WO2001009346A1 - Gene encoding novel adenylate kinase 3 (ak3)-like protein - Google Patents

Gene encoding novel adenylate kinase 3 (ak3)-like protein Download PDF

Info

Publication number
WO2001009346A1
WO2001009346A1 PCT/JP2000/005066 JP0005066W WO0109346A1 WO 2001009346 A1 WO2001009346 A1 WO 2001009346A1 JP 0005066 W JP0005066 W JP 0005066W WO 0109346 A1 WO0109346 A1 WO 0109346A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
dna
sequence
present
seq
Prior art date
Application number
PCT/JP2000/005066
Other languages
French (fr)
Japanese (ja)
Inventor
Research Institute Helix
Original Assignee
Ota, Toshio
Isogai, Takao
Hayashi, Koji
Saito, Kaoru
Yamamoto, Jun-Ichi
Ishii, Shizuko
Sugiyama, Tomoyasu
Wakamatsu, Ai
Nagai, Keiichi
Otsuki, Tetsuji
Ihara, Shigeo
Nakae, Hiroki
Nishikawa, Tetsuo
Kimura, Koichi
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 Ota, Toshio, Isogai, Takao, Hayashi, Koji, Saito, Kaoru, Yamamoto, Jun-Ichi, Ishii, Shizuko, Sugiyama, Tomoyasu, Wakamatsu, Ai, Nagai, Keiichi, Otsuki, Tetsuji, Ihara, Shigeo, Nakae, Hiroki, Nishikawa, Tetsuo, Kimura, Koichi filed Critical Ota, Toshio
Publication of WO2001009346A1 publication Critical patent/WO2001009346A1/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • 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
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • AK 3 adenylate kinase 3
  • the present invention relates to novel AK3-like proteins, their genes, and their production and use.
  • Adenylate kinase is a phosphotransferase that catalyzes the reaction that produces MgATP and AMP by reversible phosphorylation of one molecule of ADP by one molecule of MgADP.
  • Three isozymes, AK1, AK2, and AK3, have been reported for vertebrate adenylate kinase.
  • AK3 is a GTP-binding protein, AMP phosphotransferase, encoded in the nucleus of cells, and is present in the mitochondrial matrix of liver, heart, and muscle (Shahjahan M. et al., (1991) ) Gene 107: 313-317).
  • MELAS stroke-like symptoms or hyperlactic acidemia
  • central nervous disease central nervous disease
  • epilepsy central nervous disease
  • skeletal muscle pathology skeletal muscle pathology
  • muscular disease have been identified as mitochondrial-related diseases.
  • Electron transmission abnormalities Leber's disease, diabetes, Peason's disease, Perkinson's disease, metabolic abnormalities, etc. have been reported.
  • AK3 is expected to be involved in these diseases, and is considered to be an important target for drug development for the diagnosis, prevention and treatment of these diseases. Disclosure of the invention
  • the present invention has been made in view of such circumstances, and an object of the present invention is to provide a novel AK3-like protein, a gene thereof, and a method for producing the same and use thereof.
  • the present inventors isolated a plurality of full-length cDNAs from a cDNA library derived from retinoic acid-treated NT-2 cells by an oligocap method uniquely developed for isolating full-length cDNAs.
  • the nucleotide sequence of one of the isolated cDNAs was determined, and its structural analysis revealed significant homology to the known AK3, indicating that the cDNA encodes a novel AK3-like protein.
  • This clone was named "C-NT2RP20 00329").
  • AK3 is present in mitochondria and plays a role in converting extracellular information into intracellular information, changing cell morphology, etc., and plays a wide variety of diseases including MELAS and central nervous system disorders. The association has been suggested. Because C-NT2RP200 0329 is a novel human AK3, it is considered to be an important target for the diagnosis of these diseases and the development of drugs to prevent or treat these diseases.
  • the present invention relates to a novel AK3-like protein C-NT2RP2000329 and a DNA encoding the protein, and their production and use.
  • a method for screening a candidate compound for a drug for prevention or treatment of a disease caused by DNA abnormality according to (1) which comprises:
  • step (c) selecting a compound that increases or decreases the level of DNA expression according to (1), which is measured in step (b) and compared to the case where the test sample is not contacted, according to (1).
  • a DNA chip provided with the DNA according to (1) or (2) or the polynucleotide according to (8).
  • the present invention provides a novel protein "C-NT2RP2000329".
  • the amino acid sequence of the human C-NT2RP2000329 protein contained in the protein of the present invention is shown in SEQ ID NO: 2, and the nucleotide sequence of the cDNA encoding the protein is shown in SEQ ID NO: 1.
  • the C-NT2RP2000329 gene encodes a protein of 227 amino acids with significant homology to adenylate kinase 3 (AK3).
  • AK3 which is present in mitochondria, plays a role in converting extracellular information into cytoplasm information, changing cell morphology, apoptosis, cell movement, and intracellular substance transport.
  • the C-NT2RP2000329 protein is also considered to have such a function in vivo due to its structural characteristics, the C-NT2RP2000329 gene and protein are used for diagnosis of diseases caused by abnormalities in these functions. It is expected to be used for the development of drugs for preventing or treating the disease.
  • the C-NT2RP2000329 protein can be prepared as a recombinant protein or as a natural protein.
  • the recombinant protein is prepared, for example, by introducing a vector into which a DNA encoding the protein of the present invention has been inserted into an appropriate host cell as described below, and purifying the protein expressed in the transformant. Is possible
  • a natural protein can be prepared using, for example, an affinity column to which an antibody against the protein of the present invention described below is bound (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) ) Publish. John Wiley & Sons Section 16. 1-16. 19).
  • the antibody used for affinity purification may be a polyclonal antibody or a monoclonal antibody.
  • in vitro translation for example, “0n the fidelity of raR A translation in the nuc ⁇ ease-treated rabbit ret iculocyte lysate system. Dasso, MC, Jackson, RJ (1989) Nucleic Acids Res. 17: 3129 It is also possible to prepare the protein of the present invention by using, for example, -3144J.
  • the present invention includes a protein functionally equivalent to the human-derived C-NT2RP2000329 protein identified in this example.
  • Such proteins include, for example, mutants, homologs, and cryptants of the C-NT2RP2000329 protein described in SEQ ID NO: 2.
  • “functionally equivalent” means that the target protein has the same biological or biochemical function as the C-NT2RP2000329 protein. Examples of such a function include a function as a GTP-binding protein AMP phosphotransferase. Whether the protein functions as a GTP-binding protein AMP phosphotransferase can be determined, for example, by mixing AMP with GTP containing 32 P at the ⁇ -phosphate and confirming that ADP containing 32 P is purified Can be detected. GTP and ADP can be separated, for example, by thin-layer chromatography.
  • proteins functionally equivalent to the proteins identified in this example are For example, a method for introducing a mutation into an amino acid sequence in a protein (eg, a site-directed mutagenesis method (Current Protocols in Molecular Biology edit. Au subel et al. (1987) Publish. John Wiley & It can be prepared using Sons Section 8. 1-8.5)). Such proteins may also be generated by mutations in amino acids in nature. According to the present invention, one or more amino acids in the amino acid sequence (SEQ ID NO: 2) may be substituted, deleted, inserted, and substituted with Z or Z as long as it has a function equivalent to the protein identified in this example. Different proteins are included by addition and the like.
  • the number of amino acid mutations and mutation sites in a protein are not limited as long as the function is maintained.
  • the number of mutations is typically within 30 amino acids, preferably within 10 amino acids, and more preferably within 5 amino acids (eg, within 3 amino acids).
  • the amino acid to be substituted is preferably an amino acid having properties similar to the amino acid before substitution from the viewpoint of maintaining the function of the protein. For example, Ala, Val, Leu, Ile, Pro, Met, Phe, and Trp are considered to have similar properties because they are all classified as nonpolar amino acids.
  • examples of the non-charger include Gly, Ser, Thr, Cys, Tyr, Asn, and Gin.
  • acidic amino acids include Asp and Glu
  • basic amino acids include Lys, Arg, and His.
  • Proteins functionally equivalent to the proteins identified in this example can be obtained by using known techniques known to those skilled in the art (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section). 6.3.6.4) or use gene amplification technology (PCR) (Current protocols in Molecular Biologic edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 6.1.6.4.4) It is also possible to isolate them. That is, those skilled in the art can use a DNA encoding the protein identified in this example (SEQ ID NO: 1) or a part thereof as a probe, or an oligonucleotide specifically hybridizing with the DNA as a primer.
  • Isolating DNA that hybridizes with DNA it can. Further, based on the isolated DNA, a protein encoded by the DNA can be prepared.
  • the present invention includes proteins encoded by DNAs that hybridize with DNAs encoding these proteins, as long as they have the same function as the proteins identified in the present example. Examples of organisms for isolating functionally equivalent proteins include, but are not limited to, vertebrate animals such as humans, mice, rats, rabbits, pigs, and rabbits.
  • Stringent conditions for hybridization for isolation of DNA encoding functionally equivalent proteins are usually ⁇ lxSSC, 0.1% SDS, 37 ° C '', and are more stringent. The conditions were about 0.5 x SSC, 0.1% SDS, 42 ° C, and the more severe conditions were about 0.1 x SSC; 0.1% SDS, 65 ° C. It can be expected that DNA with higher homology to the probe sequence will be isolated as the conditions of the probe become stricter. However, the combination of the above SSC, SDS and temperature conditions is merely an example, and those skilled in the art will recognize the above or other factors that determine the stringency of the hybridization (eg, probe concentration, probe length, probe length, etc.). The same stringency as described above can be realized by appropriately combining the hybridization reaction time and the like.
  • the protein isolated using such a hybridization technique or gene amplification technique usually has higher homology in its amino acid sequence than the protein of the present invention described in SEQ ID NO: 2.
  • High homology refers to sequence identity of at least 50% or more, more preferably 70% or more, and even more preferably 90% or more (eg, 93% or more, 95% or more).
  • Amino acid sequence homology can be determined by homology search using BLAST X.
  • the present invention also provides a partial peptide of the protein of the present invention.
  • the partial peptide of the protein of the present invention can be used, for example, for preparing an antibody that binds to the protein of the present invention.
  • the partial peptide of the present invention comprises at least 7 amino acids, preferably 9 amino acids or more, more preferably 12 amino acids or more, and more preferably 15 amino acids or less. Consists of the above amino acid sequence.
  • the partial peptide of the present invention can be produced, for example, by a genetic engineering technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase.
  • the present invention also provides a DNA encoding the protein of the present invention.
  • the DNA of the present invention is not particularly limited in its form as long as it can encode the protein of the present invention, and includes genomic DNA, chemically synthesized DNA, and the like in addition to cDNA. Also, as long as the protein of the present invention can be encoded, it has an arbitrary base sequence based on the degeneracy of the genetic code! ) NA is included.
  • the DNA encoding the protein of the present invention was designed based on the hybridization method using the DNA sequence of SEQ ID NO: 1 or a part thereof as a probe. It can be isolated by a conventional method such as a gene amplification method (PCR) using primers.
  • PCR gene amplification method
  • the present invention also provides a vector into which a DNA encoding the protein of the present invention has been inserted.
  • the vector of the present invention is not particularly limited as long as it stably retains the inserted DNA.
  • a pBluescript vector manufactured by Stratagene
  • an expression vector is particularly useful. Examples of the expression vector include p BEST vector (manufactured by Promega) for in vitro expression, pET vector (manufactured by Invitrogen) for expression in E.
  • the FL3 vector (GenBank Accession No. AB009864) and the pME18S vector (Mol Cell Biol. 8: 466-472 (1988)) can be suitably used for expression in a living organism. Insertion of a DNA encoding the protein of the present invention into a vector can be carried out by a conventional method, for example, by using a restriction enzyme Sat. 7 protocols anti fi (Current protocols in Molecular Biology edit. Ausubel et al. 1987) Publish. John Wiley & Sons. Section 11.4 to 11.11).
  • the present invention also relates to a DNA encoding the protein of the present invention or the DNA inserted therein.
  • a transformant carrying the vector is provided.
  • the host cell into which the vector of the present invention is introduced is not particularly limited, and various host cells may be used depending on the purpose. Host cells can be used, for example, for the production of the proteins of the invention.
  • Production systems for protein production include in vitro and in vivo production systems. In vitro production systems include production systems using eukaryotic cells and production systems using prokaryotic cells. When eukaryotic cells are used, for example, animal cells, plant cells, and fungal cells can be used as hosts.
  • the host cells of the present invention also include cells intended for use in analyzing the function of the C-NT2RP2000329 protein and screening for its function inhibitor or function promoter utilizing the C-NT2RP2000329 protein.
  • Vector transfer into host cells can be performed, for example, by the calcium phosphate precipitation method, electric pulse mosquito L, & (Current protocols m Molecular Biology edit. Ausubel et al. (1 987) Publish. John Wiley & Sons. Section 9 1-9. 9), Lipofectamine method (manufactured by GIBCO-BRL), microinjection method, etc.
  • Preparation of the C-NT2RP2000329 protein from the transformant can be carried out by using a protein separation / purification method known to those skilled in the art.
  • the present invention also provides a polynucleotide comprising at least 15 nucleotides complementary to a DNA consisting of the nucleotide sequence of SEQ ID NO: 1 or a complementary strand thereof.
  • the “complementary strand” refers to one strand of a double-stranded DNA consisting of A: T, G: C base pairs with respect to the other strand.
  • the term "complementary” is not limited to a completely complementary sequence of at least 15 contiguous nucleotide regions, but is at least 70%, preferably at least 80%, more preferably 90%, Preferably, it should have 95% or more homology on the base sequence.
  • the algorithm described in this specification may be used as an algorithm for determining homology.
  • Such a polynucleotide can be used as a probe for detecting and isolating DNA encoding the protein of the present invention, and as a primer for amplifying the DNA of the present invention.
  • a primer usually It has a chain length of 15-100 bases, preferably 15-35 bases, more preferably 20-30 bases.
  • a DNA having at least a part or all of the sequence of the DNA of the present invention and a chain length of at least 15 bases is used.
  • the 3 'region must be complementary, but a restriction enzyme recognition sequence or a tag can be added to the 5' side.
  • the polynucleotide of the present invention includes an antisense for suppressing the expression of the C-NT2RP2000329 protein of the present invention.
  • the antisense has a chain length of at least 15 bp or more, preferably 100 bp, more preferably 500 bp or more, and preferably has a chain length of 2000 bp or less in order to cause an antisense effect.
  • Such an antisense can be obtained, for example, based on the DNA sequence information shown in SEQ ID NO: 1 based on the phosphorothionate method (Stem, 1988 Physicochemical properties of phosphorot hioate oligodeoxynucleotides.Nucleic Acids Res 16, 3209-21). (1988)).
  • the DNA of the present invention and its antisense can be applied to, for example, gene therapy.
  • Alzheimer's disease can be considered as a target disease for gene therapy using the DNA of the present invention.
  • ex vivo methods such as retroviral vectors, adenoviral vectors, non-inore vectors such as ribosomes, and non-inore vectors such as ribosomes are used. It may be administered to a patient by a method such as, or in vivo.
  • the present invention also provides an antibody that binds to the protein of the present invention.
  • the form of the antibody of the present invention is not particularly limited, and includes a polyclonal antibody, a monoclonal antibody, and a part thereof having antigen-binding properties. Also includes all classes of antibodies. Furthermore, the antibodies of the present invention also include special antibodies such as humanized antibodies.
  • the antibody of the present invention can be obtained by synthesizing an oligonucleotide corresponding to the amino acid sequence according to a conventional method and immunizing a rabbit.
  • a conventional method for monoclonal antibodies, E. coli is routinely used.
  • E. coli is routinely used.
  • Antibodies that bind to the protein of the present invention may be used, for example, for the examination and diagnosis of abnormal expression or structural abnormality of these proteins, in addition to the purification of the protein of the present invention.
  • proteins are extracted from, for example, tissues, blood, or cells, and examined for abnormal expression or structure through detection of the proteins of the present invention by methods such as stamping, immunoprecipitation, and ELISA. ⁇ Can be diagnosed.
  • Antibodies that bind to the protein of the present invention may also be used for purposes such as treating diseases associated with the protein of the present invention.
