が得られる。血 流改善薬には、上記以外に、医薬上許容される公知の添加剤を添加することができ る。 [0039] The drug may be an encapsulated body in which an active ingredient is encapsulated in a ribosome, a sustained-release material, or the like, or a carrier supported on a carrier. By encapsulating in ribosomes, etc. This is advantageous in that the nuclease protects the decomposition ability of the protease and the ribosome membrane binds to the cell surface and easily reaches the cell by endocytosis. Encapsulation in sustained-release materials such as collagen provides long-term sustainability of the active ingredient. In addition to the above, known pharmaceutically acceptable additives can be added to the blood flow improving drug.
[0040] 薬剤の投与量は、有効成分の種類、投与方法、症状、投与対象の種類、大きさ、 薬物特性等に応じて異なるが、通常、成人 1日当たり例えば 0. 5〜50mg程度、好ま しくは l〜20mg程度であり、 1日に 1回、又は複数回に分けて投与することができる。  [0040] The dose of the drug varies depending on the type of active ingredient, administration method, symptom, type of administration target, size, drug characteristics, etc., but is usually preferably about 0.5 to 50 mg per day for an adult. It is about 1 to 20 mg, and can be administered once a day or divided into multiple doses.
[0041] 本発明の血流改善薬は、血管収縮を緩和することにより RAMP1遺伝子の発現又は 機能を亢進する物質を有効成分として含有している。ここで、「発現」とは、蛋白質が 産生されることを意味しており、「発現を亢進する物質」は、遺伝子の転写、転写後調 節、蛋白質への翻訳、翻訳後修飾 (チロシンリン酸化)、蛋白質フォールデイング等の 何れの段階で作用するものであってもよい。また、「機能を亢進する物質」としては、 例えば、 RAMP1タンパク質; RAMP1における CRLR等との蛋白質間相互作用部位や CGRP受容体等の受容体活性部位等の機能部位に作用してシグナル伝達を増加 又は向上する物質; RAMP1又は RAMP1と相互作用する蛋白質のドミナントアクティブ 変異体等であってもよい。ここで、「ドミナントアクティブ変異体」とは、蛋白質に対する 変異の導入等によりその生理活性が向上したものをいう。これらの変異体は野生型と の相乗効果により間接的にその機能を亢進することができる。  [0041] The blood flow improving agent of the present invention contains, as an active ingredient, a substance that enhances the expression or function of the RAMP1 gene by alleviating vasoconstriction. Here, “expression” means that protein is produced, and “substance that enhances expression” means gene transcription, post-transcriptional regulation, translation into protein, and post-translational modification (tyrosine phosphorylation). It may act at any stage such as oxidation) or protein folding. “Substances that enhance function” include, for example, RAMP1 protein; it acts on functional sites such as protein-protein interaction sites with CRLR in RAMP1 and receptor active sites such as CGRP receptors to increase signal transduction. Or a substance that improves; RAMP1 or a dominant active mutant of a protein that interacts with RAMP1. As used herein, “dominant active mutant” refers to a substance whose physiological activity has been improved by introducing a mutation into a protein. These mutants can indirectly enhance their function by a synergistic effect with the wild type.
[0042] RAMP1の発現を亢進する物質としては、例えば、転写促進因子、蛋白質合成促進 剤、蛋白質安定化酵素、スプライシングゃ mRNAの細胞質移行を促進しうる因子、 mRNA安定ィヒ酵素、 mRNAに結合して活性ィヒする因子等が挙げられ、特に他の遺 伝子や蛋白質への副作用を最小限にするため、標的分子に特異的に作用しうる物 質が好ましい。これらの機能性核酸及び蛋白質は、投与後に投与対象内で産生され る形態であってもよぐ必要に応じて発現ベクター、細胞等を用いて公知の方法で調 製することができる。これらの血流改善薬の剤形、投与方法、投与量等は上記薬剤と 同様である。 [0042] Substances that enhance the expression of RAMP1 include, for example, transcription promoting factors, protein synthesis promoters, protein stabilizing enzymes, splicing factors that can promote mRNA cytoplasmic transition, mRNA stabilizing enzymes, and mRNA binding. In particular, a substance capable of acting specifically on the target molecule is preferable in order to minimize side effects on other genes and proteins. These functional nucleic acids and proteins may be produced in the administration subject after administration, and can be prepared by known methods using expression vectors, cells, etc. as necessary. The dosage form, administration method, dosage and the like of these blood flow improving drugs are the same as those for the above drugs.
[0043] 本発明の血管障害性疾患の予防及び Z又は治療薬は、また、 RAMP1遺伝子の発 現又は機能を亢進する物質を有効成分として含有していてもよい。すなわち、上記 血流改善薬を血管障害性疾患の予防薬や治療薬として利用することができる。 [0043] The preventive and Z or therapeutic agent for a vascular disorder disease of the present invention may also generate a RAMP1 gene. A substance that enhances the current or function may be contained as an active ingredient. That is, the above blood flow improving drug can be used as a prophylactic or therapeutic drug for vascular disorder diseases.
[0044] 本発明の高血圧モデル非ヒト動物は、また、機能性食品のスクリーニングや評価に 用いることができる。ここで、「機能性食品」とは、食品が本来持っている栄養機能 (第 1次機能)、味'香りなどの感覚機能 (第 2次機能)〖こ加えて、生態防御や疾病の防止 •回復 ·体調リズムの調整などの生体調節機能 (第 3次機能)があることに着目し、生 体調節を科学的に解明して機能を発揮できるように設計'加工された食品である。す なわち、第 3次機能を有する食品が機能性食品と定義される。  [0044] The non-human animal model of hypertension of the present invention can also be used for screening and evaluation of functional foods. Here, “functional food” means the nutritional function (primary function) inherent in food, sensory functions such as taste fragrance (secondary function), and ecological defense and disease prevention. • Recovery · Food that has been designed and processed so that biological regulation can be clarified scientifically by focusing on the presence of biological regulation functions (third function) such as adjustment of physical rhythm. In other words, foods with tertiary functions are defined as functional foods.
