WO2000042180A1 - Novel protein - Google Patents

Novel protein Download PDF

Info

Publication number
WO2000042180A1
WO2000042180A1 PCT/JP2000/000106 JP0000106W WO0042180A1 WO 2000042180 A1 WO2000042180 A1 WO 2000042180A1 JP 0000106 W JP0000106 W JP 0000106W WO 0042180 A1 WO0042180 A1 WO 0042180A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
dna
oligonucleotide
seq
gene encoding
Prior art date
Application number
PCT/JP2000/000106
Other languages
French (fr)
Japanese (ja)
Inventor
Yoji Yamada
Satoshi Nakagawa
Susumu Sekine
Original Assignee
Kyowa Hakko Kogyo Co., Ltd.
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 Kyowa Hakko Kogyo Co., Ltd. filed Critical Kyowa Hakko Kogyo Co., Ltd.
Priority to AU20025/00A priority Critical patent/AU2002500A/en
Publication of WO2000042180A1 publication Critical patent/WO2000042180A1/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

Definitions

  • the present invention relates to a protein having ligand-binding ability and DNA-binding ability useful as a medicament and a diagnostic agent, DNA encoding the protein, and an antibody recognizing the protein.
  • Estrogen receptor glucocorticoid receptor, androgen receptor, progesterone receptor, thyroid hormone receptor, vitamin D receptor, electrolyte corticoid receptor, retinoic acid Receptu Yuichi, 9-cis retinoic acid Receptu Yuichi, etc. are known, and the existence of subtypes of some of them is known.
  • nuclear receptors have well-preserved functional regions such as DNA-binding region, ligand-binding region, and transcription control region, and are known to form a superfamily.
  • nuclear receptors are involved in a large number of diseases, ligands that bind to nuclear receptors have been used as therapeutics for various diseases.
  • Representative examples of therapeutic agents using ligands that bind to nuclear receptors include anti-inflammatory drugs containing glucocorticoid.
  • Glucocorticoid has a catabolic effect and works to maintain blood glucose, but another important effect is to prevent the body from responding excessively to external stimuli. Inflammation is a disease caused by this excessive response.
  • Glucocorticoid has been shown to bind to glucocorticoid drecept, thereby exhibiting a medicinal effect of suppressing the inflammatory response, and has been used for a long time in therapy.
  • nuclear receptors are useful as target molecules for detection of various diseases and screening of drugs. Disclosure of the invention
  • An object of the present invention is to provide a protein having ligand-binding ability and DNA-binding ability useful for the search and development of therapeutic agents for inflammation, cancer, osteoporosis, diabetes, kidney disease, etc., a DNA encoding the protein,
  • An object of the present invention is to provide an antibody that recognizes a protein, and a method for using the same.
  • the present invention relates to the following (1) to (41).
  • DNA that is contained in the plasmid contained in Escherichia coli DH5 / pERR4 (FERM BP-6613) and encodes a protein having ligand binding ability and DNA binding ability.
  • Transformant c obtained by introducing the recombinant vector according to (8) into a host cell (10) The transformant according to (9) is cultured in a medium, the protein according to any one of (1) to (3) is produced and accumulated in the culture, and the protein is collected from the culture. And a method for producing a protein.
  • oligonucleotide corresponding to a sequence consisting of 5 to 60 consecutive nucleotides in the base sequence of the DNA according to any one of (4) to (7) or an oligonucleotide corresponding to a sequence complementary to the oligonucleotide. nucleotide.
  • (16) A method for detecting a mutation in the gene encoding the protein according to any one of (1) to (3) by using the oligonucleotide according to (12) by a polymerase 'chain reaction method.
  • a diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease comprising the DNA according to any one of (4) to (7).
  • a diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease comprising the oligonucleotide according to (12).
  • a therapeutic agent for inflammation, cancer, osteoporosis, diabetes or a sick disease comprising the DNA according to any one of (4) to (7).
  • a therapeutic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease comprising the oligonucleotide according to (12).
  • a diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease comprising the antibody according to (11).
  • a therapeutic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease comprising the antibody according to (11).