  • human antibodies or humanized antibodies are preferred because of their low immunogenicity.
  • a human antibody is a mouse in which the immune system is replaced with that of a human (e.g., ⁇ Functional transplant of megabase human immunoglobul in ⁇ oc i recapitulates human antibody response in mice, Mendez, MJ et al. (1997) Nat. Genet. 15: 146-156 ").
  • a humanized antibody can be prepared by genetic recombination using the hypervariable region of a monoclonal antibody (Methods in Enzymology 203, 99-121 (1991)).
  • the present invention also provides a method for screening a compound that binds to the protein of the present invention.
  • This screening method comprises: (a) a step of bringing a test sample into contact with the protein of the present invention or a partial peptide thereof; (b) a step of detecting the binding activity between the protein or its partial peptide and the test sample; (C) selecting a compound having an activity of binding to the protein or a partial peptide thereof.
  • Specific methods include, for example, a method of contacting and purifying a test sample with an affinity column for the protein of the present invention, a method using a two-hybrid system, a West Western blotting method, a combinatorial chemistry method. Many known methods can be used, such as high-throughput screening techniques.
  • Screening can also be performed by evaluating the binding between the protein of the present invention and a test compound using a measuring device such as BIACORE (Pharmacia).
  • the test sample used for screening is not limited to these, but includes, for example, a cell extract, an expression product of a gene library, a synthetic low-molecular compound, a synthetic peptide, a natural compound, and the like.
  • the compound isolated by this screening is a candidate for a compound (agonist, antagonist) that promotes or inhibits the activity of the protein of the present invention.
  • it is a candidate for a compound that inhibits the interaction between the protein of the present invention and a molecule that interacts with the protein in vivo.
  • the present invention also provides a method for diagnosing a disease caused by abnormal expression of the gene of the present invention.
  • This diagnosis includes, for example, MELAS (stroke-like symptoms and hyperlactemia), central nervous system, epilepsy, skeletal muscle pathology, muscular disease, abnormal electron transmission, Leber disease, diabetes, Peason disease, Perkinson disease, metabolism It is expected to be effective for diagnosis of diseases such as abnormalities.
  • MELAS stroke-like symptoms and hyperlactemia
  • central nervous system epilepsy
  • skeletal muscle pathology muscular disease
  • abnormal electron transmission Leber disease
  • diabetes diabetes
  • Peason disease Perkinson disease
  • metabolism is expected to be effective for diagnosis of diseases such as abnormalities.
  • the diagnosis of the present invention can be performed by preparing a cell sample from a patient and detecting the expression level or mutation of the gene of the present invention in the cell.
  • the cell sample prepared from the patient may be appropriately selected according to the type of the disease to be diagnosed. For example, cells derived from muscle tissue can be suitably used.
  • Expression of the gene of the present invention in cells can be detected by a method known to those skilled in the art. Examples of such a method include, but are not limited to, Northern blotting and RT-PCR.
  • RNA from cells to be tested is purified and agarose gel
  • the probe of the present invention is hybridized with a radioactive-labeled probe of the gene of the present invention, and the expression of the gene of the present invention is measured based on the presence or absence of a band that appears specifically and the density of the band. .
  • RNA of a cell to be tested is purified, converted into cDNA using reverse transcriptase, and this is designated as type ⁇ , and a sequence characteristic of the gene of the present invention is defined as a primer, and The cDNA derived from the gene transcript is amplified by PCR. Since the amount of cDNA amplified by this is considered to be proportional to the amount of type II cDNA and, consequently, the amount of the transcript of the gene of the present invention, the amount of DNA fragment amplified by this PCR is determined by electrophoresis. By using such a method, the expression level of the gene of the present invention can be measured.
  • the mutation of the gene of the present invention can be detected by a method of directly determining the nucleotide sequence by PCR or the like.
  • PCR method DNA of a cell to be tested is prepared, and the DNA is amplified into a ⁇ type, and a sequence characteristic of the gene of the present invention is used as a primer to amplify the gene portion of the present invention on the chromosome of the cell.
  • the presence or absence of a mutation can be measured by confirming the nucleotide sequence.
  • the present invention also provides a method for screening a candidate compound for a drug for preventing or treating a disease caused by DNA abnormality of the present invention.
  • the screening method of the present invention is considered to be suitable for screening candidate compounds for drugs for preventing or treating the above-mentioned diseases.
  • the screening according to the present invention provides a method in which a test sample is brought into contact with a cell in which the gene of the present invention is expressed, the expression level of the gene of the present invention in the cell is measured, and the test sample is not contacted. This can be carried out by selecting a compound that increases or decreases the expression level of the gene of the present invention in the cells that have been brought into contact with the test sample.
  • test sample is not particularly limited, and examples thereof include compounds obtained by combinatorial chemistry technology (Tetrahedron (1995) 51, 8135-8137), Alternatively, a random peptide group created by applying a phage display method (J. Mol. Biol. (1991) 222, 301-310) can be used.
  • culture supernatants of microorganisms and natural components derived from plants and marine organisms are also subject to screening.
  • Other examples include, but are not limited to, biological tissue extracts, cell extracts, expression products of gene libraries, synthetic low-molecular compounds, synthetic peptides, and natural compounds.
  • Examples of cells used for screening include, but are not limited to, COS7 cells.
  • the expression of the gene of the present invention in the cells can be measured by a method known to those skilled in the art, for example, the Northern blotting method, the RT-PCR method, and the like, as in the above-described diagnosis.
  • a compound capable of altering the expression of the gene of the present invention is expected to act, for example, to regulate the expression level of the gene of the present invention in patients with the above-mentioned diseases.
  • Candidate for therapeutic drug is expected to act, for example, to regulate the expression level of the gene of the present invention in patients with the above-mentioned diseases.
  • the gene of the present invention its protein, a compound that regulates the expression of the gene, or a compound that regulates the activity of the protein is used as a drug
  • the drug itself can be used as a drug, but a known preparation can be used. It is also possible to formulate and use it by a chemical method. For example, it may be used in the form of a formulation by appropriately combining with a pharmacologically acceptable carrier or medium, specifically, sterile water, physiological saline, vegetable oil, emulsifier, suspending agent and the like.
  • Administration to a patient can be performed by a method known to those skilled in the art, such as intraarterial injection, intravenous injection, and subcutaneous injection.
  • the dose varies depending on the weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose.
  • the DNA When DNA is used as a therapeutic agent, the DNA may be incorporated into a vector for gene therapy and administered to a patient.
  • the dose and the administration method vary depending on the patient's body weight, age, symptoms and the like, but those skilled in the art will be able to select as appropriate.
  • the present invention also provides a DNA chip on which the DNA or polynucleotide of the present invention is arranged.
  • the term "DNA chip” refers to a DNA chip having a size of 1 to 10 cm 2 , such as glass or silicon, on which a large number of DNAs are precisely arranged in a predetermined order. As a result, analysis of DNA and RA can be performed in a large amount in a short time.
  • the DNA chip can be manufactured as follows. First, a partial sequence specific to the gene of the present invention is selected as a probe. This is spotted, for example, on a commonly supplied DNA chip substrate made of glass or silicon having a size of l to 10 cm 2 using a microarrayer.
  • a method for immobilizing the probe on the substrate a method known in the art can be used.
  • an amino group for covalent bonding can be introduced into the end of the probe in advance, and immobilized on a silanized substrate surface by silane coupling.
  • regions other than the region where the probe is immobilized are coated with, for example, polylysine to avoid nonspecific binding of sample DNA and RNA to the silane-coated surface.
  • One or more probes can be fixed to a single chip base.
  • FIG. 1 is a diagram showing a comparison between the amino acid sequence of the AMP phosphotransferase, which is homologous to the amino acid sequence of the protein of the present invention, and the amino acid sequence of the protein of the present invention.
  • FIG. 2 is a diagram showing a comparison of the amino acid sequence of the rat amino acid sequence of rat mitochondrial GTP-binding protein AMP phosphotransferase showing homology with the amino acid sequence of the protein of the present invention and the protein of the present invention.
  • FIG. 3 is a diagram showing a comparison between the amino acid sequence of a human mitochondrial GTP-binding protein AMP phosphotransferase showing homology with the amino acid sequence of the protein of the present invention and the amino acid sequence of the protein of the present invention.
  • BEST MODE FOR CARRYING OUT THE INVENTION the present invention will be described more specifically with reference to examples, but the present invention is not limited to the following examples. Unless otherwise specified, the method can be carried out according to a known method (Mamatis, T. at al. (1982): Molecular Cloning-A Laboratory Manual, Old Spring Harbor Laboratory, NY).
  • NT-2 neural progenitor cells purchased from Stratagene that can be differentiated into neural cells by retinoic acid treatment in teratocarcinoma cells derived from human fetal testis are cultured, and induced to differentiate by adding retinoic acid. Cultured. From the cells, mRNA was extracted by the method described in Molecular Coating, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1989). Further, according to the method described in Molecular Cloning, A Laboratory Manual, Second Edition, and Old Spring Harbor Laboratory Press (1989), an oligo (dT) cellulose column (Col laborative labs) was used to obtain poly (A) + RNA. Was purified.
  • RNA from the pol ba! A cDNA library was prepared by the gocap method [M. Maruyama and S. Sugano, Gene, 138: 171-174 (1994)].
  • oligocaplinker synthetic RNA
  • oligo (dT) adapter consisting of the sequence represented by SEQ ID NO: 4
  • the literature [Suzuki * Sugano, Protein Nucleic Acid Enzyme, 41: 197-201 (1996), Y.
  • Example 2 Analysis of a cDNA clone derived from a cDNA library prepared from NT-2 cells
  • a part of the cDNA library prepared in Example 1 was introduced into E. coli DH10B by electroporation using Gene Pulser (manufactured by Biorad). Transformants were selected by culturing on LB agar medium containing 50 g / ml ampicillin. These transformants were cultured overnight in an LB medium containing 50 g / nLL of ampicillin, and a plasmid was extracted using an automatic plasmid extractor PI100 (manufactured by Kurabo Industries, Ltd.).
  • Plasmid DNA from clones obtained from these transformants was subjected to DNA sequencing using a DNA sequencing kit (BigDye rermmator and Ycle Sequencing FII Ready Reaction Kit, manufactured by PE Biosystems) according to the manual.
  • the nucleotide sequence from the 5 'end or 3' end of each cDNA clone was analyzed using an NA sequencer (ABI PRISM 377, manufactured by PE Biosystems).
  • the 5′-end sequence and the 3′-end sequence of the cDNA clone determined in (2) were separately clustered. That is, the determined 5 'end and 3' end of the cDNA clone
  • the BLAST analysis was performed on the single-pass sequence data from each sequence data with each sequence data, and clones considered to be derived from the same gene were grouped.
  • the 'terminal sequence group' was further processed into a group (cluster) so that the 5 'terminal sequence and the 3' terminal sequence of the same clone belonged to the same group (cluster 1).
  • 5 ′ terminal sequence data of the clone sequence was characterized based on the following method.
  • Human mRNA sequence ⁇ Identity to human EST sequence Comparison of 5'-end length
  • the identity of the 5'-end and 3'-end sequences of the clone sequence to the human or other organism's mRNA sequence is at least 94% identical to the sequence compared to each sequence when the sequence length is 200 bases or more. Were considered identical.
  • the identity to the human EST sequence was considered to be identical when the length of the comparison sequence with the 5'-terminal sequence was 200 bases or more and 90% or more matches.
  • the comparison target sequence is EST
  • the 5 'end is longer than the human EST sequence in the database, or if the difference between the two is within 50 bases even for a clone with a shorter 5' end.
  • the total length is defined as the total length, and the shorter length is defined as the non-full length.
  • ATGpr [A. Salamov, T. Nishikawa, ⁇ . ⁇ . Swindells. As sessing protein coding region integrity in cDNA sequencing projects. Bioinformatics 14: 384-390 (1998)] were used for prediction of full length. .
  • the ATGprl value predicts the possibility of the full length from the calculated value, and the higher the ATGpr1 value, the higher the possibility of the full length.
  • the maximum ATGprl value and the maximum ATGpr2 value indicate the maximum ATGpr1 and ATGpr2 values predicted from all the start codons contained in the 5 'terminal sequence of the clone sequence. Using.
  • 5'-end sequence A cluster that groups the 3'-end sequences was characterized based on the following viewpoints.
  • the cluster was regarded as the same cluster as the mRNA sequence.
  • the cluster was classified as a cluster that was non-full-length with respect to the mRNA sequence or the human EST sequence. did.
  • clusters identical to the mRNA sequences of humans and other organisms (including the licensed sequences) and non-full-length clusters were excluded. From those clusters, those that met any of the following conditions were selected.
  • At least one clone contains a clone with high novelty and full length.
  • the clusters selected in (b), (c), and (d) have a lower overall length, but are still full-length and contain new clones.
  • the nucleotide sequence of the full-length cDNA was determined for the NT-2 cell-derived cDNA clones selected as described in (1) to (10) and determined to be highly likely to be novel.
  • the nucleotide sequence is mainly based on primer walking by the dideoxy terminator method using a custom synthesized DNA primer (sequencing is performed according to the manual using a DNA sequencing reagent manufactured by PE Biosystem, using a custom synthesized DNA primer). After the reaction, the DNA sequence was analyzed using the company's sequencer). The full-length nucleotide sequence was finally determined by completely overlapping the partial nucleotide sequence determined by the above method. Next, a deduced amino acid sequence was determined from the determined nucleotide sequence of the full-length cDNA.
  • cDNA clone C- The nucleotide sequence of NT2RP2000329 is shown in SEQ ID NO: 1.
  • SEQ ID NO: 2 shows the amino acid sequence of the gene product encoded by cDNA clone C-NT2RP2000329 estimated from the full-length nucleotide sequence.
  • C-NT2RP2000329 (SEQ ID NO: 1) was searched using the in-house GenBank database BLAST program. As a result, as shown in Fig. 1, C_NT2RP2000329 was produced in the nucleus of the cell of the moss as reported in the literature (Shahjahan M. et al., (1991) Gene 107: 313'-317). It showed 92% similarity to the amino acid sequence of the AMP phosphotransferase (AK3), a GTP-binding protein from tochondria. In addition, as shown in Figure 2, mitochondria produced in the nuclei of rat cells reported in the literature (Tanabe T. et al., (1993) J. Biochem. 113: 200-207).
  • AK3 AMP phosphotransferase
  • the amino acid sequence of GTP-binding protein AMP phosphotransferase (AK3) showed 90% similarity.
  • GTP binding of mitochondria produced in the nucleus of human cells reported in the literature (Xu G. et al., (1992) Genetics 13: 537-542).
  • the amino acid sequence of protein AMP phosphotransferase (AK3) showed 60% similarity.
  • C-NT2RP2000329 had a similarity of 90% or more with AK3 of human and rat, but only 60% of similarity with human AK3. Therefore, C-NT2R P2000329 is a new and different human gene.
  • the present invention provides a novel AK3-like protein (C-NT2RP2000329), a gene encoding the protein, a vector containing the gene, a transformant containing the vector, and a method for producing the protein. It is suggested that the cDNA of the present invention is associated with abnormally caused diseases such as conversion of extracellular information into intracellular information, change in cell morphology, apoptosis, cell motility, and intracellular substance transport. Therefore, the genetics of the present invention Children or proteins are effective for the development of diagnostic markers and pharmaceuticals for these diseases.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Diabetes (AREA)
  • Neurology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Obesity (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Emergency Medicine (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Endocrinology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