[0045] 本発明の機能性食品のスクリーニング方法及び評価方法に用いる被検物質として は、例えば、ビタミン、ミネラル、オリゴ糖、糖アルコール、アミノ酸、ペプチド、タンパク 質、動物抽出物、乳酸菌や酵母等の微生物及びその抽出物、ハーブや薬草等の植 物体及びその抽出物、配糖体、油脂類等を構成成分とする物質が挙げられる。また 、被検物質の形態としては、特に限定されず、清涼飲料水、炭酸飲料、ドレッシング などの液状調味料等の液状物;テーブルシュガーなどの固体調味料、クッキーなど の菓子、インスタント食品等の固形物等が挙げられる。また、これらの機能性食品は、 錠剤、カプセル等の医薬的形態のものであってもよ 、。  [0045] Examples of test substances used in the screening method and evaluation method for functional foods of the present invention include vitamins, minerals, oligosaccharides, sugar alcohols, amino acids, peptides, proteins, animal extracts, lactic acid bacteria, yeasts, and the like. And microorganisms and extracts thereof, plants such as herbs and medicinal herbs and extracts thereof, glycosides, oils and fats, and the like. In addition, the form of the test substance is not particularly limited, and liquids such as soft drinks, carbonated drinks, liquid seasonings such as dressings; solid seasonings such as table sugar; confectionery such as cookies; and instant foods A solid substance etc. are mentioned. In addition, these functional foods may be in a pharmaceutical form such as tablets and capsules.
[0046] 本発明の機能性食品のスクリーニング方法及び評価方法は、上記本発明の高血 圧モデル非ヒト動物に被検物質を投与する力、又は該動物由来の組織、器官、若し くは細胞を被検物質と接触させることを特徴としている。好ましくは、モデル動物に被 検物質を投与する方法が好まし 、。  [0046] The functional food screening method and evaluation method of the present invention include the ability to administer a test substance to the above-described high blood pressure model non-human animal of the present invention, or a tissue, organ, or animal derived from the animal. It is characterized by contacting cells with a test substance. Preferably, a method of administering a test substance to a model animal is preferred.
[0047] 本発明の機能性食品のスクリーニング方法は、上記に例示の血管障害性疾患の予 防及び Z又は治療薬のスクリーニング方法に準じて行うことができる。機能性食品の 評価方法 (スクリーニング方法における評価を含む)は、上述する薬剤のスクリーニン グ方法(1, 2)における評価より判断基準を低く設定することができる。より詳細には、 機能性食品の評価方法は、例えば、前記薬剤のスクリーニング方法 1において、野 生型動物の平均血圧値が約 135%以下(マウスの血圧の場合には約 135mmHg以下 )程度である被検物質を、機能性食品として有用であると評価できる。上記方法によ れば、高血圧の緩和や血管障害性疾病のリスクを低減可能な機能性食品を簡便に スクリーニング (選別)、評価することができる。 [0047] The screening method for functional foods of the present invention can be carried out in accordance with the above-described prevention method for vascular disorder diseases and screening method for Z or therapeutic agent. The evaluation method for functional food (including the evaluation in the screening method) can set a lower judgment criterion than the evaluation in the above-mentioned drug screening method (1, 2). More specifically, in the method for evaluating functional food, for example, in the aforementioned screening method 1 for drugs, the average blood pressure value of wild-type animals is about 135% or less (about 135 mmHg or less in the case of mouse blood pressure). A test substance can be evaluated as useful as a functional food. According to the above method, functional food that can alleviate hypertension and reduce the risk of vascular disorder disease can be easily obtained. Can be screened and evaluated.
[0048] 本発明の機能性食品の評価方法は、例えば、特定保健用食品や栄養機能食品等 を含む保健機能食品を評価する科学的根拠としての利用可能性がある。また、上記 方法は、血圧の低下や血管弛緩作用を指標とするため、得られた機能性食品に対し 、例えば、特定保健用食品における「血圧が高めの方の食生活の改善」等の表示を 付すことにより、消費者にその機能を直接訴えることができる可能性がある。  [0048] The functional food evaluation method of the present invention can be used as a scientific basis for evaluating, for example, functional health foods including foods for specified health use and functional foods for nutrition. In addition, since the above method uses a decrease in blood pressure or a blood vessel relaxing action as an index, for example, the indication of “improvement of eating habits for people with higher blood pressure” in the food for specified health use is displayed on the obtained functional food. It may be possible to appeal the function directly to consumers.
[0049] 本発明の機能性食品は、上記スクリーニング方法又は評価方法で得ることができる 。このため、高血圧の緩和'改善を促し、高血圧に関連する疾病 (血管障害性疾患等 )のリスクを低減 ·予防する機能を発揮できる食品としてきわめて有用である。  [0049] The functional food of the present invention can be obtained by the screening method or the evaluation method described above. For this reason, it is extremely useful as a food that can promote the alleviation and improvement of hypertension and reduce or prevent the risk of diseases related to high blood pressure (eg, vascular disorder diseases).
実施例  Example
[0050] 以下に、実施例に基づいて本発明をより詳細に説明するが、本発明はこれらの実 施例により限定されるものではない。なお、「RAMP1 +/+」は野生型、「RAMP1 +/-」 は RAMP1ヘテロ接合体、「RAMP1 -/-」は RAMP1ホモ接合体をそれぞれ意味してい る。  [0050] Hereinafter, the present invention will be described in more detail based on examples, but the present invention is not limited to these examples. “RAMP1 + / +” means wild type, “RAMP1 +/-” means RAMP1 heterozygote, and “RAMP1-/-” means RAMP1 homozygote.
[0051] 実施例 1 (RAMP1ノックアウトマウスの作成)  [0051] Example 1 (creation of a RAMP1 knockout mouse)
(1) Vector作製  (1) Vector production