  • the protein of the present invention includes progesterone, androgen, vitamin D, glucocorticoid, electrolyte corticoid, etc., steroid hormones, estrogen, thyroid hormone, retinoic acid, 9-cis retinoin, or ligands such as agonist and anginoist. It is a protein that has the ability to bind to DNA (hereinafter abbreviated as ligand binding ability) and the ability to bind to DNA by having a zinc finger structure (hereinafter abbreviated as DNA binding ability).
  • Examples of the protein of the present invention include a protein having an amino acid sequence represented by SEQ ID NO: 1 (hereinafter, the protein may be referred to as ERR4), and one or more amino acids are deleted in the amino acid sequence encoding the protein.
  • ERR4 an amino acid sequence represented by SEQ ID NO: 1
  • a protein comprising a substituted or added amino acid sequence and having the ligand binding ability and the DNA binding ability of the protein, the amino acid sequence represented by SEQ ID NO: 1, and BLAST CJ-Mol. Biol., 215, 403 (1990)) and FASTA [Methods in Enzymology, 183, 63-98 (1990)], etc. contain an amino acid sequence with 96% or more homology, and have ligand binding ability and DN.
  • a protein having A-binding ability can be used.
  • a protein comprising an amino acid sequence in which one or more amino acids have been deleted, substituted or added in the amino acid sequence represented by SEQ ID NO: 1 and which has the ligand-binding ability and the DNA-binding ability of the protein, Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1989) (Moleki Yura's' Cloning Second Edition '), Current Protocols in Molecular Biology, John Wiley & Sons (1987-1997) (hereinafter abbreviated as'Current'Protocols' in 'Mole Kiura Biology') Nucleic Acids Research, 10, 6487 (1982), Proc. Natl. Acad.
  • site-specific mutagenesis method for example, site-specific to DNA encoding a protein having an amino acid sequence represented by SEQ ID NO: 1. Can be obtained by introducing a target mutation.
  • the number of amino acids to be deleted, substituted or added is not particularly limited.
  • the number is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 5.
  • Examples of the DNA of the present invention include a DNA encoding the protein of the present invention.
  • a DNA having the nucleotide sequence of nucleotides 140 to 1438 of SEQ ID NO: 2 and stringent conditions with the DNA
  • Examples of the DNA include a DNA which hybridizes under the following conditions and encodes a protein having a ligand binding ability and a DNA binding ability.
  • DNA that can be hybridized under stringent conditions is defined as DNA that has a nucleotide sequence represented by nucleotides 140 to 1438 in SEQ ID NO: 2 as a probe, and is used as a probe for hybridization and plaque.
  • DNA obtained by using the hybridization method, Southern plot, hybridization method, etc., specifically, using a filter on which DNA derived from colonies or plaques is immobilized. After performing hybridization at 65 ° C in the presence of 0.7 mol of sodium chloride and 0.1 mol of SSC solution, the concentration of SSC solution of 0.1 to 2 times DNA that can be identified by washing the filter at 65 ° C under the conditions of 150 mmol / 1 sodium chloride and 15 mmol / 1 sodium citrate).
  • hybridizable DNA when calculated using analysis software such as BLAST or FAST A, it has at least 90% homology with the nucleotide sequence represented by nucleotides 140 to 1438 in SEQ ID NO: 2.
  • DNA preferably DNA having 95% or more homology.
  • Methods for preparing total RNA from human tissues include guanidine thiocyanate-cesium trifluoroacetate method [Methods in Enzymology, 154, 3 (1987)], guanidine acid thiocyanate phenol-clonal form (AGPC) Method (Analytical Biochemistry, 162, 156 (1987), experimental medicine,, 1937 (1991)).
  • Methods for preparing mRNA as po 1 y (A) + RNA from total RNA include the oligo (dT) -immobilized cellulose column method (Molecular Cloning 2nd Edition) and the method using oligo dT latex [Cell Engineering Separate Volume 8 "New Cell Engineering Experimental Protocol” Shujunsha, pp. 48-52, Nucleic Acids Res., Symposium Series, 19, 61 (1988)].
  • Fast Track mRNA Isolation Kit Fast Track mRNA Isolation Kit; Fast Track mRNA Isolation Kit; manufactured by Invitrogen
  • Quick Prep mRNA Purification Kit Quick Prep mRNA Purification Kit
  • Pharmacia It can also be used to prepare mRNA directly from tissues or cells.
  • mRNA can be used.
  • Commercially available products include, for example, human retinal mRNA from Clontech.
  • a cDNA library is prepared from the prepared mRNA, for example, human retinal mRNA.