By an oligocap method originally developed for isolating full-length cDNA, a plural number of full-length cDNAs are isolated from a cDNA library originating in NT-2 cells treated with retinoic acid. Among these cDNAs, a clone (C-NT2RP2000329) encoding a novel AK-like protein is isolated. It is considered that C-NT2RP2000329 has functions of converting extracellular signals into intracellular signals and changing cell morphology. Therefore, it is expected that C-NT2RP2000329 is usable as an important target in diagnosing diseases associated with abnormality in these functions and developing drugs for preventing or treating these diseases.

Description

新規なアデ二レートキナーゼ 3 (A K 3 ) 様蛋白質をコードする遺伝子 技術分野  Gene encoding a novel adenylate kinase 3 (AK 3) -like protein
本発明は、新規な AK3様蛋白質、 その遺伝子、 並びにそれらの製造および用途 に関する。 背景技術  The present invention relates to novel AK3-like proteins, their genes, and their production and use. Background art
アデ二レートキナーゼ (AK) は、 MgADPの 1分子による ADPの 1分子の可逆的リ ン酸化により MgATPおよび AMPを生成する反応を触媒するホスホトランスフェラ ーゼである。 脊椎動物のアデ-レートキナ一ゼには、 これまでに AK1、 AK2、 AK 3の 3つのアイソザィムが報告れている。 このうち AK3は、細胞の核内においてコ ードされる GTP結合蛋白質 AMPホスホトランスフェラーゼであり、 肝臓、 心臓、 筋肉のミ トコンドリアのマトリクスに存在している (Shahjahan M. et al. , (1 991) Gene 107 : 313-317) 。 最近、 AK3がアポト一シスに関連していることが示 唆された (Kohler C. et al. , (1999) FEBS Lett 447 : 10) 。 また、 他の文献に おいて、 AK2がアポト一シス初期にミ トコンドリァのィンターメンブレンから放 出されるのに対し、 AK3はアポト一シスの間でもインターメンブレンに放出され なレヽことカ示された (Noma T. et al. , (1999) Biochem. Biophys. Res. Commun 2 64 : 990) 。 さらに、 AK3に関しては、 ラッ トにおいて神経の成熟再生に関与して レヽること力 s示された (Inoue S. et al. , (1999) Biochem. Biophys. Res. Commun 2 54 : 681) 。 これら報告から、 AK3は、 ミ トコンドリアに存在し、 細胞外情報の細 胞内情報への変換、 細胞の形態変化、 アポトーシス、 細胞運動、 細胞内物質輸 送などの役割を担っていると考えられる。 Adenylate kinase (AK) is a phosphotransferase that catalyzes the reaction that produces MgATP and AMP by reversible phosphorylation of one molecule of ADP by one molecule of MgADP. Three isozymes, AK1, AK2, and AK3, have been reported for vertebrate adenylate kinase. Among them, AK3 is a GTP-binding protein, AMP phosphotransferase, encoded in the nucleus of cells, and is present in the mitochondrial matrix of liver, heart, and muscle (Shahjahan M. et al., (1991) ) Gene 107: 313-317). Recently, it has been suggested that AK3 is involved in apoptosis (Kohler C. et al., (1999) FEBS Lett 447: 10). In other literature, AK2 was released from the mitochondrial intermembrane early in apoptosis, whereas AK3 was not released into the intermembrane during apoptosis. (Noma T. et al., (1999) Biochem. Biophys. Res. Commun 264: 990). Additionally, for AK3, it was shown Rereru that force s involved in the maturation regeneration of nerve in rats (Inoue S. et al, (1999 ) Biochem Biophys Res Commun 2 54:.... 681). These reports suggest that AK3 is present in mitochondria and plays a role in converting extracellular information into intracellular information, changing cell morphology, apoptosis, cell motility, and transporting intracellular substances. .
ミ トコンドリアと核との相互作用についてはいまだ十分に解析されておらず 、 医学的あるいは生物学的文献も多くないが、 ミ トコンドリアに関係する疾患 として、 これまでに、 MELAS (脳卒中様症状ち高乳酸血症) 、 中枢神経症、 てん かん、 骨格筋病理、 筋肉疾患、 電子伝達異常、 Leber病、 糖尿病、 Peason病、 P erkinson病、 代謝異常などが報告されている。 The interaction between mitochondria and the nucleus has not yet been fully analyzed. Although there is not much medical or biological literature, MELAS (stroke-like symptoms or hyperlactic acidemia), central nervous disease, epilepsy, skeletal muscle pathology, and muscular disease have been identified as mitochondrial-related diseases. , Electron transmission abnormalities, Leber's disease, diabetes, Peason's disease, Perkinson's disease, metabolic abnormalities, etc. have been reported.
AK3は、その特性からこれら疾患に関係していることが予想され、 これら疾患 の診断や予防や治療のための医薬品開発の重要な標的となると考えられる。 発明の開示  Because of its properties, AK3 is expected to be involved in these diseases, and is considered to be an important target for drug development for the diagnosis, prevention and treatment of these diseases. Disclosure of the invention
本発明は、 このような状況に鑑みてなされたものであり、 その目的は、 新規 な AK3様蛋白質、その遺伝子、並びにそれらの製造方法および用途を提供するこ とにある。  The present invention has been made in view of such circumstances, and an object of the present invention is to provide a novel AK3-like protein, a gene thereof, and a method for producing the same and use thereof.
本発明者らは、 完全長 cDNAを単離するために独自に開発したオリゴキャップ 法により、レチノイン酸処理した NT- 2細胞由来の cDNAライブラリ一から完全長 c DNAを複数単離した。単離した cDNAの一つにつき塩基配列を決定し、その構造解 析を行なったところ、公知の AK3と有意な相同性を示したため、該 cDNAが新規な AK3様の蛋白質をコードしていることが判明した (このクローンを 「C - NT2RP20 00329」 と命名した) 。 AK3は、 上記したように、 ミ トコンドリアに存在し、 細 胞外情報の細胞内情報への変換、細胞の形態変化などの役割を担い、 MELASや中 枢神経症を初めとして多岐に渡る疾患との関連が示唆されている。 C - NT2RP200 0329は、新規なヒ トの AK3であるため、 これら疾患の診断やこれら疾患の予防や 治療を行なうための薬剤の開発のための重要な標的となると考えられる。  The present inventors isolated a plurality of full-length cDNAs from a cDNA library derived from retinoic acid-treated NT-2 cells by an oligocap method uniquely developed for isolating full-length cDNAs. The nucleotide sequence of one of the isolated cDNAs was determined, and its structural analysis revealed significant homology to the known AK3, indicating that the cDNA encodes a novel AK3-like protein. (This clone was named "C-NT2RP20 00329"). As described above, AK3 is present in mitochondria and plays a role in converting extracellular information into intracellular information, changing cell morphology, etc., and plays a wide variety of diseases including MELAS and central nervous system disorders. The association has been suggested. Because C-NT2RP200 0329 is a novel human AK3, it is considered to be an important target for the diagnosis of these diseases and the development of drugs to prevent or treat these diseases.
本発明は、 新規な AK3様蛋白質 C - NT2RP2000329および該蛋白質をコードする D NA、 並びにそれらの製造および用途に関し、 より詳しくは、  The present invention relates to a novel AK3-like protein C-NT2RP2000329 and a DNA encoding the protein, and their production and use.
( 1 ) 下記 (a ) から (d ) のいずれかに記載の DNA、  (1) a DNA according to any of (a) to (d) below,
( a ) 配列番号: 2に記載のアミノ酸配列からなる蛋白質をコードする DNA。 ( b ) 配列番号: 1に記載の塩基配列のコード領域を含む DNA。 (c) 配列番号: 2に記載のアミノ酸配列において 1若しくは複数のアミノ酸 が置換、 欠失、 挿入、 および/または付加したアミノ酸配列を有し、 配列番号(a) DNA encoding a protein consisting of the amino acid sequence of SEQ ID NO: 2. (b) DNA containing the coding region of the nucleotide sequence of SEQ ID NO: 1. (c) having an amino acid sequence in which one or more amino acids have been substituted, deleted, inserted, and / or added in the amino acid sequence of SEQ ID NO: 2,
: 2に記載のアミノ酸配列からなる蛋白質と機能的に同等な蛋白質をコードす る DNA。 : DNA encoding a protein functionally equivalent to the protein consisting of the amino acid sequence of 2.
(d)配列番号: 1に記載の塩基配列からなる DNAとス トリンジェン卜な条件下 でハイブリダィズし、 配列番号: 2に記載のアミノ酸配列からなる蛋白質と機 能的に同等な蛋白質をコードする DNA。  (d) a DNA that hybridizes under stringent conditions to a DNA consisting of the nucleotide sequence of SEQ ID NO: 1 and encodes a protein functionally equivalent to the protein consisting of the amino acid sequence of SEQ ID NO: 2 .
(2) 配列番号: 2に記載のアミノ酸配列からなる蛋白質の部分べプチ ドをコ一ドする DNA、  (2) DNA encoding a partial peptide of a protein consisting of the amino acid sequence of SEQ ID NO: 2,
(3) (1) に記載の DNAによりコードされる蛋白質またはペプチド、 (3) a protein or peptide encoded by the DNA according to (1),
(4) (1) に記載の DNAが挿入されたベクター、 (4) a vector into which the DNA of (1) has been inserted,
(5) (1) に記載の DNAまたは (4) に記載のベクタ一を保持する形質 転換細胞、  (5) a transformed cell carrying the DNA of (1) or the vector of (4),
(6) (5) に記載の形質転換細胞を培養し、 該形質転換細胞またはそ の培養上清から発現させた蛋白質またはべプチドを回収する工程を含む、 ( 3 ) に記載の蛋白質またはペプチドの製造方法、  (6) The protein or peptide according to (3), which comprises a step of culturing the transformed cell according to (5) and collecting a protein or a peptide expressed from the transformed cell or a culture supernatant thereof. Manufacturing method,
(7) (3) に記載の蛋白質に結合する抗体、  (7) an antibody that binds to the protein according to (3),
(8) 配列番号: 1に記載の塩基配列からなる DNAまたはその相補鎖に相 補的な少なく とも 15ヌクレオチドを含むポリヌクレオチド、  (8) a polynucleotide comprising at least 15 nucleotides complementary to a DNA consisting of the nucleotide sequence of SEQ ID NO: 1 or a complementary strand thereof,
(9) (3) に記載の蛋白質に結合する化合物のスク リーニング方法で あって、  (9) A method for screening a compound that binds to a protein according to (3),
(a) 該蛋白質またはその部分べプチドに被検試料を接触させる工程、  (a) contacting a test sample with the protein or a partial peptide thereof,
( b ) 該蛋白質またはその部分べプチドと被検試料との結合活性を検出するェ 程、  (b) detecting the binding activity between the protein or its partial peptide and a test sample,
(c) 該蛋白質またはその部分べプチドに結合する活性を有する化合物を選択 する工程、 を含む方法、 および (1 0) (1) に記載の DNAの異常に起因する疾患を診断する方法であつ て、 (c) selecting a compound having an activity of binding to the protein or a partial peptide thereof, and (10) A method for diagnosing a disease caused by a DNA abnormality according to (1),
(a) 患者から細胞試料を調製する工程、 および  (a) preparing a cell sample from a patient; and
(b) 該細胞における (1) に記載の DNAの発現量または変異を検出する工程 、 を含む方法、  (b) detecting the expression level or mutation of the DNA according to (1) in the cell,
(1 1) (1) に記載の DNAの異常に起因する疾患の予防または治療の ための薬剤の候補化合物をスク リーニングする方法であって、  (11) A method for screening a candidate compound for a drug for prevention or treatment of a disease caused by DNA abnormality according to (1), which comprises:
(a) (1) に記載の DNAが発現している細胞に被検試料を接触させる工程、 (a) contacting the test sample with cells expressing the DNA according to (1),
(b) 該細胞における (1) に記載の DNAの発現量を測定する工程、 および(b) measuring the expression level of the DNA according to (1) in the cells; and
(c) 被検試料を接触させない場合と比較して、 工程 (b) において測定され る (1) に記載の DNA発現量を増加または低下させる化合物を選択する工程、 を含む方法、 (c) selecting a compound that increases or decreases the level of DNA expression according to (1), which is measured in step (b) and compared to the case where the test sample is not contacted, according to (1).
(1 2) (1) 若しくは (2) に記載の DNA、 (3) に記載の蛋白質若 しくはペプチド、 (4) に記載のベクター、 または (9) 若しくは (1 1) に 記載のスクリーニングにより単離される化合物を含有する医薬組成物、  (1 2) The DNA according to (1) or (2), the protein or peptide according to (3), the vector according to (4), or the screening according to (9) or (11). A pharmaceutical composition containing the compound to be isolated,
(1 3) (1) 若しくは (2) に記載の DNAまたは (8) に記載のポリ ヌクレオチドが配置された DNAチップ、 を提供するものである。  (13) A DNA chip provided with the DNA according to (1) or (2) or the polynucleotide according to (8).
本発明は、 新規な蛋白質 「C- NT2RP2000329」 を提供する。 本発明の蛋白質に 含まれるヒ ト由来の C- NT2RP2000329蛋白質のアミノ酸配列を配列番号: 2に、 該蛋白質をコードする cDNAの塩基配列を配列番号: 1に示す。 C - NT2RP200032 9遺伝子は、 アデ二レートキナーゼ 3 (AK3) に対して有意な相同性を有する、 227アミノ酸からなる蛋白質をコードする。 AK3は、 ミ トコンドリアに存在して 、 細胞外情報の細胞內情報への変換、 細胞の形態変化、 アポトーシス、 細胞運 動、 細胞内物質輸送などの役割を演じている。 C- NT2RP2000329蛋白質もその構 造上の特徴から生体内においてこのような機能を有していると考えられるため 、 C- NT2RP2000329遺伝子や蛋白質は、 これらの機能の異常に起因する疾患の診 断ゃ該疾患の予防や治療を行なうための薬剤開発への利用が期待される。 The present invention provides a novel protein "C-NT2RP2000329". The amino acid sequence of the human C-NT2RP2000329 protein contained in the protein of the present invention is shown in SEQ ID NO: 2, and the nucleotide sequence of the cDNA encoding the protein is shown in SEQ ID NO: 1. The C-NT2RP2000329 gene encodes a protein of 227 amino acids with significant homology to adenylate kinase 3 (AK3). AK3, which is present in mitochondria, plays a role in converting extracellular information into cytoplasm information, changing cell morphology, apoptosis, cell movement, and intracellular substance transport. Since the C-NT2RP2000329 protein is also considered to have such a function in vivo due to its structural characteristics, the C-NT2RP2000329 gene and protein are used for diagnosis of diseases caused by abnormalities in these functions. It is expected to be used for the development of drugs for preventing or treating the disease.
C - NT2RP2000329蛋白質は、 組み換え蛋白質として、 また天然の蛋白質として 調製することが可能である。 組み換え蛋白質は、 例えば、 後述するように本発 明の蛋白質をコ一ドする DNAを挿入したベクターを適当な宿主細胞に導入し、形 質転換体内で発現した蛋白質を精製することにより調製することが可能である The C-NT2RP2000329 protein can be prepared as a recombinant protein or as a natural protein. The recombinant protein is prepared, for example, by introducing a vector into which a DNA encoding the protein of the present invention has been inserted into an appropriate host cell as described below, and purifying the protein expressed in the transformant. Is possible
。 一方、 天然の蛋白質は、 例えば、 後述する本発明の蛋白質に対する抗体を結 合したァフィ二ティ一カラムを利用して調製することができる (Current Prot ocols in Molecular Biology edit. Ausubel et al. (1987) Publ ish. John W i ley & Sons Sect ion 16. 1-16. 19)。 ァフィ二ティ一精製に用いる抗体は、 ポリ クローナル抗体であってもモノクローナル抗体であってもよい。 また、 インビ トロ トランスレーション (例えば、 「0n the fidel ity of raR A translat ion in the nuc丄 ease - treated rabbit ret iculocyte lysate system. Dasso, M. C. , Jackson, R. J. (1989) Nucleic Aci ds Res. 17: 3129-3144 J 参照) などにより本 発明の蛋白質を調製することも可能である。 . On the other hand, a natural protein can be prepared using, for example, an affinity column to which an antibody against the protein of the present invention described below is bound (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) ) Publish. John Wiley & Sons Section 16. 1-16. 19). The antibody used for affinity purification may be a polyclonal antibody or a monoclonal antibody. In addition, in vitro translation (for example, “0n the fidelity of raR A translation in the nuc 丄 ease-treated rabbit ret iculocyte lysate system. Dasso, MC, Jackson, RJ (1989) Nucleic Acids Res. 17: 3129 It is also possible to prepare the protein of the present invention by using, for example, -3144J.
本発明には、本実施例において同定されたヒ ト由来の C - NT2RP2000329蛋白質 と機能的に同等な蛋白質が含まれる。 このような蛋白質には、 例えば、 配列番 号: 2に記載の C - NT2RP2000329蛋白質の変異体、 ホモログ、 ノくリアントなどが 含まれる。 ここで 「機能的に同等」 とは、 対象となる蛋白質が C - NT2RP2000329 蛋白質と同様の生物学的機能あるいは生化学的機能を有することを指す。 この ような機能としては、 例えば、 GTP結合蛋白質 AMPホスホトランスフェラ一ゼと しての機能が挙げられる。 蛋白質が GTP結合蛋白質 AMPホスホトランスフェラー ゼとして機能するか否かは、 例えば、 γ位のリン酸に32 Pを含む GTPと AMPおよび 蛋白質を混合し、 32Pを含む ADPが精製することを確認することにより検出する ことができる。 GTPと ADPは、 例えば、 薄層クロマトグラフで分離することがで きる。 The present invention includes a protein functionally equivalent to the human-derived C-NT2RP2000329 protein identified in this example. Such proteins include, for example, mutants, homologs, and cryptants of the C-NT2RP2000329 protein described in SEQ ID NO: 2. Here, “functionally equivalent” means that the target protein has the same biological or biochemical function as the C-NT2RP2000329 protein. Examples of such a function include a function as a GTP-binding protein AMP phosphotransferase. Whether the protein functions as a GTP-binding protein AMP phosphotransferase can be determined, for example, by mixing AMP with GTP containing 32 P at the γ-phosphate and confirming that ADP containing 32 P is purified Can be detected. GTP and ADP can be separated, for example, by thin-layer chromatography.