BAC clone : GSll- "201 (倉敷紡績株式会社)から RAMP1の exonを含むゲノムを、 Ba mHlで切り出される 9kbpのフラグメントの中に見つけた。サブクローニングにより、 RA MP1の exon2を確認した。 exon2の 200bp上流に LoxP配列を、 2kbp下流に LoxP- neom ycin而性遺伝子(pPNTより) -LoxPカセットを挿入した。 Exon2の上流 5。 7kbpに thymid ine kinase遺伝子(pPNTより)を挿入し、ターゲティングベクターとした。 BAC clone: GSll- " 2 01 (Kurashiki Spinning Co., Ltd.) found a genome containing exon of RAMP1 in a 9 kbp fragment excised with Ba mHl. By subcloning, we confirmed exon2 of RA MP1. The LoxP sequence was inserted 200 bp upstream, and the LoxP-neomycin metagene (from pPNT) -LoxP cassette was inserted 2 kbp downstream 5. Upon of Exon2 5. The thymid ine kinase gene (from pPNT) was inserted 7 bp into the targeting vector did.
pPNT: Victorし J. cell: 1991 (65)1153—63  pPNT: Victor J. cell: 1991 (65) 1153—63
[0052] (2) ES細胞作製 [0052] (2) ES cell production
Notlで線状化したターゲティングベクター 20 gを D3 ES細胞に Gene Puis er (Bio- Ra d)を用いてエレクト口ポレーシヨンでトランスフエクシヨンした(条件は 250≡V, 500≡ F)。翌日より選択培地として G418を 150 μ g/mL添カ卩し、さらに翌日より gancyclovir を 2 mol/L添カ卩した。エレクト口ポレーシヨン一週間後、コロニー化した ES細胞をピッ クアップしゲノム抽出用と凍結用に継代した。細胞よりゲノムを抽出し、合成オリゴヌク レオチド Primer456 (配列番号 1)と Primer298 (配列番号 2)とをプライマーに用い、 94 °C-30sec + (94°C-30sec 68°C- lmin) X 30cycle + 72°C- 2minの条件で PCR反応を行 うことにより、短腕側が相同組換えを起こしているクローンを選別した。短腕側の相同 組換えが確認できたクローンにつ 、て、合成オリゴヌクレオチド Primer494 (配列番号 3)と Primer495 (配列番号 4)とをプライマーに用い、 94°C-30sec + (94°C-30sec 70°C -lmin) X 30cycle + 72°C-lmin30secの条件で PCR反応を行うことにより、長腕側の Lo xPが残っていることを検出し、更にサザンブロッテイングで短腕、長腕両側での相同 組換えを最終的に確認した。 643クローンから 5クローンの相同組換えを確認し、その うち正常な核型を有していた 2クローンを RAMPl-floxed ES細胞とした。 20 g of the targeting vector linearized with Notl was transferred to D3 ES cells using Gene Puis er (Bio-Rad) with an electoral position (conditions 250≡V, 500≡F). From the following day, G418 was added as a selective medium at 150 μg / mL, and gancyclovir was added at 2 mol / L from the next day. One week after the electoral position, colonized ES cells were picked up and passaged for genome extraction and freezing. Genome is extracted from cells and synthesized oligonucleotides PCR using the primers Primer456 (SEQ ID NO: 1) and Primer298 (SEQ ID NO: 2) as primers and 94 ° C-30 sec + (94 ° C-30 sec 68 ° C- lmin) X 30cycle + 72 ° C-2 min By carrying out the reaction, clones that had homologous recombination on the short arm side were selected. For clones in which short-arm homologous recombination was confirmed, synthetic oligonucleotides Primer494 (SEQ ID NO: 3) and Primer495 (SEQ ID NO: 4) were used as primers, and 94 ° C-30sec + (94 ° C- 30sec 70 ° C -lmin) X 30cycle + 72 ° C-lmin By performing PCR reaction under the condition of 30sec, it is detected that Lo xP remains on the long arm side, and further, Southern blotting is used to detect short arm and long arm Homologous recombination on both sides was finally confirmed. Homologous recombination from 643 clones to 5 clones was confirmed, and 2 clones having normal karyotypes were designated as RAMPl-floxed ES cells.
配列番号 1 : 5,- TGTACCCTGT GCGGTTTGTC TGGGGGCGCG- 3, (Primer456) 配列番号 2 : 5,- AGGGGAGGAG TAGAAGGTGG CGCGAAGGGG-3 ' (Primer298) 配列番号 3 : 5,- GTCTTAACTG CACTGTGGGC- 3 ' (Primer494) SEQ ID NO: 1: 5,-TGTACCCTGT GCGGTTTGTC TGGGGGCGCG-3, (Primer456) SEQ ID NO: 2: 5,-AGGGGAGGAG TAGAAGGTGG CGCGAAGGGG-3 '(Primer298) SEQ ID NO: 3: 5,-GTCTTAACTG CACTGTGGGC-3' (Primer494)
配列番号 4 : 5,- GTGATAAAAG CAACTCCGGG- 3 ' (Primer495) SEQ ID NO: 4: 5,-GTGATAAAAG CAACTCCGGG-3 '(Primer495)
(3) Cre/LoxPシステムの確認 (3) Check Cre / LoxP system
得られた RAMPl- floxed ES細胞に Notlで線状化した pCAGGS- Cre 10 gを Gene Pu lser(Bio-Rad)を用いてエレクト口ポレーシヨンでトランスフエクシヨンした(条件は 250 ≡V and 500≡ μ F)。翌日より選択培地として G418 (150 μ g/mL)を添カ卩した。エレク トロポレーシヨンより一週間後、コロニー化した ES細胞をピックアップしゲノム抽出用と 凍結用に継代した。抽出したゲノムに対し、合成オリゴヌクレオチド Primer258 (配列 番号 5)と Primer259 (配列番号 6)とをプライマーに用い、 94°C-30sec + (94°C-30sec 60°C-30sec 72°C-30sec) X 40cycle + 72°C- 2minの条件で PCR反応を行うことにより 、 Cre recombinaseのインテグレーションを確認した。さらに、 Creが導入されたクロー ンに対し、合成オリゴヌクレオチド Primer494 (配列番号 7)と Primer496 (配列番号 8)と をプライマーに用い、 94°C-30sec + (94°C- 30sec 65°C- 30sec 74°C-30sec) X 40cycle + 72°C-2minの条件で PCR反応を行った。野生型のゲノムだと 4kbp以上離れている ので PCRは走らな!/ヽが、 Creにより recombinasionが起こって!/、ると 500bpのバンドが確 認できる。これにより 2つのクローン双方において Cre recombinaseの導入が確認され た全てのクローンでゲノムの recombinasionを確認した。 The resulting RAMPl-floxed ES cells were transfected with 10 g of pCAGGS-Cre linearized with Notl using an erect mouth position using Gene Puller (Bio-Rad) (conditions were 250 ≡ V and 500 ≡ μ F). From the next day, G418 (150 μg / mL) was added as a selective medium. One week after electroporation, colonized ES cells were picked up and passaged for genome extraction and freezing. Using the extracted oligonucleotides Primer258 (SEQ ID NO: 5) and Primer259 (SEQ ID NO: 6) as primers, 94 ° C-30sec + (94 ° C-30sec 60 ° C-30sec 72 ° C-30sec ) Cre recombinase integration was confirmed by PCR reaction under conditions of X 40cycle + 72 ° C-2min. In addition, the clones with Cre were introduced using primers Primer494 (SEQ ID NO: 7) and Primer496 (SEQ ID NO: 8) as primers, and 94 ° C-30sec + (94 ° C-30sec 65 ° C- PCR was performed under the conditions of 30 sec 74 ° C-30 sec) X 40cycle + 72 ° C-2 min. Since the wild-type genome is more than 4kbp away, PCR does not run! / ヽ, but when Creombi has recombinasion! /, A 500bp band can be confirmed. This confirmed the introduction of Cre recombinase in both clones. Genomic recombinasion was confirmed in all clones.