これら本実施例において同定された蛋白質と機能的に同等な蛋白質は、 当業 者であれば、 例えば、 蛋白質中のアミノ酸配列に変異を導入する方法 (例えば 、 部位特異的変異誘発法(Current Protocols in Molecular Biology edit. Au sub el et al. (1987) Publish. John Wi ley & Sons Section 8. 1 - 8· 5) )を利用 して調製することができる。 また、 このような蛋白質は、 自然界におけるアミ ノ酸の変異により生じることもある。 本発明には、 このように本実施例におい て同定された蛋白質と同等の機能を有する限り、 そのアミノ酸配列 (配列番号 : 2 ) において 1もしくは複数のアミノ酸が置換、 欠失、 挿入および Zもしくは 付加などにより異なる蛋白質が含まれる。 These proteins functionally equivalent to the proteins identified in this example are For example, a method for introducing a mutation into an amino acid sequence in a protein (eg, a site-directed mutagenesis method (Current Protocols in Molecular Biology edit. Au subel et al. (1987) Publish. John Wiley & It can be prepared using Sons Section 8. 1-8.5)). Such proteins may also be generated by mutations in amino acids in nature. According to the present invention, one or more amino acids in the amino acid sequence (SEQ ID NO: 2) may be substituted, deleted, inserted, and substituted with Z or Z as long as it has a function equivalent to the protein identified in this example. Different proteins are included by addition and the like.
蛋白質におけるァミノ酸の変異数や変異部位は、 その機能が保持される限り 制限はない。 変異数は、 典型的には、 30アミノ酸以内であり、 好ましくは 10ァ ミノ酸以内であり、 さらに好ましくは 5アミノ酸以内 (例えば、 3アミノ酸以内 ) である。 置換されるアミノ酸は、 蛋白質の機能の保持の観点から、 置換前の アミノ酸と似た性質を有するアミノ酸であることが好ましい。 例えば、 Ala、 V al、 Leu, Ile、 Pro, Met, Phe、 Trpは、 共に非極性アミノ酸に分類されるため 、 互いに似た性質を有すると考えられる。 また、 非荷電性としては、 Gly、 Ser 、 Thr、 Cys、 Tyr、 Asn、 Ginが挙げられる。 また、 酸性アミノ酸としては、 Asp および Gluが、 塩基性アミノ酸としては、 Lys、 Arg、 Hisが挙げられる。  The number of amino acid mutations and mutation sites in a protein are not limited as long as the function is maintained. The number of mutations is typically within 30 amino acids, preferably within 10 amino acids, and more preferably within 5 amino acids (eg, within 3 amino acids). The amino acid to be substituted is preferably an amino acid having properties similar to the amino acid before substitution from the viewpoint of maintaining the function of the protein. For example, Ala, Val, Leu, Ile, Pro, Met, Phe, and Trp are considered to have similar properties because they are all classified as nonpolar amino acids. In addition, examples of the non-charger include Gly, Ser, Thr, Cys, Tyr, Asn, and Gin. In addition, acidic amino acids include Asp and Glu, and basic amino acids include Lys, Arg, and His.
本実施例において同定された蛋白質と機能的に同等な蛋白質は、当業者に周 知のノヽィブジダイゼ一ション技術(Current Protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 6. 3-6. 4 )、 あるいは 遺伝子増幅技術 (PCR) (Current protocols in Molecular Biol ogy edit. Ausubel et al. (1987) Publish. John Wi ley & Sons Section 6. 1 -6. 4) を利用して単離することも可能である。 即ち、 当業者であれば、 本実施 例において同定された蛋白質をコードする DNA (配列番号: 1 ) またはその一部 をプローブとして、あるいは該 DNAと特異的にハイブリダィズするオリゴヌクレ ォチドをプライマーとして、 該 DNAとハイブリダィズする DNAを単離することが できる。 さらに単離した DNAを基に、 該 DNAによりコードされる蛋白質を調製す ることができる。 本発明には、 本実施例において同定された蛋白質と同等の機 能を有する限り、 これら蛋白質をコ一ドする DNAとハイブリダィズする DNAによ りコ一ドされる蛋白質が含まれる。 機能的に同等な蛋白質を単離するための生 物としては、 例えば、 ヒ ト、 マウス、 ラット、 ゥサギ、 ブタ、 ゥシ等の脊椎動 物が挙げられるが、 これらに制限されない。 Proteins functionally equivalent to the proteins identified in this example can be obtained by using known techniques known to those skilled in the art (Current Protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section). 6.3.6.4) or use gene amplification technology (PCR) (Current protocols in Molecular Biologic edit. Ausubel et al. (1987) Publish. John Wiley & Sons Section 6.1.6.4.4) It is also possible to isolate them. That is, those skilled in the art can use a DNA encoding the protein identified in this example (SEQ ID NO: 1) or a part thereof as a probe, or an oligonucleotide specifically hybridizing with the DNA as a primer. Isolating DNA that hybridizes with DNA it can. Further, based on the isolated DNA, a protein encoded by the DNA can be prepared. The present invention includes proteins encoded by DNAs that hybridize with DNAs encoding these proteins, as long as they have the same function as the proteins identified in the present example. Examples of organisms for isolating functionally equivalent proteins include, but are not limited to, vertebrate animals such as humans, mice, rats, rabbits, pigs, and rabbits.
機能的に同等な蛋白質をコ一ドする DNAを単離するためのハイプリダイゼ一 ションのス トリンジェン卜な条件は、 通常 「lxSSC、 0. 1% SDS, 37°C」 程度であ り、 より厳しい条件としては 「0. 5xSSC、 0. 1% SDS、 42°C」 程度であり、 さらに 厳しい条件としては 「0. lxSSC;、 0. 1% SDS、 65°C」 程度であり、 ハイブリダィゼ —シヨ ンの条件が厳しくなるほどプロ一ブ配列と高い相同性を有する DNAの単 離を期待しうる。 但し、 上記 SSC、 SDSおよび温度の条件の組み合わせは例示で あり、 当業者であれば、 ハイブリダィゼ一シヨンのス トリンジエンシーを決定 する上記若しくは他の要素 (例えば、 プローブ濃度、 プローブの長さ、 ハイブ リダィゼーシヨン反応時間など) を適宜組み合わせることにより、 上記と同様 のストリンジヱンシーを実現することが可能である。  Stringent conditions for hybridization for isolation of DNA encoding functionally equivalent proteins are usually `` lxSSC, 0.1% SDS, 37 ° C '', and are more stringent. The conditions were about 0.5 x SSC, 0.1% SDS, 42 ° C, and the more severe conditions were about 0.1 x SSC; 0.1% SDS, 65 ° C. It can be expected that DNA with higher homology to the probe sequence will be isolated as the conditions of the probe become stricter. However, the combination of the above SSC, SDS and temperature conditions is merely an example, and those skilled in the art will recognize the above or other factors that determine the stringency of the hybridization (eg, probe concentration, probe length, probe length, etc.). The same stringency as described above can be realized by appropriately combining the hybridization reaction time and the like.
このようなハイプリダイゼーション技術あるいは遺伝子増幅技術を利用して 単離される蛋白質は、 配列番号: 2に記載の本発明の蛋白質と比較して、 通常 、 そのアミノ酸配列において高い相同性を有する。 高い相同性とは、 少なく と も 50%以上、 さらに好ましくは 70%以上、 さらに好ましくは 90%以上 (例えば 、 93%以上、 95%以上) の配列の同一性を指す。 アミノ酸配列の相同性は、 BL AST Xによる相同性検索により決定することができる。  The protein isolated using such a hybridization technique or gene amplification technique usually has higher homology in its amino acid sequence than the protein of the present invention described in SEQ ID NO: 2. High homology refers to sequence identity of at least 50% or more, more preferably 70% or more, and even more preferably 90% or more (eg, 93% or more, 95% or more). Amino acid sequence homology can be determined by homology search using BLAST X.
本発明は、 また、 本発明の蛋白質の部分ペプチドを提供する。 本発明の蛋白 質の部分ペプチドは、 例えば、 本発明の蛋白質に結合する抗体の調製に利用す ることができる。 本発明の部分べプチドは、 少なく とも 7アミノ酸、 好ましくは 9ァミノ酸以上、 より好ましくは 12ァミノ酸以上、 より好ましくは 15ァミノ酸以 上のアミノ酸配列からなる。 本発明の部分ペプチドは、 例えば、 遺伝子工学的 手法、 公知のペプチド合成法、 あるいは本発明の蛋白質を適当なぺプチダーゼ で切断することによつて製造することができる。 The present invention also provides a partial peptide of the protein of the present invention. The partial peptide of the protein of the present invention can be used, for example, for preparing an antibody that binds to the protein of the present invention. The partial peptide of the present invention comprises at least 7 amino acids, preferably 9 amino acids or more, more preferably 12 amino acids or more, and more preferably 15 amino acids or less. Consists of the above amino acid sequence. The partial peptide of the present invention can be produced, for example, by a genetic engineering technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase.
本発明は、 また、 本発明の蛋白質をコードする DNAを提供する。 本発明の DNA としては、 本発明の蛋白質をコードしうるものであれば、 その形態に特に制限 はなく、 cDNAの他、 ゲノム DNA、 化学合成 DNAなども含まれる。 また、 本発明の 蛋白質をコ一ドしうる限り、遺伝暗号の縮重に基づく任意の塩基配列を有する!) NAが含まれる。 本発明の蛋白質をコ一ドする DNAは、 上記の'ように、 配列番号: 1に記載の DNA配列もしくはその一部をプローブとしたハイブリダィゼ一ショ ン法ゃこれら DNA配列の情報に基づき設計したプライマーを用いた遺伝子増幅 法 (PCR) 等の常法により単離することが可能である。  The present invention also provides a DNA encoding the protein of the present invention. The DNA of the present invention is not particularly limited in its form as long as it can encode the protein of the present invention, and includes genomic DNA, chemically synthesized DNA, and the like in addition to cDNA. Also, as long as the protein of the present invention can be encoded, it has an arbitrary base sequence based on the degeneracy of the genetic code! ) NA is included. As described above, the DNA encoding the protein of the present invention was designed based on the hybridization method using the DNA sequence of SEQ ID NO: 1 or a part thereof as a probe. It can be isolated by a conventional method such as a gene amplification method (PCR) using primers.
本発明は、 また、本発明の蛋白質をコードする DNAが挿入されたべクタ一を提 供する。本発明のベクタ一としては、挿入した DNAを安定に保持するものであれ ば特に制限されず、例えば 宿主に大腸菌を用いるのであれば、 クローニング用 ベクタ一としては pBluescriptベクター(Stratagene社製) などが好ましレ、。 本 発明の蛋白質を生産する目的においてべクタ一を用いる場合には、 特に発現べ クタ一が有用である。 発現べクタ一としては、例えば、試験管内発現であれば p BESTベクター (プロメガ社製) を、 大腸菌における発現であれば pETベクタ一 ( Invitrogen社製) を、 培養細胞における発現であれば pME18S- FL3ベクタ一 (Ge nBank Accession No. AB009864) を、 生物個体における発現であれば pME18Sベ クタ一 (Mol Cell Biol. 8 : 466〜472 (1988) ) を、 好適に用いることができる。 本発明の蛋白質をコ一ドする DNAのベクターへの挿入は常法、例えば、制限酵素 サ トを用レヽ 7こリ刀— ΐ 反 fi (Current protocols in Molecular Biology edi t. Ausubel et al. (1987) Publish. John Wiley & Sons. Section 11. 4〜11. 11) により行うことができる。  The present invention also provides a vector into which a DNA encoding the protein of the present invention has been inserted. The vector of the present invention is not particularly limited as long as it stably retains the inserted DNA. For example, if Escherichia coli is used as a host, a pBluescript vector (manufactured by Stratagene) or the like may be used as a cloning vector. I like it. When a vector is used for the purpose of producing the protein of the present invention, an expression vector is particularly useful. Examples of the expression vector include p BEST vector (manufactured by Promega) for in vitro expression, pET vector (manufactured by Invitrogen) for expression in E. coli, and pME18S- for expression in cultured cells. The FL3 vector (GenBank Accession No. AB009864) and the pME18S vector (Mol Cell Biol. 8: 466-472 (1988)) can be suitably used for expression in a living organism. Insertion of a DNA encoding the protein of the present invention into a vector can be carried out by a conventional method, for example, by using a restriction enzyme Sat. 7 protocols anti fi (Current protocols in Molecular Biology edit. Ausubel et al. 1987) Publish. John Wiley & Sons. Section 11.4 to 11.11).
本発明は、 また、 本発明の蛋白質をコードする DNAまたは該 DNAが挿入された ベクタ一を保持する形質転換体を提供する。 本発明のベクターが導入される宿 主細胞としては特に制限はなく、 目的に応じて種々の宿主細胞が用いられる。 宿主細胞は、 例えば、 本発明のタンパク質の製造のために使用することができ る。 タンパク質製造のための産生系は、 in vitroおよび in vivo の産生系があ る。 in vitroの産生系としては、 真核細胞を使用する産生系や原核細胞を使用 する産生系が挙げられる。 真核細胞を使用する場合、 例えば、 動物細胞、 植物 細胞、 真菌細胞を宿主に用いることができる。 また、 本発明の宿主細胞には、 C - NT2RP2000329蛋白質の機能解析や C-NT2RP2000329蛋白質を利用したその機能 阻害剤や機能促進剤のスクリ一ニングのために用いる目的の細胞も含まれる。 宿主細胞へのベクタ一導入は、 例えば、 リン酸カルシウム沈殿法、 電気パルス 牙孑 L、& (Current protocols m Molecular Biology edit. Ausubel et al. (1 987) Publ ish. John Wi ley & Sons. Sect ion 9. 1-9. 9) 、 リポフエクタミン法 (GIBCO- BRL社製) 、マイクロインジェクション法などの方法で行うことが可能 である。 形質転換体からの C-NT2RP2000329蛋白質の調製は、 当業者に公知の蛋 白質の分離 ·精製法を利用して行なうことができる。 The present invention also relates to a DNA encoding the protein of the present invention or the DNA inserted therein. A transformant carrying the vector is provided. The host cell into which the vector of the present invention is introduced is not particularly limited, and various host cells may be used depending on the purpose. Host cells can be used, for example, for the production of the proteins of the invention. Production systems for protein production include in vitro and in vivo production systems. In vitro production systems include production systems using eukaryotic cells and production systems using prokaryotic cells. When eukaryotic cells are used, for example, animal cells, plant cells, and fungal cells can be used as hosts. The host cells of the present invention also include cells intended for use in analyzing the function of the C-NT2RP2000329 protein and screening for its function inhibitor or function promoter utilizing the C-NT2RP2000329 protein. Vector transfer into host cells can be performed, for example, by the calcium phosphate precipitation method, electric pulse mosquito L, & (Current protocols m Molecular Biology edit. Ausubel et al. (1 987) Publish. John Wiley & Sons. Section 9 1-9. 9), Lipofectamine method (manufactured by GIBCO-BRL), microinjection method, etc. Preparation of the C-NT2RP2000329 protein from the transformant can be carried out by using a protein separation / purification method known to those skilled in the art.
本発明はまた、配列番号: 1に記載の塩基配列からなる DNAまたはその相補鎖 に相補的な少なく とも 15ヌクレオチドを含むポリヌクレオチドを提供する。 こ こで 「相補鎖」 とは、 A : T、 G : Cの塩基対からなる 2本鎖 DNAの一方の鎖に対する 他方の鎖を指す。 また、 「相補的」 とは、 少なく とも 15個の連続したヌクレオ チド領域で完全に相補配列である場合に限られず、 少なく とも 70% 、 好ましく は少なく とも 80% 、 より好ましくは 90% 、 さらに好ましくは 95% 以上の塩基配 列上の相同性を有すればよい。 相同性を決定するためのアルゴリズムは本明細 書に記載したものを使用すればよい。  The present invention also provides a polynucleotide comprising at least 15 nucleotides complementary to a DNA consisting of the nucleotide sequence of SEQ ID NO: 1 or a complementary strand thereof. Here, the “complementary strand” refers to one strand of a double-stranded DNA consisting of A: T, G: C base pairs with respect to the other strand. The term "complementary" is not limited to a completely complementary sequence of at least 15 contiguous nucleotide regions, but is at least 70%, preferably at least 80%, more preferably 90%, Preferably, it should have 95% or more homology on the base sequence. The algorithm described in this specification may be used as an algorithm for determining homology.
このようなポリヌクレオチドは、 本発明の蛋白質をコ一ドする DNAを検出、 単離するためのプローブとして、 また、本発明の DNAを増幅するためのプライマ 一として利用することが可能である。 プライマ一として用いる場合には、 通常 、 15〜100塩基、 好ましくは 15〜35塩基、 さらに好ましくは 20〜30塩基の鎖長を 有する。 また、 プローブとして用いる場合には、 本発明の DNAの少なく とも一部 若しくは全部の配列を有し、少なく とも 15塩基の鎖長の DNAが用いられる。 ブラ イマ一として用いる場合、 3'側の領域は相補的である必要があるが、 5'側には 制限酵素認識配列やタグなどを付加することができる。 これらプライマ一ゃプ 口一ブは、 本発明の DNAが関係する疾患の診断に有効である。 Such a polynucleotide can be used as a probe for detecting and isolating DNA encoding the protein of the present invention, and as a primer for amplifying the DNA of the present invention. When used as a primer, usually It has a chain length of 15-100 bases, preferably 15-35 bases, more preferably 20-30 bases. When used as a probe, a DNA having at least a part or all of the sequence of the DNA of the present invention and a chain length of at least 15 bases is used. When used as a primer, the 3 'region must be complementary, but a restriction enzyme recognition sequence or a tag can be added to the 5' side. These primers are effective in diagnosing a disease associated with the DNA of the present invention.