配列番号 5 : 5,- GTTTCACTGG TTATGCGGCG G- 3, (Primer258)  SEQ ID NO: 5: 5,-GTTTCACTGG TTATGCGGCG G-3, (Primer258)
配列番号 6 : 5,- TTCCAGGGCG CGAGTTGATA G- 3, (Primer259)  SEQ ID NO: 6: 5, TTCCAGGGCG CGAGTTGATA G-3, (Primer259)
配列番号 7 : 5,- GTCTTAACTG CACTGTGGGC- 3 ' (Primer494)  SEQ ID NO: 7: 5,-GTCTTAACTG CACTGTGGGC-3 '(Primer494)
配列番号 8 : 5,- TTGGAACATC ACCACGCACG- 3, (Primer496)  SEQ ID NO: 8: 5,-TTGGAACATC ACCACGCACG-3, (Primer496)
[0054] (4)キメラマウス作製、 germ line transmission  [0054] (4) Chimera mouse production, germ line transmission
C57BL/6マウス(日本 SLC)より採取した未受精卵の胚盤胞へ 4〜8個の RAMP1- flo xed ES細胞を注入し、偽妊娠 ICRマウスへ戻した。 3週間後に得られた産子の毛色で キメラマウスであることを確認した。さらにキメラマウスと野生型の C57BL/6マウスと交 配させ、その産子の中に ES細胞由来の RAMP1 +/-マウス力 ^、ることを毛色およびジ エノタイピングより解析し、 2つのクローンに germ line transmissionを確認した。ジエノ タイピングは尾部より抽出したゲノムを前記と同様に Primer456と Primer298とを用いた PCR反応(合成オリゴヌクレオチド Primer456 (配列番号 1)と Primer298 (配列番号 2) とをプライマーに用いた 94°C- 30sec + (94°C- 30sec 68°C- lmin) X 30cycle + 72°C-2m inの条件での PCR反応)及びサザンブロッテイングにより行った。  Four to eight RAMP1-floxed ES cells were injected into blastocysts of unfertilized eggs collected from C57BL / 6 mice (Japan SLC) and returned to pseudopregnant ICR mice. The color of the litter obtained after 3 weeks was confirmed to be a chimeric mouse. Furthermore, it was crossed with chimeric mice and wild-type C57BL / 6 mice, and it was analyzed by hair color and dienotyping that the ESP-derived RAMP1 +/- mouse power ^, and the two clones were analyzed. Confirmed germ line transmission. In the dienotyping, the genome extracted from the tail was subjected to PCR reaction using Primer456 and Primer298 as described above (94 ° C-30 sec using primers of synthetic oligonucleotide Primer456 (SEQ ID NO: 1) and Primer298 (SEQ ID NO: 2)). + (94 ° C-30 sec, 68 ° C-lmin) X PCR at 30 cycles + 72 ° C-2 min) and Southern blotting.
[0055] (5)ホモ接合体とその表現形  [0055] (5) Homozygote and its phenotype
得られた RAMP1 +/-マウス同士を交配させて RAMP1 -/-マウス、及びコントロール として同腹の RAMP1 +/+マウス、 RAMP1 +/-マウスを得た。ジエノタイピングは、尾部 より抽出したゲノムに対し、前記(3)と同様の条件下で PCR反応により行った。  The obtained RAMP1 +/− mice were mated to obtain RAMP1 − / − mice, and the same lump RAMP1 + / + mice and RAMP1 +/− mice as controls. Dienotyping was performed on the genome extracted from the tail by a PCR reaction under the same conditions as in (3) above.
8週齢の RAMP1 +/+、 RAMP1 +/-、 RAMP1 -/-の各マウスについて、脳、胸腺、脾臓 、肺の各臓器における RAMP1と CRLRの mRNAレベルでの発現を確認した。その結 果を図 1に示す。  For 8-week-old RAMP1 + / +, RAMP1 +/−, and RAMP1 − / − mice, the expression of RAMP1 and CRLR at the mRNA level in the brain, thymus, spleen, and lung organs was confirmed. The results are shown in Fig. 1.
[0056] 実施例 2 (恒常的 RAMP1 KOマウス作製)  Example 2 (Constant RAMP1 KO mouse production)
全身で Cre recombinaseを発現する pCAGGS- Cre transgenicマウスと RAMPl-floxed マウスを交配させることで、 pCAGGS- Cre陽性かつ RAMP1- floxedマウスが産子として 得られた。そのマウスと wild typeマウスとの交配により RAMP1ヘテロ欠損マウスが生 まれた。得られたヘテロ欠損マウス同士を交配させて RAMP1ホモ欠損マウスが生ま れた。ジエノタイピングは、尾部より抽出したゲノムを铸型にし、合成オリゴヌクレオチ ド Primer494 (配列番号 7)と Primer496 (配列番号 8)とをプライマーに用いた、 94°C-3 Osec + (94°C-30sec 65°C- 30sec 74°C-30sec) X 40cycle + 72°C- 2minの条件での PC R反応、及び合成オリゴヌクレオチド Primer494 (配列番号 3)と Primer495 (配列番号 4 )とをプライマーに用いた 94°C- 30sec + (94°C- 30sec 70°C- lmin) X 30cycle + 72°C- 1 min30secの条件での PCR反応との組み合わせで、ヘテロ欠損マウスかホモ欠損マウ スかを判断した。 By mating pCAGGS- Cre transgenic mice and RAMPl-floxed mice that express Cre recombinase systemically, pCAGGS- Cre positive and RAMP1- floxed mice were obtained as offspring. By mating the mouse with a wild type mouse, a RAMP1 hetero-deficient mouse was born. The heterozygous mice obtained were mated to produce RAMP1-homozygous mice. In dienotyping, the genome extracted from the tail is made into a saddle shape, and synthetic oligonucleotides are used. 94 ° C-3 Osec + (94 ° C-30sec 65 ° C-30sec 74 ° C-30sec) X 40cycle + 72 ° using Primer494 (SEQ ID NO: 7) and Primer496 (SEQ ID NO: 8) as primers 94 ° C-30 sec + (94 ° C-30 sec 70 ° C- using the PCR reaction under the condition of C-2 min, and the synthetic oligonucleotides Primer494 (SEQ ID NO: 3) and Primer495 (SEQ ID NO: 4) as primers lmin) X 30cycle + 72 ° C-1 min 30 sec was used in combination with the PCR reaction to determine whether it was a hetero-deficient mouse or a homo-deficient mouse.
[0057] 実施例 3 [0057] Example 3
実施例 1で得た RAMP1 -/-マウスに、被検物質を生理的食塩水に溶解して調製し た 15mg/0.5mlの注射液を静脈注射し、経時的に後述する方法に従って平均血圧を 測定する。また、対照として、生後 6週目の RAMP1 +/+マウスと RAMP1 - /-マウスに 0. 5mlの生理的食塩水を注射して経時的に同様に血圧を測定する。その結果、被検物 質を注射された RAMP1 -/-マウスの血圧が 120mmHg以下であった場合に、その被 検物質は血管障害性疾患の予防及び Z又は治療薬として有用である。  The RAMP1 − / − mice obtained in Example 1 were intravenously injected with a 15 mg / 0.5 ml injection prepared by dissolving the test substance in physiological saline, and the mean blood pressure was measured over time according to the method described later. taking measurement. As a control, RAMP1 + / + mice and RAMP1 − / − mice at 6 weeks of age are injected with 0.5 ml of physiological saline, and blood pressure is similarly measured over time. As a result, when the blood pressure of RAMP1 − / − mice injected with the test substance is 120 mmHg or less, the test substance is useful as a prophylactic and Z or therapeutic agent for vascular disorder diseases.
[0058] 実施例 4 [0058] Example 4
被検物質を生理的食塩水に溶解して調製した 15mg/0.5mlの注射液を静脈注射し 、経時的に後述する方法に従って収縮期血圧を測定する。また、対照として、生後 6 週目の RAMP1 +/+マウスと RAMP1 -/-マウスに 0.5mlの生理的食塩水を注射して経 時的に同様に血圧を測定する。その結果、生理的食塩水が投与された RAMP1-/-マ ウスの血圧が 140mmHgであるのに対し、被検物質を注射された RAMP1 -/-マウスの 血圧が 120mmHg以下であった場合、その被検物質は血管障害性疾患の予防及び Z又は治療薬として有用である。  A 15 mg / 0.5 ml injection prepared by dissolving the test substance in physiological saline is intravenously injected, and the systolic blood pressure is measured over time according to the method described later. As a control, RAMP1 + / + and RAMP1-/-mice at 6 weeks of age are injected with 0.5 ml of physiological saline and blood pressure is similarly measured over time. As a result, if the blood pressure of RAMP1-/-mouse administered with physiological saline is 140 mmHg, while the blood pressure of RAMP1-/-mice injected with the test substance is 120 mmHg or less, The test substance is useful as a preventive and Z or therapeutic agent for vascular disorder diseases.
[0059] 実施例 5 [0059] Example 5
特表 2005— 503809号公報の記載に基づき、 RAMP1のアミノ酸配列 74番のリジ ンをトリブトファンに変換することにより RAMP1の機能が亢進された RAMP1変異タンパ ク質を調製し、生理的食塩水に溶解して 15mg/0.5mlの注射液として用いて実施例 1 で得た RAMP1 -/-マウスに静脈注射し、経時的に後述する方法に従って収縮期血 圧を測定する。その結果、注射後の RAMP1 - /-マウスの血圧が 120mmHg以下を示 すことから、上記 RAMP1変異タンパク質は、血流改善薬として、また、血管障害性疾 患の予防及び z又は治療薬として有用である。 Based on the description in JP 2005-503809, a RAMP1 mutant protein with enhanced RAMP1 function was prepared by converting the 74th amino acid sequence of RAMP1 into tributophan, and dissolved in physiological saline. Then, it is intravenously injected into the RAMP1 − / − mouse obtained in Example 1 using it as an injection solution of 15 mg / 0.5 ml, and the systolic blood pressure is measured over time according to the method described later. As a result, the blood pressure of RAMP1 − / − mice after injection showed 120 mmHg or less. Therefore, the RAMP1 mutein was used as a blood flow improving agent and as a vascular disorder disease. It is useful as a preventive and z or therapeutic agent for patients.
[0060] 実施例 6  [0060] Example 6
マウス用の固形状の餌を粉砕し、実施例 6で調製した RAMP1変異タンパク質を 15m g/g濃度で混合して再度固形状に成形したものを被検物質として用い、平均血圧が 1 40mmHgの高血圧モデルマウスに、通常の餌の代わりに 1週間摂取させた後、後述す る方法に従って収縮期血圧を測定する。その結果、被検物質を摂取させた RAMP1- /-マウスの血圧が 120mmHg以下に低下することから、その被検物質は、血管障害性 疾患の予防に有用な機能性食品として有用である。  The solid food for mice was crushed, and the RAMP1 mutant protein prepared in Example 6 was mixed at a concentration of 15 mg / g and molded again into a solid form as a test substance, with an average blood pressure of 140 mmHg. After taking hypertension model mice for 1 week instead of normal food, measure systolic blood pressure according to the method described below. As a result, the blood pressure of RAMP1-/-mice fed with the test substance decreases to 120 mmHg or less, so that the test substance is useful as a functional food useful for the prevention of vascular disorder diseases.
[0061] (評価方法) [0061] (Evaluation method)
心拍数及び収縮期血圧の測定  Measurement of heart rate and systolic blood pressure
実施例 1で作製した RAMP1 +/+、 RAMP1 +/-、 RAMP1 -/-の各マウスについて、 心拍数と収縮期血圧を以下の方法で測定した。  For the RAMP1 + / +, RAMP1 +/−, and RAMP1 − / − mice prepared in Example 1, the heart rate and systolic blood pressure were measured by the following methods.
無麻酔条件下でマウスを保定し、 MK2000 (室町機械社製)を用いて tan- cuff?去によ り心拍数および収縮期血圧を測定した。なお測定時には環境に慣らすため、各個体 にっき 30回以上の測定を行った後、本測定として 10回の測定を行い、その平均値を その個体の値とした。その結果を図 3に示す。図 3に示されるように、野生型マウスな らびにヘテロ接合体の平均血圧は約 lOOmmHgであるのに対し、上記で得た RAMP1 KOマウスの平均血圧は約 140mmHgであり、明かな高血圧を示した。また、同様の方 法により、 6〜18週齢のマウスについて心拍数と血圧の変化を測定した。その結果を 図 4に示す。図 4に示されるように、心拍数はいずれのマウスにおいても変化は認め られなかった。 RAMP1 KOマウスの血圧は調べた 6週齢において既に高ぐ週齢とと もに徐々に高くなる傾向が認められた。  Mice were held under anesthesia, and heart rate and systolic blood pressure were measured by tan- cuff removal using MK2000 (Muromachi Kikai Co., Ltd.). In order to acclimatize to the environment at the time of measurement, each individual was measured 30 times or more, then 10 measurements were performed as the main measurement, and the average value was taken as the value of that individual. The results are shown in Fig. 3. As shown in Figure 3, the mean blood pressure of wild-type mice and heterozygotes is about lOOmmHg, while the mean blood pressure of RAMP1 KO mice obtained above is about 140 mmHg, indicating clear hypertension. It was. In the same manner, changes in heart rate and blood pressure were measured for 6-18 week old mice. The results are shown in Fig. 4. As shown in Figure 4, heart rate did not change in any mouse. The blood pressure of the RAMP1 KO mice tended to gradually increase with the age of 6 weeks, which was already high.
[0062] 血管収縮実験 [0062] Vasoconstriction experiment