また、 本発明のポリヌクレオチドには、 本発明の C- NT2RP2000329蛋白質の発 現を抑制するためのアンチセンスが含まれる。 アンチセンスは、 アンチセンス 効果を引き起こすために、 少なく とも 15bp以上、 好ましくは 100bp、 さらに好ま しくは 500bp以上の鎖長を有し、好ましくは 2000bp以内の鎖長を有する。 このよ うなアンチセンスは、 例えば、 配列番号: 1に記載の DNAの配列情報を基にホス ホロチォネー卜法 (Stem, 1988 Phys icochemical propert ies of phosphorot hioate ol igodeoxynucleot ides. Nucleic Acids Res 16, 3209-21 (1988) ) な どにより調製することが可能である。  Further, the polynucleotide of the present invention includes an antisense for suppressing the expression of the C-NT2RP2000329 protein of the present invention. The antisense has a chain length of at least 15 bp or more, preferably 100 bp, more preferably 500 bp or more, and preferably has a chain length of 2000 bp or less in order to cause an antisense effect. Such an antisense can be obtained, for example, based on the DNA sequence information shown in SEQ ID NO: 1 based on the phosphorothionate method (Stem, 1988 Physicochemical properties of phosphorot hioate oligodeoxynucleotides.Nucleic Acids Res 16, 3209-21). (1988)).
本発明の DNAやそのアンチセンスには、 例えば、 遺伝子治療への応用が考え られる。本発明の DNAを利用した遺伝子治療の標的となる疾患としては、例えば 、 アルツハイマーが考えられる。 これら分子を遺伝子治療に用いる場合には、 例えば、 レトロウイルスベクター、 アデノウイルスベクタ一、 アデノ随伴ウイ ノレスベタターなどのゥイノレスべクタ一やリボソームなどの非ゥイノレスベクター などを利用して、 ex vivo法や in vivo法などにより患者へ投与を行えばよい。  The DNA of the present invention and its antisense can be applied to, for example, gene therapy. Alzheimer's disease can be considered as a target disease for gene therapy using the DNA of the present invention. When these molecules are used for gene therapy, for example, ex vivo methods such as retroviral vectors, adenoviral vectors, non-inore vectors such as ribosomes, and non-inore vectors such as ribosomes are used. It may be administered to a patient by a method such as, or in vivo.
本発明は、 また、 本発明の蛋白質に結合する抗体を提供する。 本発明の抗体 の形態には特に制限はなく、 ポリクローナル抗体やモノクローナル抗体または 抗原結合性を有するそれらの一部も含まれる。 また、 全てのクラスの抗体が含 まれる。 さらに、 本発明の抗体には、 ヒ ト化抗体などの特殊抗体も含まれる。  The present invention also provides an antibody that binds to the protein of the present invention. The form of the antibody of the present invention is not particularly limited, and includes a polyclonal antibody, a monoclonal antibody, and a part thereof having antigen-binding properties. Also includes all classes of antibodies. Furthermore, the antibodies of the present invention also include special antibodies such as humanized antibodies.
本発明の抗体は、ポリクロ一ナル抗体の場合には、常法に従いアミノ酸配列 に相当するオリゴぺプチドを合成して家兎に免疫することにより得ることが可 能であり (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publ ish. John Wiley Sons. Section 11. 12〜11. 13) 、 一方、 モノク ローナル抗体の場合には、 常法に従い大腸菌で発現し精製した蛋白質を用いて マウスを免疫し、 脾臓細胞と骨髄腫細胞を細胞融合させたハイプリ ドーマ細胞 の中 T らネ守ること; 0 でさる (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons . Section 11. 4〜11. 11In the case of a polyclonal antibody, the antibody of the present invention can be obtained by synthesizing an oligonucleotide corresponding to the amino acid sequence according to a conventional method and immunizing a rabbit. (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley Sons. Section 11.12-11.13), whereas for monoclonal antibodies, E. coli is routinely used. Immunize mice with the expressed and purified protein and protect them from the hybridoma cells of spleen cells and myeloma cells; protect them by T; 0 (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons.Section 11.4 to 11.11
) o ) o
本発明の蛋白質に結合する抗体は、 本発明の蛋白質の精製に加え、 例えば、 これら蛋白質の発現異常や構造異常の検査 ·診断に利用することも考えられる 。 具体的には、 例えば組織、 血液、 または細胞などから蛋白質を抽出し、 ゥェ スタンプ口ッティング、免疫沈降、 ELISA等の方法による本発明の蛋白質の検出 を通して、 発現や構造の異常の有無を検査 ·診断することができる。  Antibodies that bind to the protein of the present invention may be used, for example, for the examination and diagnosis of abnormal expression or structural abnormality of these proteins, in addition to the purification of the protein of the present invention. Specifically, proteins are extracted from, for example, tissues, blood, or cells, and examined for abnormal expression or structure through detection of the proteins of the present invention by methods such as stamping, immunoprecipitation, and ELISA. · Can be diagnosed.
本発明の蛋白質に結合する抗体は、本発明の蛋白質に関連した疾患の治療な どの目的に利用することも考えられる。 抗体を患者の治療目的で用いる場合に は、 ヒ ト抗体またはヒ ト化抗体が免疫原性の少ない点で好ましい。 ヒ ト抗体は 、 免疫系をヒ トのものと入れ換えたマウス (例えば、 「Functional transplan t of megabase human immunoglobul in 丄 oc i recapitulates human antibody r esponse in mice, Mendez, M. J. et al. (1997) Nat. Genet. 15 : 146 - 156」 参照) に免疫することにより調製することができる。 また、 ヒ ト化抗体は、 モノクロ ーナル抗体の超可変領域を用いた遺伝子組み換えによって調製することができ る(Methods in Enzymology 203, 99—121 (1991) )。  Antibodies that bind to the protein of the present invention may also be used for purposes such as treating diseases associated with the protein of the present invention. When antibodies are used for the purpose of treating patients, human antibodies or humanized antibodies are preferred because of their low immunogenicity. A human antibody is a mouse in which the immune system is replaced with that of a human (e.g., `` Functional transplant of megabase human immunoglobul in 丄 oc i recapitulates human antibody response in mice, Mendez, MJ et al. (1997) Nat. Genet. 15: 146-156 "). Moreover, a humanized antibody can be prepared by genetic recombination using the hypervariable region of a monoclonal antibody (Methods in Enzymology 203, 99-121 (1991)).
また、本発明は、本発明の蛋白質に結合する化合物のスクリ一ニング方法を 提供する。 このスクリーニング方法は、 (a ) 本発明の蛋白質またはその部分 ペプチドに被検試料を接触させる工程、 (b ) 該蛋白質またはその部分べプチ ドと被検試料との結合活性を検出する工程、 および (c ) 該蛋白質またはその 部分べプチドに結合する活性を有する化合物を選択する工程、 を含む。 具体的な方法としては、例えば、本発明の蛋白質のァフィ二ティ一カラムに 被検試料を接触させ精製する方法、 twoハイプリ ッドシステムを利用する方法、 ウェス トウェスタンブロッティング法、 コンビナトリァルケミス トリ一技術に おけるハイスループッ トスクリ一二ングによる方法など多くの公知の方法を利 用することができる。 また、 BIACORE (Pharmacia社) などの測定装置を利用し て、 本発明の蛋白質と被検化合物との結合を評価することによりスクリーニン グを行うこともできる。 スク リーニングに用いる被検試料としては、 これらに 制限されないが、 例えば、 細胞抽出液、 遺伝子ライブラリ一の発現産物、 合成 低分子化合物、 合成ペプチド、 天然化合物などが挙げられる。 The present invention also provides a method for screening a compound that binds to the protein of the present invention. This screening method comprises: (a) a step of bringing a test sample into contact with the protein of the present invention or a partial peptide thereof; (b) a step of detecting the binding activity between the protein or its partial peptide and the test sample; (C) selecting a compound having an activity of binding to the protein or a partial peptide thereof. Specific methods include, for example, a method of contacting and purifying a test sample with an affinity column for the protein of the present invention, a method using a two-hybrid system, a West Western blotting method, a combinatorial chemistry method. Many known methods can be used, such as high-throughput screening techniques. Screening can also be performed by evaluating the binding between the protein of the present invention and a test compound using a measuring device such as BIACORE (Pharmacia). The test sample used for screening is not limited to these, but includes, for example, a cell extract, an expression product of a gene library, a synthetic low-molecular compound, a synthetic peptide, a natural compound, and the like.
このスクリ一二ングにより単離される化合物は、 本発明の蛋白質の活性を促 進または阻害する化合物 (ァゴ二ス ト、 アンタゴニス ト) の候補となる。 また 、 生体内において、 本発明の蛋白質とこれと相互作用する分子との該相互作用 を阻害する化合物の候補となる。  The compound isolated by this screening is a candidate for a compound (agonist, antagonist) that promotes or inhibits the activity of the protein of the present invention. In addition, it is a candidate for a compound that inhibits the interaction between the protein of the present invention and a molecule that interacts with the protein in vivo.
また、 本発明は、 本発明の遺伝子の発現異常に起因する疾患を診断する方法 を提供する。  The present invention also provides a method for diagnosing a disease caused by abnormal expression of the gene of the present invention.
この診断は、 例えば、 MELAS (脳卒中様の症状と高乳酸血症) 、 中枢神経症、 て んかん、 骨格筋病理、 筋肉疾患、 電子伝達異常、 Leber病、 糖尿病、 Peason病 、 Perkinson病、 代謝異常などの疾患の診断への有効性が期待される。 This diagnosis includes, for example, MELAS (stroke-like symptoms and hyperlactemia), central nervous system, epilepsy, skeletal muscle pathology, muscular disease, abnormal electron transmission, Leber disease, diabetes, Peason disease, Perkinson disease, metabolism It is expected to be effective for diagnosis of diseases such as abnormalities.
本発明の診断は、 患者から細胞試料を調製し、 該細胞における本発明の遺伝 子の発現量または変異を検出することにより行なうことができる。 患者から調 製する細胞試料としては、 診断する疾患の種類に応じて適宜選択すればよい。 例えば、 筋組織由来の細胞を好適に用いることができる。  The diagnosis of the present invention can be performed by preparing a cell sample from a patient and detecting the expression level or mutation of the gene of the present invention in the cell. The cell sample prepared from the patient may be appropriately selected according to the type of the disease to be diagnosed. For example, cells derived from muscle tissue can be suitably used.
細胞における本発明の遺伝子の発現は、 当業者に公知の方法で検出すること ができる。 このような方法としては、 例えば、 ノーザンブロッテイング法や RT - PCR法などを例示することができるがこれらに制限されない。 ノーザンプロッ ティング法においては、 検査対象となる細胞の RNAを精製し、 ァガロースゲル 中にて電気泳動し、 メンブレンにプロットしたものに、 本発明の遺伝子を放射 能などで標識したプローブをハイブリダィズさせ、 特異的に出現するバンドの 有無、 濃淡で本発明の遺伝子の発現を測定する。 また、 RT- PCR法では、 検査対 象となる細胞の RNAを精製し、逆転写酵素により cDNAとし、 これを铸型として 、 本発明の遺伝子に特徴的な配列をプライマ一として、 本発明の遺伝子の転写 産物由来の cDNAを PCRにより増幅する。 これにより増幅された cDNAの量は铸 型となった cDNAの量、ひいては本発明の遺伝子の転写産物の量に比例すると考 えられるので、 この PCRにより増幅された DNA断片の量を電気泳動法などを用 いて測定することで、 本発明の遺伝子の発現量を測定できる。 Expression of the gene of the present invention in cells can be detected by a method known to those skilled in the art. Examples of such a method include, but are not limited to, Northern blotting and RT-PCR. In the Northern Plotting method, RNA from cells to be tested is purified and agarose gel The probe of the present invention is hybridized with a radioactive-labeled probe of the gene of the present invention, and the expression of the gene of the present invention is measured based on the presence or absence of a band that appears specifically and the density of the band. . In the RT-PCR method, RNA of a cell to be tested is purified, converted into cDNA using reverse transcriptase, and this is designated as type 铸, and a sequence characteristic of the gene of the present invention is defined as a primer, and The cDNA derived from the gene transcript is amplified by PCR. Since the amount of cDNA amplified by this is considered to be proportional to the amount of type II cDNA and, consequently, the amount of the transcript of the gene of the present invention, the amount of DNA fragment amplified by this PCR is determined by electrophoresis. By using such a method, the expression level of the gene of the present invention can be measured.
また、 本発明の遺伝子の変異の検出は、 PCR 法などにより直接塩基配列を決 定する方法を用いることができる。 PCR法においては、検査対象となる細胞の D NAを調製し、 その DNAを铸型に、 本発明の遺伝子に特徴的な配列をプライマー として、 細胞の染色体上の本発明の遺伝子部分を増幅し、 その塩基配列を確認 することで変異の有無を測定できる。 その他、 大量の検体がある場合には、 SS CP法などを用いると好適である。  The mutation of the gene of the present invention can be detected by a method of directly determining the nucleotide sequence by PCR or the like. In the PCR method, DNA of a cell to be tested is prepared, and the DNA is amplified into a 铸 type, and a sequence characteristic of the gene of the present invention is used as a primer to amplify the gene portion of the present invention on the chromosome of the cell. However, the presence or absence of a mutation can be measured by confirming the nucleotide sequence. In addition, when there is a large amount of sample, it is preferable to use the SSCP method.
また、 本発明は、 本発明の DNAの異常に起因する疾患の予防または治療のた めの薬剤の候補化合物をスクリ一ニングする方法を提供する。 本発明のスクリ 一二ング方法は、 上記した疾患の予防や治療のための薬剤の候補化合物をスク リーニングするのに好適であると考えられる。  The present invention also provides a method for screening a candidate compound for a drug for preventing or treating a disease caused by DNA abnormality of the present invention. The screening method of the present invention is considered to be suitable for screening candidate compounds for drugs for preventing or treating the above-mentioned diseases.
本発明のスクリ一ユングは、 本発明の遺伝子が発現している細胞に被検試料 を接触させ、 該細胞における本発明の遺伝子の発現量を測定し、 被検試料を接 触させなレヽ場合と比較して、 被検試料を接触させた細胞における本発明の遺伝 子の発現量を増加または低下させる化合物を選択することにより実施すること ができる。  The screening according to the present invention provides a method in which a test sample is brought into contact with a cell in which the gene of the present invention is expressed, the expression level of the gene of the present invention in the cell is measured, and the test sample is not contacted. This can be carried out by selecting a compound that increases or decreases the expression level of the gene of the present invention in the cells that have been brought into contact with the test sample.
被検試料としては、 特に制限はなく、 例えば、 コンビナトリアル 'ケミスト リー技術 (Tetrahedron ( 1995) 51 , 8135-8137) によって得られた化合物群、 あるいはファージ 'ディスプレイ法 (J. Mol. Biol. (1991) 222, 301- 310) な どを応用して作成されたランダム ·ペプチド群を用いることができる。 また、 微生物の培養上清や、 植物、 海洋生物由来の天然成分などもスク リーニングの 対象となる。 その他、 生体組織抽出物、 細胞抽出液、 遺伝子ライブラリーの発 現産物、 合成低分子化合物、 合成ペプチド、 天然化合物などが挙げられるが、 これらに制限されない。 The test sample is not particularly limited, and examples thereof include compounds obtained by combinatorial chemistry technology (Tetrahedron (1995) 51, 8135-8137), Alternatively, a random peptide group created by applying a phage display method (J. Mol. Biol. (1991) 222, 301-310) can be used. In addition, culture supernatants of microorganisms and natural components derived from plants and marine organisms are also subject to screening. Other examples include, but are not limited to, biological tissue extracts, cell extracts, expression products of gene libraries, synthetic low-molecular compounds, synthetic peptides, and natural compounds.
スクリーニングに用いる細胞としては、 例えば、 C0S7細胞が挙げられるが、 これに制限されない。 細胞における本発明の遺伝子の発現は、 上記診断の場合 と同様に、 例えば、 ノーザンブロッテイング法、 RT - PCR法など、 当業者に公知 の方法で測定することができる。  Examples of cells used for screening include, but are not limited to, COS7 cells. The expression of the gene of the present invention in the cells can be measured by a method known to those skilled in the art, for example, the Northern blotting method, the RT-PCR method, and the like, as in the above-described diagnosis.