実施例 1で作製した RAMP1 +/+、 RAMP1 +/-、 RAMP1 -/-の各マウスについて、 以下の方法で作成した血管標本における張力変化を測定し、血管収縮率に換算す ることにより血管収縮特性を評価した。  For each of the RAMP1 + / +, RAMP1 +/-, and RAMP1-/-mice prepared in Example 1, the change in tension in the blood vessel specimen prepared by the following method was measured, and converted into a vasoconstriction rate. Shrinkage properties were evaluated.
ペントバルビタールナトリウム麻酔下、マウス頸動脈を切断し、脱血した。次に胸部 大動脈を摘出し、 37°Cの Klebs- Henseleit液(118.4 mM NaCl、 4.7 mM KC1、 2.5 mM CaCl、 1.2 mM KH PO、 1.2 mM MgSO - 7H 0、 25 mM NaHCO、11.1 mMダルコ一Under pentobarbital sodium anesthesia, the mouse carotid artery was cut and blood was removed. Next, the thoracic aorta was removed and 37 ° C Klebs-Henseleit solution (118.4 mM NaCl, 4.7 mM KC1, 2.5 mM) CaCl, 1.2 mM KH PO, 1.2 mM MgSO-7H 0, 25 mM NaHCO, 11.1 mM Darco
2 2 4 4 2 3 2 2 4 4 2 3
ス)中で血管の結合組織を除去後、 3 mm幅に血管を切断しリング標本を作製した。 血管標本にアクリル榭脂製支持棒を取り付けたタングステン線を挿入し、 95% 0  After removing the connective tissue of the blood vessel in 3), the blood vessel was cut to a width of 3 mm to prepare a ring specimen. Insert a tungsten wire with a support rod made of acrylic resin into the blood vessel specimen, 95% 0
2 I 5% 2 I 5%
CO混合ガスを通気して 37°Cに保温した Klebs-Henseleit液 10 mL中に固定した。タンThe mixture was fixed in 10 mL of Klebs-Henseleit solution kept at 37 ° C by ventilating CO gas mixture. Tan
2 2
ダステン線をもう一本標本に挿入し、 FDピックアップに連結した。なお、標本の張力 変化は FDピックアップとキヤリヤーアンプを介して等尺性にレコーダー(model 8K21; NEC)上に記録し(1.0 g = 5 cm)、簡易マ-ュピレーター(model t- 7; NEC)を操作し て標本に 0.7 gの基礎張力をかけた。 30分後、フエ-レフリン (終濃度 3 X 10— 7 M)によ る血管収縮反応と、アセチルコリン (終濃度 10— 7 M)による血管弛緩反応を確認した。 血管標本の確認終了後、再び Klebs-Henseleit液中で 60分間平衡化し、フ 二レフリ ン(10— 6 M)添カ卩により血管を収縮させ、 a CGRP、 j8 CGRPまたはアドレノメジユリン (A M)を終濃度 lx 10— u-3xl0— 6 Mとなるように累積添加し血管の弛緩応答を記録した。 再び 30分間平衡ィ匕させた後、フエ-レフリン(10— 6 M)で収縮させた後,パパべリン(10 — 4 M)を添加し血管を弛緩させ、最大弛緩能を求めた。これらの結果を図 5に示す。 Another dust wire was inserted into the specimen and connected to the FD pickup. The change in tension of the specimen is recorded on the recorder (model 8K21; NEC) isometrically via an FD pickup and carrier amplifier (1.0 g = 5 cm), and a simple manipulator (model t-7; NEC). ) To apply 0.7 g basic tension. After 30 minutes, Hue - Refurin and vasoconstriction that by the (final concentration of 3 X 10- 7 M), confirmed the blood vessel relaxation reaction by acetylcholine (final concentration of 10- 7 M). After confirmation of the blood vessel specimens finished, again equilibrated for 60 min at Klebs-Henseleit solution, the vessel is contracted by off two Refuri emissions (10- 6 M)添Ka卩, a CGRP, j8 CGRP or address drink Jiyu phosphorus (AM ) was recorded relaxation response of the cumulative added vessels to a final concentration of lx 10- u -3xl0- 6 M. After equilibration spoon again 30 minutes, Hue - after contracted with Refurin (10- 6 M), Papabe phosphorus (10 - 4 M) added to relax blood vessels to determine the maximum relaxation ability. These results are shown in FIG.
[0063] 図 5に示されるように、フ -レフリンにより収縮させた RAMP1+/+マウス胸部大動脈 は、 a CGRP添カ卩により顕著に弛緩した。一方、 RAMP1- /-マウスの胸部大動脈は α CGRPによる弛緩反応を示さなかった。このことは RAMP1が a CGRPの受容体を構成 することを示す。一方、 i8 CGRPは野生型マウス胸部大動脈に対しても顕著な血管弛 緩活性を認めな力 た。また、強い血管拡張作用を有することが知られている AMの 活性も、フエ-レフリンにより収縮させた RAMP1- /-マウスの胸部大動脈では RAMP1+ /+マウスに比べあきらかに減弱した。このことはアドレノメデュリンの血管拡張作用も 少なくとも一部は CGRPと同様 RAMP1で構成される受容体が関与していることが示さ れた。 [0063] As shown in FIG. 5, the RAMP1 + / + mouse thoracic aorta contracted with furephrine was remarkably relaxed by a CGRP supplementation. On the other hand, the thoracic aorta of RAMP1-/-mice showed no relaxation reaction by α-CGRP. This indicates that RAMP1 constitutes a CGRP receptor. On the other hand, i8 CGRP did not show significant vasorelaxing activity in wild-type mouse thoracic aorta. In addition, the activity of AM, which is known to have a strong vasodilatory effect, was clearly attenuated in the thoracic aorta of RAMP1 − / − mice contracted by fe-rephrin compared to RAMP1 + / + mice. This indicates that the vasodilatory effect of adrenomedullin is involved, at least in part, in the receptor composed of RAMP1 as in CGRP.
[0064] 免疫組織化学染色  [0064] Immunohistochemical staining
ヒト病理組織を 10%中性緩衝ホルマリンにより固定し、ノラフィン包埋後、 4 /z mに切 片化した。脱パラフィンィ匕後、切片を室温にて 1時間、抗 RAMP1抗体 (SantaCruz)とィ ンキュペートした。反応に引き続き Histofine SAB-PO kit (-チレイ社製)及び色素と して diaminobenzidineを用いて可視化し、へマトキシリンにて対比染色した。その結果 、脳、大腸粘膜細胞等とともに大腸癌細胞においても RAMP1の発現が認められた。 このことは CGRPのシグナルが血管拡張作用とともに癌細胞の増殖 ·転移にも関与す ることを示すものである。 Human pathological tissue was fixed with 10% neutral buffered formalin, embedded in norafine, and cut into 4 / zm. After deparaffinization, the sections were incubated with anti-RAMP1 antibody (SantaCruz) for 1 hour at room temperature. Subsequent to the reaction, Histofine SAB-PO kit (manufactured by Chirei Co., Ltd.) and diaminobenzidine as a dye were visualized and counterstained with hematoxylin. as a result In addition, expression of RAMP1 was observed in colon cancer cells as well as brain and colon mucosa cells. This indicates that CGRP signals are involved in the growth and metastasis of cancer cells as well as vasodilatory effects.

Claims

請求の範囲 The scope of the claims
[1] 染色体上の RAMP1遺伝子の一部又は全部の改変により、当該 RAMP1遺伝子の機 能が完全に欠損していることを特徴とする高血圧モデル非ヒト動物。  [1] A hypertensive model non-human animal, wherein the function of the RAMP1 gene is completely deficient due to partial or complete modification of the RAMP1 gene on the chromosome.
[2] 染色体上に選択マーカー遺伝子が挿入されていない請求の範囲第 1項記載の高 血圧モデル非ヒト動物。  [2] The hypertension model non-human animal according to claim 1, wherein a selection marker gene is not inserted on the chromosome.
[3] 染色体上に、 RAMP1遺伝子のェキソンの少なくとも一部が LoxP配列に挟まれた領 域と、選択マーカー遺伝子が LoxP配列に挟まれた領域とを含む ES細胞に、 Cre酵 素を作用させて得られる ES細胞を用いる方法、又は染色体上に、 RAMP1遺伝子の ェキソンの少なくとも一部が LoxP配列に挟まれた領域を含む ES細胞を用いて作製さ れた RAMPl-floxedマウスと、 Cre酵素を発現可能なマウスとの交配による方法の!/ヽず れかで作製された請求の範囲第 1項又は第 2項記載の高血圧モデル非ヒト動物。  [3] Cre enzyme is allowed to act on ES cells containing a region where at least part of the exon of the RAMP1 gene is sandwiched between LoxP sequences and a region where a selectable marker gene is sandwiched between LoxP sequences on the chromosome. A method using ES cells obtained by the above, or a RAMPl-floxed mouse prepared using ES cells containing a region in which at least part of the exon of the RAMP1 gene is sandwiched between LoxP sequences on the chromosome, and Cre enzyme The hypertensive model non-human animal according to claim 1 or 2, which is prepared by crossing the method with an expressible mouse!
[4] RAMP1遺伝子の機能がコンデイショナルに欠損されている請求の範囲第 1項〜第 [4] Claims 1 to 4 in which the function of the RAMP1 gene is conditionally defective
3項の何れかの項に記載の高血圧モデル非ヒト動物。 4. The hypertensive model non-human animal according to any one of items 3.
[5] 非ヒト動物がラット又はマウスである請求の範囲第 1項〜第 4項の何れかの項に記 載の高血圧モデル非ヒト動物。  [5] The hypertensive model non-human animal according to any one of claims 1 to 4, wherein the non-human animal is a rat or a mouse.
[6] 請求の範囲第 1項〜第 5項の何れかの項に記載の高血圧モデル非ヒト動物に被検 物質を投与するか、又は該動物由来の組織、器官、若しくは細胞を被検物質と接触 させることを特徴とする血管障害性疾患の予防及び Z又は治療用薬剤のスクリー- ング方法。  [6] The test substance is administered to the hypertensive model non-human animal according to any one of claims 1 to 5, or a tissue, organ or cell derived from the animal is used as the test substance. A method for screening a drug for prevention and Z or treatment of a vascular disorder disease, which comprises contacting with a blood vessel.
[7] 血圧及び Z又は血管拡張作用を指標とする請求の範囲第 6項記載の薬剤のスクリ 一ユング方法。  7. The drug screening method according to claim 6, wherein blood pressure and Z or vasodilatory action are used as indices.
[8] 血管障害性疾患が、高血圧、血管内皮障害、動脈硬化性疾患、及び記憶障害,認 知症,末梢血流障害性疾患を含む血流障害性疾患からなる群から選択される少なく とも一の疾患である請求の範囲第 6項又は第 7項の何れかの項に記載の薬剤のスク リーニング方法。  [8] The vascular disorder is at least selected from the group consisting of hypertension, vascular endothelial disorder, arteriosclerotic disorder, and blood flow disorder including memory disorder, dementia and peripheral blood flow disorder. The method for screening a drug according to any one of claims 6 and 7, which is a disease.
[9] 請求の範囲第 6項〜第 8項の何れかの項に記載の薬剤のスクリーニング方法により 得られる血管障害性疾患の予防及び Z又は治療薬。  [9] A preventive and Z or therapeutic agent for a vascular disorder disease obtained by the method for screening a drug according to any one of claims 6 to 8.
[10] RAMP1遺伝子の発現又は機能を亢進する物質を有効成分として含有する血流改 善薬。 [10] Blood flow modification containing a substance that enhances the expression or function of the RAMP1 gene as an active ingredient Good medicine.
[11] RAMP1遺伝子の発現又は機能を亢進する物質を有効成分として含有する血管障 害性疾患の予防及び Z又は治療薬。  [11] A prophylactic and Z or therapeutic agent for a vascular disorder disease comprising a substance that enhances the expression or function of the RAMP1 gene as an active ingredient.
[12] 血管障害性疾患が、高血圧、血管内皮障害、動脈硬化性疾患、及び記憶障害,認 知症,末梢血流障害性疾患を含む血流障害性疾患からなる群から選択される少なく とも一の疾患である請求の範囲第 11項記載の血管障害性疾患の予防及び Z又は 治療薬。 [12] The vascular disorder is at least selected from the group consisting of hypertension, vascular endothelial disorder, arteriosclerotic disorder, and blood flow disorder including memory disorder, dementia and peripheral blood flow disorder. 12. The prophylactic and Z or therapeutic agent for vascular disorder according to claim 11, which is a disease.
[13] 請求の範囲第 1項〜第 5項の何れかの項に記載の高血圧モデル非ヒト動物に被検 物質を投与するか、又は該動物由来の組織、器官、若しくは細胞を被検物質と接触 させることを特徴とする機能性食品のスクリーニング方法。  [13] The test substance is administered to the hypertensive model non-human animal according to any one of claims 1 to 5, or a tissue, organ, or cell derived from the animal is used as the test substance. A method for screening functional foods, characterized in that the functional foods are brought into contact with each other.
[14] 血圧及び Z又は血管拡張作用を指標とする請求の範囲第 13項記載の機能性食 品のスクリーニング方法。 [14] The method for screening a functional food according to claim 13, wherein blood pressure and Z or vasodilatory action are used as indices.
[15] 請求の範囲第 13項又は第 14項記載の機能性食品のスクリーニング方法により得ら れる機能性食品。 [15] A functional food obtained by the method for screening a functional food according to claim 13 or 14.
[16] 請求の範囲第 1項〜第 5項の何れかの項に記載の高血圧モデル非ヒト動物に被検 物質を投与するか、又は該動物由来の組織、器官、若しくは細胞を被検物質と接触 させることを特徴とする機能性食品の評価方法。  [16] The test substance is administered to the hypertensive model non-human animal according to any one of claims 1 to 5, or a tissue, organ or cell derived from the animal is used as the test substance. A method for evaluating functional foods, characterized in that it is brought into contact with a food product.
[17] 血圧及び Z又は血管拡張作用を指標とする請求の範囲第 16項記載の機能性食 品の評価方法。  [17] The method for evaluating a functional food according to claim 16, wherein blood pressure and Z or vasodilatory action are used as indices.
PCT/JP2007/059316 2006-06-02 2007-05-01 Nonhuman model animal of hypertension WO2007141974A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2006154336A JP2009201355A (en) 2006-06-02 2006-06-02 Nonhuman model animal of hypertension
JP2006-154336 2006-06-02

Publications (1)