この測定の結果、 本発明の遺伝子の発現を変化させることができる化合物は 、 例えば、 上記した疾患の患者における本発明の遺伝子の発現量を調節する作 用が期待され、 これら疾患の予防薬や治療薬の候補となる。  As a result of this measurement, a compound capable of altering the expression of the gene of the present invention is expected to act, for example, to regulate the expression level of the gene of the present invention in patients with the above-mentioned diseases. Candidate for therapeutic drug.
本発明の遺伝子、 その蛋白質、 該遺伝子の発現を制御する化合物、 あるいは 該蛋白質の活性を制御する化合物を医薬品として用いる場合には、 それ自体を 医薬品として用いることも可能であるが、 公知の製剤学的方法により製剤化し て用いることも可能である。 例えば、 薬理学上許容される担体もしくは媒体、 具体的には、 滅菌水や生理食塩水、 植物油、 乳化剤、 懸濁剤などと適宜組み合 わせて製剤化して用いることが考えられる。 患者への投与は、 例えば、 動脈内 注射、 静脈内注射、 皮下注射など当業者に公知の方法により行いうる。 投与量 は、 患者の体重や年齢、 投与方法などにより変動するが、 当業者であれば適当 な投与量を適宜選択することが可能である。 また、 DNAを治療薬として使用する 場合には、該 DNAを遺伝子治療用ベクターに組込み、患者に投与することも考え られる。 投与量、 投与方法は、 患者の体重や年齢、 症状などにより変動するが 、 当業者であれば適宜選択することが可能であろう。  When the gene of the present invention, its protein, a compound that regulates the expression of the gene, or a compound that regulates the activity of the protein is used as a drug, the drug itself can be used as a drug, but a known preparation can be used. It is also possible to formulate and use it by a chemical method. For example, it may be used in the form of a formulation by appropriately combining with a pharmacologically acceptable carrier or medium, specifically, sterile water, physiological saline, vegetable oil, emulsifier, suspending agent and the like. Administration to a patient can be performed by a method known to those skilled in the art, such as intraarterial injection, intravenous injection, and subcutaneous injection. The dose varies depending on the weight and age of the patient, the administration method, and the like, but those skilled in the art can appropriately select an appropriate dose. When DNA is used as a therapeutic agent, the DNA may be incorporated into a vector for gene therapy and administered to a patient. The dose and the administration method vary depending on the patient's body weight, age, symptoms and the like, but those skilled in the art will be able to select as appropriate.
また、 本発明は、 本発明の DNAまたはポリヌクレオチドが配置された DNAチッ プを提供する。 本発明において 「DNAチップ」 とは、 l〜10cm2の大きさのガラス 、 シリコンなどの DNAチップ基板上に多数の DNAを決められた順に精密に並べた ものを意味する。 これにより DNA、 R Aの解析を短時間で大量に行なうことがで きる。 DNAチップは、 以下のようにして製造することができる。 まず、 本発明の 遺伝子に特異的な部分配列をプローブとして選択しておく。 これを、 例えば、 一般的に供給されている l〜10cm2の大きさのガラス、シリコンなどの DNAチップ 基板上にマイクロアレイヤーを用いてスポットする。 基盤上へのプローブの固 定方法は、 当技術分野で公知の方法を用いることができる。 例えば、 予めプロ —ブの末端に共有結合のためのアミノ基を導入し、 シラン化した基盤表面上に シランカップリングによって固定することができる。 プローブの固定後、 プロ —ブを固定した領域以外の領域は、 例えば、 ポリ リジンでコートして、 サンプ ルの DNAや RNAなどがシランコート面に非特異的に結合するのを回避する。 1枚の チップ用基盤に固定するプローブは、 1種類でも複数でもよレ、。 図面の簡単な説明 The present invention also provides a DNA chip on which the DNA or polynucleotide of the present invention is arranged. Offer In the present invention, the term "DNA chip" refers to a DNA chip having a size of 1 to 10 cm 2 , such as glass or silicon, on which a large number of DNAs are precisely arranged in a predetermined order. As a result, analysis of DNA and RA can be performed in a large amount in a short time. The DNA chip can be manufactured as follows. First, a partial sequence specific to the gene of the present invention is selected as a probe. This is spotted, for example, on a commonly supplied DNA chip substrate made of glass or silicon having a size of l to 10 cm 2 using a microarrayer. As a method for immobilizing the probe on the substrate, a method known in the art can be used. For example, an amino group for covalent bonding can be introduced into the end of the probe in advance, and immobilized on a silanized substrate surface by silane coupling. After immobilization of the probe, regions other than the region where the probe is immobilized are coated with, for example, polylysine to avoid nonspecific binding of sample DNA and RNA to the silane-coated surface. One or more probes can be fixed to a single chip base. BRIEF DESCRIPTION OF THE FIGURES
図 1は、 本発明の蛋白質のアミノ酸配列と相同性を示したゥシミ トコンドリ ァ GTP結合蛋白質 AMPホスホトランスフェラ一ゼのアミノ酸配列と本発明の蛋白 質とのアミノ酸配列の比較を示す図である。  FIG. 1 is a diagram showing a comparison between the amino acid sequence of the AMP phosphotransferase, which is homologous to the amino acid sequence of the protein of the present invention, and the amino acid sequence of the protein of the present invention.
図 2は、 本発明の蛋白質のアミノ酸配列と相同性を示したラッ トミ トコンド リァ GTP結合蛋白質 AMPホスホトランスフェラーゼのァミノ酸配列と本発明の蛋 白質とのアミノ酸配列の比較を示す図である。  FIG. 2 is a diagram showing a comparison of the amino acid sequence of the rat amino acid sequence of rat mitochondrial GTP-binding protein AMP phosphotransferase showing homology with the amino acid sequence of the protein of the present invention and the protein of the present invention.
図 3は、 本発明の蛋白質のアミノ酸配列と相同性を示したヒ トミ トコンドリ ァ GTP結合蛋白質 AMPホスホトランスフェラ一ゼのアミノ酸配列と本発明の蛋白 質とのアミノ酸配列の比較を示す図である。 発明を実施するための最良の形態 次に、 本発明を実施例によりさらに具体的に説明するが、 本発明は下記実施 例に限定されるものではない。 なお、 特に断りがない場合は、 公知の方法 (Ma matis, T. at al. (1982) : Molecular Cloning - A Laboratory Manual C old Spring Harbor Laboratory, NY) に従って実施可能である。 FIG. 3 is a diagram showing a comparison between the amino acid sequence of a human mitochondrial GTP-binding protein AMP phosphotransferase showing homology with the amino acid sequence of the protein of the present invention and the amino acid sequence of the protein of the present invention. . BEST MODE FOR CARRYING OUT THE INVENTION Next, the present invention will be described more specifically with reference to examples, but the present invention is not limited to the following examples. Unless otherwise specified, the method can be carried out according to a known method (Mamatis, T. at al. (1982): Molecular Cloning-A Laboratory Manual, Old Spring Harbor Laboratory, NY).
[実施例 1 ] オリゴキャップ法による NT- 2細胞からの cDNAライブラリ一の作 製  [Example 1] Production of a cDNA library from NT-2 cells by the oligocap method
ヒ ト胎児精巣由来のテラトカルシノーマ細胞でレチノイン酸処理により神経 細胞に分化可能な NT- 2神経前駆細胞 (Stratagene社より購入) を培養し、 レチ ノイン酸を添加して分化誘導させ、 2週間培養した。 該細胞より、 Molecular C 丄 oning, A Laboratory Manual, Second Edition, Cold Spring Harbor Labora tory Press (1989)記載の方法により mRNAを抽出した。 さらに、 Molecular Clo ning, A Laboratory Manual, Second Edit ion, し old Spring Harbor Laborato ry Press (1989)記載の方法にしたがって、 オリゴ(dT)セルロースカラム (Col laborative labs) を用い、 poly (A) + RNAを精製した。  NT-2 neural progenitor cells (purchased from Stratagene) that can be differentiated into neural cells by retinoic acid treatment in teratocarcinoma cells derived from human fetal testis are cultured, and induced to differentiate by adding retinoic acid. Cultured. From the cells, mRNA was extracted by the method described in Molecular Coating, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1989). Further, according to the method described in Molecular Cloning, A Laboratory Manual, Second Edition, and Old Spring Harbor Laboratory Press (1989), an oligo (dT) cellulose column (Col laborative labs) was used to obtain poly (A) + RNA. Was purified.
該 polバ A) + RNAより、 ォ!;ゴキャップ法 [M. Maruyama and S. Sugano, Gen e, 138 : 171-174 (1994) ]により cDNAライブラリーを作製した。配列番号: 3で 表される配列からなるオリゴキヤップリンカ一 (合成 RNA) および配列番号: 4 で表される配列からなるオリゴ(dT)アダプターを用いて、 文献 [鈴木 *菅野, 蛋白質 核酸 酵素, 41 : 197-201 (1996)、 Y. Suzuki et al. , Gene, 200: 14 9-156 (1997) ]に記載してあるように BAP (Bacterial Alkal ine Phosphatase) 処理、 TAP (Tobacco Acid Pyrophosphatase) 処理、 RNAライゲーシヨン、 第一 鎖 cDNAの合成と RNAの除去を行った。 次いで、 配列番号: 5で表される 5'末端側 および配列番号: 6で表される 3'末端側の PCRプライマ一を用い、 PCR (polyme rase chain reaction)により 2本鎖 cDNAに変換し、 得られた DNA断片を Sf ilで切 断した。 次いで、 Dralllで切断したベクタ一 pME18SFL3 (GenBank AB009864) に cDNAの方向性を決めてクロ一ニングし、 cDNAライブラリーを作製した。 pME18S FL3のクロ一ン化部位は非対称性の Dralllサイ トとなっており、cDNA断片の末端 にはこれと相補的な Sfi l部位を付加しているので、クローン化した cDNA断片は S R etプロモータ一の下流に一方向性に揷入される。 A) + RNA from the pol ba! A cDNA library was prepared by the gocap method [M. Maruyama and S. Sugano, Gene, 138: 171-174 (1994)]. Using an oligocaplinker (synthetic RNA) consisting of the sequence represented by SEQ ID NO: 3 and an oligo (dT) adapter consisting of the sequence represented by SEQ ID NO: 4, the literature [Suzuki * Sugano, Protein Nucleic Acid Enzyme, 41: 197-201 (1996), Y. Suzuki et al., Gene, 200: 149-156 (1997)], BAP (Bacterial Alkaline Phosphatase) treatment, TAP (Tobacco Acid Pyrophosphatase) Treatment, RNA ligation, synthesis of first-strand cDNA and removal of RNA were performed. Then, using a 5′-end PCR primer represented by SEQ ID NO: 5 and a 3′-end PCR primer represented by SEQ ID NO: 6, it was converted into double-stranded cDNA by PCR (polymerase chain reaction). The obtained DNA fragment was cut with Sfil. Next, the direction of the cDNA was determined and cloned into the vector pME18SFL3 (GenBank AB009864) cut with Dralll to prepare a cDNA library. pME18S The cloned site of FL3 is an asymmetric Dralll site, and a complementary Sfil site is added to the end of the cDNA fragment. Is introduced in a one-way direction downstream of.
[実施例 2 ] NT - 2細胞から作製した cDNAライブラリ一由来の cDNAクローンの 解析 [Example 2] Analysis of a cDNA clone derived from a cDNA library prepared from NT-2 cells
( 1 ) cDNAクローンの単離  (1) Isolation of cDNA clone
実施例 1で作製した cDNAライブラリ一の一部をジーンパルサー (Biorad社製 ) を用いてエレク トロポレーシヨン法で大腸菌 DH10B株に導入した。形質転換体 は、 アンピシリンを 50 g/ml含有する LB寒天培地上で培養して選択した。 これ らの形質転換体をアンピシリンを 50 g/nLL含有する LB培地で一晩培養し、 ブラ スミ ド自動抽出機 PI100 (クラボウ社製) を用いてプラスミ ドを抽出した。  A part of the cDNA library prepared in Example 1 was introduced into E. coli DH10B by electroporation using Gene Pulser (manufactured by Biorad). Transformants were selected by culturing on LB agar medium containing 50 g / ml ampicillin. These transformants were cultured overnight in an LB medium containing 50 g / nLL of ampicillin, and a plasmid was extracted using an automatic plasmid extractor PI100 (manufactured by Kurabo Industries, Ltd.).
( 2 ) 単離された cDNAクローンの塩基配列の解析 (2) Analysis of the nucleotide sequence of the isolated cDNA clone
これらの形質転換体より得たクローンのプラスミ ド DNAについて、 DNAシ一ケ ンンング^桑 (BigDye rermmator し ycle Sequencing F¾ Ready Reaction Kit , PE Biosystems社製) を用い、 マニュアルに従ってシ一ケンシング反応後、 D NAシーケンサー (ABI PRISM 377, PE Biosystems社製) で各 cDNAクローンの 5' 末端または 3'末端からの塩基配列を解析した。  Plasmid DNA from clones obtained from these transformants was subjected to DNA sequencing using a DNA sequencing kit (BigDye rermmator and Ycle Sequencing FII Ready Reaction Kit, manufactured by PE Biosystems) according to the manual. The nucleotide sequence from the 5 'end or 3' end of each cDNA clone was analyzed using an NA sequencer (ABI PRISM 377, manufactured by PE Biosystems).
5'末端側からの塩基配列の決定には配列番号: 7で表される ME761FWを、 3' 末端側からの塩基配列の決定には配列番号: 8で表される ME1250RVをシ一ケン ス用プライマーとして用いた。  For determination of the base sequence from the 5 'end, use ME761FW represented by SEQ ID NO: 7, and for determination of the base sequence from the 3' end, use ME1250RV represented by SEQ ID NO: 8. Used as primer.
( 3 ) cDNAクローンの 5'末端配列と 3'末端配列のクラスター化 (3) Clustering of 5 'and 3' end sequences of cDNA clones
( 2 ) で決定した cDNAクローンの 5'末端配列と 3'末端配列を、 それぞれ別々 にクラスタリングした。 すなわち、 cDNAクローンの決定した 5'末端及び 3'末端 からのシングルパスシーケンスデータは、各配列デ一タとの間で BLAST解析を行 い、 同一遺伝子に由来すると思われるクローンのグループ化を行った。 5'末端 配列では相同性 95%以上のコンセンサス配列が 300塩基対以上、 3'末端配列では 相同性 90%以上のコンセンサス配列が 200塩基対以上の場合、 同一グループとし た 5'末端配列グループ 3'末端配列グループはさらに、 同一クローンの 5'末端配 列と 3'末端配列が同一グループ (クラスタ一) に属するようグループ (クラス ター) 化処理を行った。 The 5′-end sequence and the 3′-end sequence of the cDNA clone determined in (2) were separately clustered. That is, the determined 5 'end and 3' end of the cDNA clone The BLAST analysis was performed on the single-pass sequence data from each sequence data with each sequence data, and clones considered to be derived from the same gene were grouped. If the consensus sequence with a homology of 95% or more is 300 base pairs or more in the 5 'end sequence and the consensus sequence with a homology of 90% or more is 200 base pairs or more in the 3' end sequence The 'terminal sequence group' was further processed into a group (cluster) so that the 5 'terminal sequence and the 3' terminal sequence of the same clone belonged to the same group (cluster 1).
( 4 ) cDNAクローン配列の特徴付け (4) Characterization of cDNA clone sequence
クローン配列の 5'末端配列データは、 次の方法に基づいて特徴付けした。 5 ′ terminal sequence data of the clone sequence was characterized based on the following method.
(1) GenBankを対象にした BlastNによる相同性検索により、 ヒ トゃ他生物の mRNA 配列(権利化された配列を含む)ゃヒ ト EST配列に対して同一であるかを確認す る。 (1) By homology search using BlastN for GenBank, it is confirmed whether the sequence is identical to the human {other mRNA sequence (including the licensed sequence)} / human EST sequence.
(2)ヒ ト mRNA配列ゃヒ ト EST配列より 5'末端端が長いかを確認する。  (2) Confirm that the human mRNA sequence is longer at the 5 'end than the human EST sequence.
(3)全長性を予測する ATGprプログラム [A. Salamov, T. Nishikawa, M. B. Sw indells. Assessing protein coding region integrity in cDNA sequencing projects. Bioinf ormatics 14: 384-390 (1998) ]により 5'末端配列中のすべて の開始コ ドンに由来する ATGprl、 ATGpr2値を決定する。  (3) The ATGpr program [A. Salamov, T. Nishikawa, MB Sw indells. Assessing protein coding region integrity in cDNA sequencing projects. Bioinf ormatics 14: 384-390 (1998)] Determine ATGprl and ATGpr2 values from all start codons.
(4) GenBankを対象にした BlastNによる相同性検索により同一としたヒ ト EST配 列数を決定する。  (4) Determine the number of identical human EST sequences by homology search using BlastN for GenBank.
また、 クローン配列の 3'末端配列データの特徴付けは前出の(1)および(4)に ついて行った。  Characterization of the 3'-terminal sequence data of the clone sequence was performed for (1) and (4) above.
これら特徴付けを行ったクローン配列のデータをもとに新規でかつ全長であ る可能性の高い cDNAクローンの選抜を行った。  Based on the data of the clone sequences thus characterized, a new and potentially full-length cDNA clone was selected.
( 5 ) ヒ ト mRNA配列ゃヒ ト EST配列に対しての同一性 5'末端の長さの比較 クローン配列の 5'末端、 および 3'末端配列の、 ヒ トゃ他生物の mR A配列に対 する同一性は、 各配列との比較配列部分の長さが 200塩基以上で、 94%以上一致 の場合に同一と見なした。ヒ ト EST配列に対する同一性は 5'末端配列との比較配 列部分の長さが 200塩基以上で、 90%以上で一致の場合に同一と見なした。 (5) Human mRNA sequence ゃ Identity to human EST sequence Comparison of 5'-end length The identity of the 5'-end and 3'-end sequences of the clone sequence to the human or other organism's mRNA sequence is at least 94% identical to the sequence compared to each sequence when the sequence length is 200 bases or more. Were considered identical. The identity to the human EST sequence was considered to be identical when the length of the comparison sequence with the 5'-terminal sequence was 200 bases or more and 90% or more matches.