Publication Number Publication Date
WO2007141974A1 true WO2007141974A1 (en) 2007-12-13

Family

ID=38801239

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/059316 WO2007141974A1 (en) 2006-06-02 2007-05-01 Nonhuman model animal of hypertension

Country Status (2)

Country Link
JP (1) JP2009201355A (en)
WO (1) WO2007141974A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102511447A (en) * 2012-01-04 2012-06-27 上海中医药大学附属岳阳中西医结合医院 Blood stasis-YANG hyperactivity-Phlegm hypertensive animal model and preparation method thereof
JP2013083504A (en) * 2011-10-07 2013-05-09 Pola Chem Ind Inc Screening method
WO2021201100A1 (en) * 2020-03-31 2021-10-07 学校法人 慶應義塾 Therapeutic agent using genome-edited pluripotent stem cell

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103416346A (en) * 2012-05-22 2013-12-04 浙江中医药大学 Production method of animal model for tension-type hypertension caused by multi-factor stimulation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2005503809A (en) * 2001-09-27 2005-02-10 メルク エンド カムパニー インコーポレーテッド Isolated DNA molecule encoding a humanized calcitonin gene-related peptide receptor, related non-human transgenic animals and assay methods
JP3639555B2 (en) * 2000-11-30 2005-04-20 ファイザー・プロダクツ・インク RAMP activity change method

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3639555B2 (en) * 2000-11-30 2005-04-20 ファイザー・プロダクツ・インク RAMP activity change method
JP2005503809A (en) * 2001-09-27 2005-02-10 メルク エンド カムパニー インコーポレーテッド Isolated DNA molecule encoding a humanized calcitonin gene-related peptide receptor, related non-human transgenic animals and assay methods

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
FUNAYA K. ET AL.: "CGRP Juyotai Knockout Mouse wa, Atarashii Koketsuatsu Model ni Nariuruka?", THE JOURNAL OF THE JAPANESE SOCIETY OF INTERNAL MEDICINE, vol. 96, pages 120 + ABSTR. NO. 68, XP003019754 *
KATO T. ET AL.: "Hifu Ensho ni okeru Shinkei Peptide CGRP no Yakuwari", THE PHARMACEUTICAL SOCIETY OF JAPAN NENKAI YOSHISHU, vol. 127, no. 2, 5 March 2007 (2007-03-05), pages 29 + ABSTR. NO. 30S-PM08, XP003019753 *
MORENO M.J. ET AL.: "Characterization of calcitonin gene-related peptide (CGRP) receptors and their receptor-activity-modifying proteins (RAMPs) in human brain microvascular and astroglial cells in culture", NEUROPHARMACOLOGY, vol. 42, 2002, pages 270 - 280, XP003019755 *
OGITANI Y. ET AL.: "Shinkei Peptide CGRP ni yoru Shizen Men'eki.Kakutoku Men'eki Seigyo Kiko Kaiseki", THE PHARMACEUTICAL SOCIETY JAPAN NENKAI YOSHISHU, vol. 127, no. 2, 5 March 2007 (2007-03-05), pages 29 + ABSTR. NO. 30S-PM04, XP003019752 *
QING X. ET AL.: "Targeted blocking of gene expression for CGRP receptors elevates pulmonary artery pressure in hypoxic rats", AM. J. PHYSIOL. LUNG CELL MOL. PHYSIOL., vol. 285, 2003, pages L86 - L96, XP003019750 *
TSUJIKAWA K. ET AL.: "RAMP/CRLR Juyotai o Kaishita Calcitonin Idenshi Kanren Peptide (CGR) ni yoru Jujo Saibo, Macrophage no Cytokine Sansei to LPS Yuhatsu Chishi Seigyo", THE JAPANESE SOCIETY FOR IMMUNOLOGY GAKUJUTSU SHUKAI KIROKU, vol. 34, 2004, pages 155 + ABSTR. NO. 2-B-W20-11-O/P, XP003019751 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013083504A (en) * 2011-10-07 2013-05-09 Pola Chem Ind Inc Screening method
CN102511447A (en) * 2012-01-04 2012-06-27 上海中医药大学附属岳阳中西医结合医院 Blood stasis-YANG hyperactivity-Phlegm hypertensive animal model and preparation method thereof
CN102511447B (en) * 2012-01-04 2013-08-14 上海中医药大学附属岳阳中西医结合医院 Blood stasis-YANG hyperactivity-Phlegm hypertensive animal model and preparation method thereof
WO2021201100A1 (en) * 2020-03-31 2021-10-07 学校法人 慶應義塾 Therapeutic agent using genome-edited pluripotent stem cell

Also Published As

Publication number Publication date
JP2009201355A (en) 2009-09-10

Similar Documents

Publication Publication Date Title
Hernández-Hoyos et al. Lck activity controls CD4/CD8 T cell lineage commitment
CN112638154B (en) Non-human animal model of DITRA disease and uses thereof
JP2021169461A (en) Animal models of longevity and related methods for increasing longevity and inhibiting tumorigenesis
Martin et al. Deficiency in IL-1 receptor type 2 aggravates K/BxN serum transfer-induced arthritis in mice but has no impact on systemic inflammatory responses
WO2007141974A1 (en) Nonhuman model animal of hypertension
WO2008075796A1 (en) Agent for promoting recovery from blood cell depletion
Yonebayashi et al. Generation of transgenic mice that conditionally overexpress Tenascin-C
US20160345553A1 (en) Transgenic mouse models for mc4r
JP4778446B2 (en) A mouse model for psoriasis and psoriatic arthritis
US20230363362A1 (en) Genetically modified non-human animal with human or chimeric glp1r
US6077990A (en) PAR2 modified transgenic mice
IL303724A (en) Non-human animals having a humanized tslp gene, a humanized tslp receptor gene, and/or a humanized il7ra gene
KR20230004472A (en) Non-human animals with a humanized CXCL13 gene
JP2008109925A (en) Adiponectin receptor gene-deficient nonhuman animal and use thereof
JP5099856B2 (en) Metabolic syndrome treatment or prevention agent, test method, test drug, and screening method for candidate compound of metabolic syndrome treatment drug
RU2804448C2 (en) Ditra models in non-human animals and their use
CA2284833A1 (en) Non human transgenic animal in which the expression of the gene coding for insulin is deleted
JP5565786B2 (en) esRAGE overexpressing mouse
US8835711B2 (en) ANF-RGC in-gene knock-out animal
WO2001009314A1 (en) Novel uses of abca-type transporters
JPH10298100A (en) Preventive and/or therapeutic agent for disease due to insulin resistance
WO2004113531A1 (en) Conditioned autophagy-deficient animal and disease model animal
CN117815363A (en) Application of Star-PAP in preparation of medicine for improving fatty insulin sensitivity
JP2009082090A (en) Erk2 knockdown non-human animal
JP2007097434A (en) Respiratory distress syndrome(rds) model mouse

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: 07742751

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 07742751

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: JP