ヒ ト mR A配列を比較配列とし、 5'末端の長さを比較する際には 5'末端配列の 長さがヒ ト mRNA配列より長い場合、 または 5'末端配列が翻訳開始コドンを含む 場合、 全長とした。 比較対象配列が ESTの場合には、 データベース中のヒ ト EST 配列より長く 5'末端が伸びている場合、 あるいは 5'末端が短いクローンでも両 者の差が 50塩基以内である場合を便宜的に全長とし、 それ以上短い場合を非全 長とした。  When comparing the length of the 5 'end with the human mRNA sequence as a comparison sequence, when the length of the 5' end sequence is longer than the human mRNA sequence, or when the 5 'end sequence contains a translation initiation codon , And the total length. When the comparison target sequence is EST, it is convenient if the 5 'end is longer than the human EST sequence in the database, or if the difference between the two is within 50 bases even for a clone with a shorter 5' end. The total length is defined as the total length, and the shorter length is defined as the non-full length.
( 6 ) ATGprによる全長性の予測 (6) Prediction of full length by ATGpr
全長性の予測には ATGpr [A. Salamov, T. Nishikawa, Μ. Β. Swindells. As sessing protein coding region integrity in cDNA sequencing projects. B ioinformatics 14: 384-390 (1998) ] による解析結果を用いた。 ATGprl値は計 算値から全長である可能性を予測する値であり、 ATGpr 1値が高いほど全長であ る可能性が高い。 なお、最大 ATGprl値及び最大 ATGpr2値とは、 クローン配列の 5 '末端配列に含まれるすべての開始コドンから予測される ATGpr 1値及び ATGpr 2 値の最大値を示し、 特徴付けにはこの値を用いた。  The results of ATGpr [A. Salamov, T. Nishikawa, Μ. Β. Swindells. As sessing protein coding region integrity in cDNA sequencing projects. Bioinformatics 14: 384-390 (1998)] were used for prediction of full length. . The ATGprl value predicts the possibility of the full length from the calculated value, and the higher the ATGpr1 value, the higher the possibility of the full length. The maximum ATGprl value and the maximum ATGpr2 value indicate the maximum ATGpr1 and ATGpr2 values predicted from all the start codons contained in the 5 'terminal sequence of the clone sequence. Using.
( 7 ) 相同性検索による同一 EST配列数からの新規性の予測 (7) Prediction of novelty from the number of identical EST sequences by homology search
5'末端配列 3'末端配列それぞれに対して、 GenBankを用いた相同性検索から求 めた。 ヒ ト EST配列に対しては、 5'末端配列との比較配列部分の長さが 200塩基 以上にわたって 90%以上で一致する場合に同一とした。 EST配列数はそのまま特 徴付けに用い、 新規性の指標とした。 mRNA配列ばかりでなく、 EST配列に対して も同一でない 5'末端配列および 3'末端配列をもつクローンは、 新規な配列をコ —ドする遺伝子である。 同様に、 同一の EST配列数が少ない 5'末端配列、 あるい は 3,末端配列をもつクローンもまた、 新規な配列をコードする cDNAクローンで あると判定した。 5 'end sequence Each 3' end sequence was determined by homology search using GenBank. The human EST sequence was determined to be the same when the length of the comparison sequence portion with the 5'-terminal sequence was 90% or more over 200 bases or more. The number of EST sequences was used directly for the characterization and used as an index of novelty. Clones with 5 'and 3' end sequences that are not identical to mRNA sequences but also to EST sequences —The gene that is Similarly, clones having a small number of 5'-terminal sequences or 3'-terminal sequences having a small number of identical EST sequences were also determined to be cDNA clones encoding the novel sequences.
( 8 ) クラスターの特徴付け (8) Cluster characterization
5'末端配列 3'末端配列をグループ化したクラスターを、 次の観点に基づいて特 徴付けした。 5'-end sequence A cluster that groups the 3'-end sequences was characterized based on the following viewpoints.
(1) GenBankを対象にした BlastNによる相同性検索により、 ヒ トゃ他生物の mRNA 配列 (権利化された配列を含む) ゃヒ ト EST配列に対して同一であるカ  (1) By homology search using BlastN for GenBank, the human mRNA sequence (including the licensed sequence) of other organisms and the human EST sequence are identical.
クラスターに含まれるすべての 5'末端配列 3'末端配列のうち、 1配列でも mRN A配列に対して同一であった場合、そのクラスターは mRNA配列に対して同一なク ラスタ一とした。  When at least one of the 5′-terminal sequences contained in the cluster was identical to the mRNA sequence, the cluster was regarded as the same cluster as the mRNA sequence.
(2)ヒ ト mRNA配列ゃヒ ト EST配列より 5'末端が長いか。  (2) Is the human mRNA sequence longer at the 5 'end than the human EST sequence?
クラスターに含まれるすべての 5'末端配列が mRNA配列やヒ ト EST配列に対し て非全長であった場合、そのクラスタ一は mRNA配列ゃヒ ト EST配列に対して非全 長であるクラスタ一とした。  If all the 5 'terminal sequences included in the cluster were not full-length with respect to the mRNA sequence or the human EST sequence, the cluster was classified as a cluster that was non-full-length with respect to the mRNA sequence or the human EST sequence. did.
(3)全長性を予測する ATGprプログラムによる 5'末端配列中のすべての開始コド ンに由来する ATGpr 1値および ATGpr 2値。  (3) ATGpr 1 value and ATGpr 2 value derived from all start codons in the 5 ′ terminal sequence by the ATGpr program for predicting full length.
全長性を予測する ATGprプログラム [A. Salamov, T. Nishikawa, M. B. Swi ndells. Assessing protein coding region integrity in cDNA sequencing p rojects. Bioinformat ics 14 : 384-390 (1998) ] による 5'末端配列中のすべて の開始コドンに由来する ATGprl値は、 クラスタ一に含まれる 5'末端配列すベて に対して ATGprl値の最大値を、 クラスターにおける ATGprl値とした。 ATGpr2値 も同様にした。  All in the 5 'end sequence by the ATGpr program [A. Salamov, T. Nishikawa, MB Swindells. Assessing protein coding region integrity in cDNA sequencing projects. Bioinformatics 14: 384-390 (1998)] For the ATGprl value derived from the start codon of the above, the maximum value of the ATGprl value for all the 5 'terminal sequences included in the cluster was defined as the ATGprl value in the cluster. ATGpr2 values were also the same.
(4) GenBankを対象にした BlastNによる相同性検索により同一としたヒ 卜 EST配 列数。 クラスターに含まれる 5'末端配列 3'末端配列それぞれに対して EST配列数の 最大値を求め、クラスターにおける 5'末端配列の同一 EST配列数 3'末端配列の同 一 EST配列数とした。 (4) Number of human EST sequences determined to be identical by homology search using BlastN for GenBank. The maximum value of the number of EST sequences was determined for each of the 5'-terminal sequences and the 3'-terminal sequences included in the cluster, and the number of the same EST sequences of the 5'-terminal sequence in the cluster and the same number of EST sequences of the 3'-terminal sequence were determined.
( 9 ) 特徴付けからのクラスタ一の選抜方法 (9) Selection method of cluster best from characterization
特徴付けにより得られたデータから、 まず、 ヒ トゃ他生物の mR A配列 (権利 化された配列を含む) と同一なクラスター、 及び非全長なクラスタ一を除いた 。 それらクラスターの中から、 次の条件のいずれかを満たすものを選抜した。 First, from the data obtained by the characterization, clusters identical to the mRNA sequences of humans and other organisms (including the licensed sequences) and non-full-length clusters were excluded. From those clusters, those that met any of the following conditions were selected.
(a)クラスターにおける 5'末端配列の同一 EST配列数が 20以下で、 クラスターに おける ATGprl値が 0. 3を越えるクラスタ一。 (a) A cluster in which the number of identical EST sequences in the 5'-terminal sequence in the cluster is 20 or less and the ATGprl value in the cluster exceeds 0.3.
(b)クラスターにおける ATGprl値が 0. 3以下のクラスターであっても、 クラスタ —における 5'末端配列の同一 EST配列数力^以下で、かつ、 クラスターにおける 3 '末端配列の同一 EST配列数も 5以下で、かつ、 クラスター内に複数のクローンが 含まれるクラスター。  (b) Even if the ATGprl value in the cluster is 0.3 or less, the number of identical EST sequences in the 5'-terminal sequence in the cluster is not more than ^^, and the number of identical EST sequences in the 3'-terminal sequence in the cluster is also A cluster with 5 or less and multiple clones in the cluster.
(c)クラスターにおける ATGprl値が 0. 3以下のクラスターであっても、 クラスタ —における 5'末端配列の同一 EST配列数が 0で、 かつ、 クラスターにおける 3'末 端配列の同一 EST配列数が 1以上であるクラスタ一。  (c) Even if the cluster has an ATGprl value of 0.3 or less, the number of identical EST sequences in the 5 'terminal sequence in the cluster is 0, and the number of identical EST sequences in the 3' terminal sequence in the cluster is 0. Cluster one that is 1 or more.
(d)クラスタ一における ATGprl値が 0. 3以下のクラスターであっても、 クラスタ 一における 5'末端配列の同一 EST配列数が 1以上 5以下で、かつ、 クラスタ一にお ける 3'末端配列の同一 EST配列数力 SOであるクラスタ一。  (d) Even if the ATGprl value in cluster 1 is 0.3 or less, the number of identical EST sequences in the 5 'end sequence in cluster 1 is 1 or more and 5 or less, and the 3' end sequence in cluster 1 The same EST sequence number of clusters that are SO.
(a)で選抜されたクラスターには、 少なく とも 1クローンは新規性も、 全長性 も高いクローンが含まれている。 (b) , (c) , (d)で選抜されたクラスターには、全 長率は低くなるものの、 依然として全長で、 新規なクローンが含まれている。  In the cluster selected in (a), at least one clone contains a clone with high novelty and full length. The clusters selected in (b), (c), and (d) have a lower overall length, but are still full-length and contain new clones.
( 1 0 ) クラスタ一からのクローンの選抜方法 (10) How to select clones from cluster 1
同一クラスタ一内に 1クローンしか含まないものについては、そのクローンを 選抜した。 同一クラスタ一内に複数のクローンを含む場合で、 ATGprl値が 0. 3 より大のクローンが複数ある場合は、 ATGprl値がより大きい方のクローンを選 択した。 同一クラスタ一内に複数のクローンを含む場合で、 ATGprl値が 0. 3以下 のクローンが複数ある場合、 ATGpr2値が 0. 3より大ならば、 ATGpr2値がより大き い方のクローンを選択した。 また、 同一クラスター内に複数のクローンを含む 場合で、 ATGprl値、 ATGpr2値ともに 0. 3以下でも、 クラスター内で ATGprl値、 A TGpr2値がともに最大値をとるクローンがあるならば、そのクローンを選択した 。 同一クラスタ一内に複数のクローンを含む場合で、上記のような ATGpr値での 選抜ができなかった場合は、 5'末端配列 3'末端配列及びヒ ト EST配列を用いてァ センブルすることにより、 より 5'末端側に長いクロ一ンを選抜した。 ァセンブ ルには、 Sequencher (Gene Codes社製) 等を利用し、 一部、 アセンブルするこ とによっても決められなかった場合は、 対象クローンすべてを全長と判断した If only one clone is included in the same cluster, Selected. When multiple clones were included in the same cluster and there were multiple clones with an ATGprl value greater than 0.3, the clone with the higher ATGprl value was selected. If the same cluster contains multiple clones and there are multiple clones with an ATGprl value of 0.3 or less, if the ATGpr2 value is greater than 0.3, the clone with the larger ATGpr2 value was selected. . In addition, when a plurality of clones are included in the same cluster, and the ATGprl value and ATGpr2 value are both 0.3 or less, if there is a clone having the maximum ATGprl value and ATGpr2 value in the cluster, the clone is replaced. Selected . If multiple clones are included in the same cluster and selection with the ATGpr value as described above is not possible, assemble using the 5 'end sequence and the 3' end sequence and the human EST sequence. A longer clone was selected on the 5 'end side. For assembly, Sequencher (manufactured by Gene Codes) was used, and if some parts could not be determined by assembling, all target clones were judged to be full length.
( 1 1 ) cDNAクローンの全長配列の解析 (11) Analysis of full-length sequence of cDNA clone
( 1 ) 〜 (1 0 ) のようにして選抜した、 新規である可能性が高いと判断さ れた NT- 2細胞由来の cDNAクローンについて、 全長 cDNAの塩基配列を決定した。 塩基配列は主に、カスタム合成 DNAプライマ一を用いたダイデォキシターミネ一 ター法によるプライマ一ウォーキング (カスタム合成 DNAプライマーを用い、 P E Biosystem社製の DNAシーケンシング試薬でマニュアルに従ってシ一ケンシン グ反応後、 同社製のシーケンサーで DNA塩基配列を解析) によって決定した。 全 長塩基配列は上記方法により決定された部分塩基配列を完全にオーバーラップ させ最終的に確定した。 次に、 決定された全長の cDNAの塩基配列から推定アミ ノ酸配列を求めた。  The nucleotide sequence of the full-length cDNA was determined for the NT-2 cell-derived cDNA clones selected as described in (1) to (10) and determined to be highly likely to be novel. The nucleotide sequence is mainly based on primer walking by the dideoxy terminator method using a custom synthesized DNA primer (sequencing is performed according to the manual using a DNA sequencing reagent manufactured by PE Biosystem, using a custom synthesized DNA primer). After the reaction, the DNA sequence was analyzed using the company's sequencer). The full-length nucleotide sequence was finally determined by completely overlapping the partial nucleotide sequence determined by the above method. Next, a deduced amino acid sequence was determined from the determined nucleotide sequence of the full-length cDNA.
( 1 ) 〜 (1 0 ) のようにして選抜した、 新規でかつ完全長である可能性が 高いと判断された NT- 2細胞由来の cDNAクローンの一例として、 cDNAクローン C- NT2RP2000329の塩基配列を配列番号: 1に示した。 また全長塩基配列から推定 された cDNAクローン C - NT2RP2000329がコ一ドする遺伝子産物のァミノ酸配列 を配列番号: 2に示した。 As an example of a cDNA clone derived from NT-2 cells selected as described in (1) to (10) and determined to be highly likely to be new and full-length, cDNA clone C- The nucleotide sequence of NT2RP2000329 is shown in SEQ ID NO: 1. SEQ ID NO: 2 shows the amino acid sequence of the gene product encoded by cDNA clone C-NT2RP2000329 estimated from the full-length nucleotide sequence.
[実施例 3 ] C- NT2RP2000329の機能解析 [Example 3] Function analysis of C-NT2RP2000329
C- NT2RP2000329 (配列番号: 1 ) をインハウスの GenBankデーターべ一ス BL ASTプログラムを用いて検索した。 その結果、 C_ NT2RP2000329は、 図 1に示す ように、 文献 (Shahjahan M. et al., (1991) Gene 107 : 313' - 317) に報告されて いるゥシの細胞の核内で作られるミ トコンドリアの GTP結合蛋白質 AMPホスホト ランスフェラーゼ (AK3) のアミノ酸配列に類似度 92%を示した。 また、 図 2に 示すように、 文献 (Tanabe T. et al. , ( 1993) J. Biochem. 113 : 200-207) に報 告されているラッ 卜の細胞の核内で作られるミ トコンドリアの GTP結合蛋白質 A MPホスホトランスフェラ一ゼ (AK3) のアミノ酸配列に類似度 90%を示した。 ま た、 図 3に示すように、 文献 (Xu G. et al. , (1992) Genet ics 13 : 537 - 542) に 報告されているヒ トの細胞の核内で作られるミ トコンドリアの GTP結合蛋白質 A MPホスホトランスフェラーゼ (AK3) のアミノ酸配列に類似度 60%を示した。 こ のように C- NT2RP2000329は、 ゥシおよびラッ 卜の AK3と類似度が 90%以上であ つたのに対し、 ヒ トの AK3とは類似度が 60%にすぎなかった。 従って、 C- NT2R P2000329は、 従来のものとは異なった、 新しいヒ ト遺伝子である。 産業上の利用の可能性  C-NT2RP2000329 (SEQ ID NO: 1) was searched using the in-house GenBank database BLAST program. As a result, as shown in Fig. 1, C_NT2RP2000329 was produced in the nucleus of the cell of the moss as reported in the literature (Shahjahan M. et al., (1991) Gene 107: 313'-317). It showed 92% similarity to the amino acid sequence of the AMP phosphotransferase (AK3), a GTP-binding protein from tochondria. In addition, as shown in Figure 2, mitochondria produced in the nuclei of rat cells reported in the literature (Tanabe T. et al., (1993) J. Biochem. 113: 200-207). The amino acid sequence of GTP-binding protein AMP phosphotransferase (AK3) showed 90% similarity. In addition, as shown in Figure 3, GTP binding of mitochondria produced in the nucleus of human cells reported in the literature (Xu G. et al., (1992) Genetics 13: 537-542). The amino acid sequence of protein AMP phosphotransferase (AK3) showed 60% similarity. Thus, C-NT2RP2000329 had a similarity of 90% or more with AK3 of human and rat, but only 60% of similarity with human AK3. Therefore, C-NT2R P2000329 is a new and different human gene. Industrial applicability
本発明により、 新規な AK3様蛋白質 (C- NT2RP2000329) 、 当該蛋白質をコード する遺伝子、 当該遺伝子を含むベクター、 当該ベクターを含む形質転換体、 当 該蛋白質の製造方法が提供された。 本発明の cDNAは、 細胞外情報の細胞内情報 への変換、 細胞の形態変化、 アポト一シス、 細胞運動、 細胞内物質輸送など異 常に起因する疾患に関連していることが示唆される。 このため、 本発明の遺伝 子あるいは蛋白質は、 これら疾患の診断マーカーや医薬品の開発等に有効であ る。 The present invention provides a novel AK3-like protein (C-NT2RP2000329), a gene encoding the protein, a vector containing the gene, a transformant containing the vector, and a method for producing the protein. It is suggested that the cDNA of the present invention is associated with abnormally caused diseases such as conversion of extracellular information into intracellular information, change in cell morphology, apoptosis, cell motility, and intracellular substance transport. Therefore, the genetics of the present invention Children or proteins are effective for the development of diagnostic markers and pharmaceuticals for these diseases.

Claims

請求の範囲 The scope of the claims
1 . 下記 (a ) から (d ) のいずれかに記載の DNA。 1. The DNA according to any one of (a) to (d) below.
( a ) 配列番号: 2に記載のアミノ酸配列からなる蛋白質をコードする DNA。 (a) DNA encoding a protein consisting of the amino acid sequence of SEQ ID NO: 2.
( b ) 配列番号: 1に記載の塩基配列のコ一ド領域を含む DNA。 (b) DNA containing the coding region of the nucleotide sequence of SEQ ID NO: 1.
( c ) 配列番号: 2に記載のアミノ酸配列において 1若しくは複数のアミノ酸 が置換、 欠失、 挿入、 および/または付加したアミノ酸配列を有し、 配列番号 (c) one or more amino acids in the amino acid sequence of SEQ ID NO: 2 having substitution, deletion, insertion, and / or addition of an amino acid sequence;
: 2に記載のアミノ酸配列からなる蛋白質と機能的に同等な蛋白質をコードす る DNA。 : DNA encoding a protein functionally equivalent to the protein consisting of the amino acid sequence of 2.
( d )配列番号: 1に記載の塩基配列からなる DNAとス トリンジェントな条件下 でハイブリダィズし、 配列番号: 2に記載のアミノ酸配列からなる蛋白質と機 能的に同等な蛋白質をコ一ドする DNA。  (d) hybridizing with a DNA consisting of the nucleotide sequence of SEQ ID NO: 1 under stringent conditions, and encoding a protein functionally equivalent to the protein consisting of the amino acid sequence of SEQ ID NO: 2; DNA.
2 . 配列番号: 2に記載のアミノ酸配列からなる蛋白質の部分ペプチドをコ ―ドする DNA。  2. A DNA encoding a partial peptide of a protein consisting of the amino acid sequence of SEQ ID NO: 2.
3 . 請求項 1に記載の DNAによりコードされる蛋白質またはべプチド。  3. A protein or peptide encoded by the DNA of claim 1.
4 . 請求項 1に記載の DNAが挿入されたべクター。  4. A vector into which the DNA according to claim 1 has been inserted.
5 . 請求項 1に記載の DNAまたは請求項 4に記載のベクターを保持する形質転 換細胞。  5. A transformed cell carrying the DNA according to claim 1 or the vector according to claim 4.
6 . 請求項 5に記載の形質転換細胞を培養し、 該形質転換細胞またはその培 養上清から発現させた蛋白質またはべプチドを回収する工程を含む、 請求項 3 に記載の蛋白質またはべプチドの製造方法。  6. The protein or peptide according to claim 3, comprising a step of culturing the transformed cell according to claim 5, and recovering the expressed protein or peptide from the transformed cell or a culture supernatant thereof. Manufacturing method.
7 . 請求項 3に記載の蛋白質に結合する抗体。  7. An antibody that binds to the protein of claim 3.
8 . 配列番号: 1に記載の塩基配列からなる DNAまたはその相補鎖に相補的な 少なくとも 15ヌクレオチドを含むポリヌクレオチド。  8. A polynucleotide comprising at least 15 nucleotides complementary to DNA consisting of the nucleotide sequence of SEQ ID NO: 1 or a complementary strand thereof.
9 . 請求項 3に記載の蛋白質に結合する化合物のスクリーニング方法であつ て、 (a) 該蛋白質またはその部分べプチドに被検試料を接触させる工程、 9. A method for screening a compound that binds to a protein according to claim 3, wherein (a) contacting a test sample with the protein or a partial peptide thereof,
(b) 該蛋白質またはその部分べプチドと被検試料との結合活性を検出するェ 程、  (b) detecting the binding activity between the protein or its partial peptide and a test sample,
(c) 該蛋白質またはその部分べプチドに結合する活性を有する化合物を選択 する工程、 を含む方法。  (c) selecting a compound having an activity of binding to the protein or a partial peptide thereof.
1 0. 請求項 1に記載の DNAの異常に起因する疾患を診断する方法であって、 10. A method for diagnosing a disease caused by a DNA abnormality according to claim 1,
(a) 患者から細胞試料を調製する工程、 および (a) preparing a cell sample from a patient; and
(b) 該細胞における請求項 1に記載の DNAの発現量または変異を検出するェ 程、 を含む方法。  (b) detecting the expression level or mutation of the DNA according to claim 1 in the cell.
1 1. 請求項 1に記載の DNAの異常に起因する疾患の予防または治療のため の薬剤の候補化合物をスクリーニングする方法であって、  1 1. A method for screening a candidate compound for a drug for preventing or treating a disease caused by DNA abnormality according to claim 1,
(a) 請求項 1に記載の DNAが発現している細胞に被検試料を接触させる工程  (a) contacting a test sample with cells expressing the DNA of claim 1
(b) 該細胞における請求項 1に記載の DNAの発現量を測定する工程、 および(b) measuring the expression level of the DNA according to claim 1 in the cells, and
(c) 被検試料を接触させない場合と比較して、 工程 (b) において測定され る請求項 1に記載の DNA発現量を増加または低下させる化合物を選択する工程 、 を含む方法。 (c) selecting a compound that increases or decreases the DNA expression level according to claim 1, which is measured in step (b), as compared with a case where the test sample is not contacted.
1 2. 請求項 1若しくは 2に記載の DNA、請求項 3に記載の蛋白質若しくはぺ プチド、 請求項 4に記載のベクタ一、 または請求項 9若しくは 1 1に記載のス クリーニングにより単離される化合物を含有する医薬組成物。  1 2. The DNA according to claim 1 or 2, the protein or peptide according to claim 3, the vector according to claim 4, or the compound isolated by the screening according to claim 9 or 11. A pharmaceutical composition comprising
1 3. 請求項 1若しくは 2に記載の DNAまたは請求項 8に記載のポリヌクレ ォチドが配置された DNAチップ。  1 3. A DNA chip on which the DNA according to claim 1 or 2 or the polynucleotide according to claim 8 is arranged.
PCT/JP2000/005066 1999-07-29 2000-07-28 Gene encoding novel adenylate kinase 3 (ak3)-like protein WO2001009346A1 (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
JP11/248036 1999-07-29
JP24803699 1999-07-29
JP11/300253 1999-08-27
JP30025399 1999-08-27
US15959099P 1999-10-18 1999-10-18
US60/159,590 1999-10-18
JP2000118776 2000-01-11
JP2000/118776 2000-01-11
US18332200P 2000-02-17 2000-02-17
US60/183,32220000217 2000-02-17

Publications (1)

Publication Number Publication Date
WO2001009346A1 true WO2001009346A1 (en) 2001-02-08

Family

ID=27530161

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/JP2000/005066 WO2001009346A1 (en) 1999-07-29 2000-07-28 Gene encoding novel adenylate kinase 3 (ak3)-like protein
PCT/JP2000/005067 WO2001009320A1 (en) 1999-07-29 2000-07-28 Gene encoding novel adp-ribosylation factor (arf)-like protein

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/JP2000/005067 WO2001009320A1 (en) 1999-07-29 2000-07-28 Gene encoding novel adp-ribosylation factor (arf)-like protein

Country Status (1)

Country Link
WO (2) WO2001009346A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102009005912A1 (en) * 2009-01-23 2010-07-29 Sponeta Gmbh Arrangement for locking table tennis table during playing and transportation, has snap-arm connected with guide part by pivot joint, where snap-arm is engaged in openings of guide rail in end positions of plate halves
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003289859A1 (en) * 2002-11-11 2004-06-03 Evotec Neurosciences Gmbh Diagnostic and therapeutic use of arl7 for alzheimer's disease
CN100368435C (en) * 2006-02-21 2008-02-13 中国科学技术大学 Arf6 activator protein and its coding gene and application
CN111662912A (en) * 2020-06-01 2020-09-15 云南省烟草农业科学研究院 Tobacco NtARF6 gene mutant and molecular identification method and application

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998044124A1 (en) * 1997-03-31 1998-10-08 Incyte Pharmaceuticals, Inc. Human mitochondrial adenylate kinase similar to gtp:amp phosphotransferase derivable from bovine or rat

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998044124A1 (en) * 1997-03-31 1998-10-08 Incyte Pharmaceuticals, Inc. Human mitochondrial adenylate kinase similar to gtp:amp phosphotransferase derivable from bovine or rat

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
SHAHJAHAN M. ET AL.: "Cloning and characterization of the gene encoding bovine mitochondrial adenylate kinase isozyme 3", GENE, vol. 107, no. 2, 1991, pages 313 - 317, XP002933133 *
TOMASSELLI A.G. ET AL.: "The complete primary structure of GTP:AMP phosphotransferase from beef heart mitochondria", FEBS LETT., vol. 202, no. 2, 1986, pages 303 - 308, XP002933134 *
YAMADA M. ET AL.: "Cloning and characterization of cDNA for mitochondrial GTP:AMP phosphotransferase of bovine liver", J. BIOL. CHEM., vol. 264, no. 32, 1989, pages 19192 - 19199, XP002933132 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9920123B2 (en) 2008-12-09 2018-03-20 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
DE102009005912A1 (en) * 2009-01-23 2010-07-29 Sponeta Gmbh Arrangement for locking table tennis table during playing and transportation, has snap-arm connected with guide part by pivot joint, where snap-arm is engaged in openings of guide rail in end positions of plate halves

Also Published As

Publication number Publication date
WO2001009320A1 (en) 2001-02-08

Similar Documents

Publication Publication Date Title
US7129338B1 (en) Secretory protein or membrane protein
US20070082345A1 (en) Secretory protein or membrane protein
US20020187472A1 (en) Steap-related protein
US6602667B1 (en) Inflammation-associated polynucleotides
JP2002507124A (en) Human extracellular matrix protein
US6262247B1 (en) Polycyclic aromatic hydrocarbon induced molecules
US20030166903A1 (en) Genes associated with vascular disease
US20130243781A1 (en) Signal peptide-containing proteins
WO2001009346A1 (en) Gene encoding novel adenylate kinase 3 (ak3)-like protein
US6566066B1 (en) Aquaporin-8 variant
JP2003521215A (en) 83 human secreted proteins
US6590089B1 (en) RVP-1 variant differentially expressed in Crohn's disease
JP2004527240A (en) Polynucleotides useful for regulating the growth of cancer cells
US20030175754A1 (en) RVP-1 variant differentially expressed in crohns disease
JP2000308488A (en) Protein which is related to blood vessel neogenesis and gene coding for the protein
JP2002171977A (en) New human sh2 protein
CA2486583C (en) Marker molecules associated with lung tumors
US20030104418A1 (en) Diagnostic markers for breast cancer
US20030175787A1 (en) Vesicle membrane proteins
JP2000300263A (en) Proteins '410' and 'new' involved in blood vessel neogenesis, and genes coding for the proteins
WO2001009322A1 (en) Guanosine triphosphate-binding protein coupled receptors, genes thereof and production and use of the same
US20030087253A1 (en) Polynucleotide markers for ovarian cancer
MXPA01012717A (en) Gene expression modulated in gastrointestinal inflammation.
US20030129655A1 (en) Nucleic acids encoding GTPase activating proteins
US20020137166A1 (en) ASIP-related proteins

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): JP KR US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
WA Withdrawal of international application