  • Methods for preparing a cDNA library include Molecular 'Cloning 2nd Edition, Current 'Protocols' in 'Molecular' Biology, A Laboratory Manual, 2nd Ed. (1989) N DN ACloning 1: Core Techniques, A Practical Approach, Second Edition, Oxford University Press (1995), etc. Or a method using a commercially available kit, for example, Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning (manufactured by Life Technologies), ZAP-cDNA Synthesis Kit (manufactured by Stratagene), or the like.
  • a phage vector, a plasmid vector, or the like can be used as a closing vector for preparing a cDNA library as long as it can replicate autonomously in the E. coli K12 strain.
  • any microorganism belonging to Escherichia coli can be used. Specifically, Escherichia coli XLl-Blue MRF, [Strategene Gene, Strategies 5, 81 (1992)], Escherichia coli C600 Genetics, 39, 440 (1954)], Escherichia coli Y1088CScience, 222, 778 (1983) ) ⁇ Escherichia coli Y1090 (Science, 222, 778 (1983)), Escherichia coli NM522 (J. Mol. Biol., 166, 1 (1983)), Escherichia coli K802 (J. Mol.
  • a commercially available cDNA library can also be used.
  • Examples of commercially available cDNA libraries include cDNA libraries of various organs derived from human, mouse, mouse, rat, egret, etc., such as Clothtec and Lifetech Oriental. Each clone was isolated from the prepared cDNA library, and the nucleotide sequence of the cDNA for each clone was determined from the end using a commonly used nucleotide sequence analysis method, for example, the dideoxy method of Sanger et al. [Proc. Natl. Acad. Sci. USA, 74, 5463 (1977)) or a nucleotide sequence analyzer such as 373A DNA Sequencer (Perkin Elmer) is used to determine the nucleotide sequence of the DNA.
  • a commonly used nucleotide sequence analysis method for example, the dideoxy method of Sanger et al. [Proc. Natl. Acad. Sci. USA, 74, 5463 (1977)
  • a nucleotide sequence analyzer such as 373A DNA Sequ
  • nucleotide sequence of each cDNA is a novel sequence can be determined on a data base by searching base sequence databases such as GenBank, EMBL and DD BJ using a homology search program such as BLAST. This can be confirmed by the fact that there is no nucleotide sequence showing obvious homology that seems to match the nucleotide sequence of the existing gene.
  • the nucleotide sequence of the novel DNA obtained by the above method includes, for example, the nucleotide sequence of SEQ ID NO: 2.
  • a protein consisting of the amino acid sequence represented by SEQ ID NO: 1 (ERR 4), which encodes a DNA consisting of the nucleotide sequence represented by nucleotides 140 to 1438 of SEQ ID NO: 2, is a human estrogen-related receptor. ) Are 95.6% and 56.9%, respectively, with hE RR2 and hERR l CNature, 331, 91-94 (1988)].
  • the region from the cysteine residue at position 103 to the methionine residue at position 168 is hERR2 and 100%, hERRl and 91%, and hER (human estrogen receptor). And 73% homology, respectively.
  • the region consisting of the amino acid sequence corresponding to hER is a DNA binding region having a zinc finger structure, and this region is known to be a highly homologous region in nuclear receptors. Also in ERR4, the positions and numbers of cysteine residues essential for forming a characteristic finger structure in the region are completely conserved.
  • the region from the proline residue at position 208 to the residue at position 433 is 94% in 11 £ 1 ⁇ 2, 63% in 11 £ 1 ⁇ 1 and 63% in hER.
  • the region consisting of the corresponding amino acid sequence of hER is regarded as the ligand binding region, and this region corresponds to the nuclear receptor family. It is known that the region is highly homogenous.
  • ERR4 has ligand binding ability and DNA binding ability.
  • ERR 4 shows high homology with hERR 2 but low homology with hERR 1 and hER. It is known that the N-terminal region of the nuclear receptor is a transcription control region, and that this region has relatively low homology between families.
  • the amino acid sequence of ERR4 has a unique structure in the nuclear receptor, such as a DNA binding region, a ligand binding region, and a transcription control region.
  • primers based on the nucleotide sequences at the 5 ′ and 3 ′ ends of the nucleotide sequence are used.
  • a cDNA or cDNA library prepared from the mRNA contained in tissues or cells of the retina, placenta, etc. of human or non-human animals using the PCR method (PCR Protocols, Academic Press (1990))
  • the DNA of the present invention can be obtained by performing DNA amplification.
  • cDNA or cDNA synthesized from mRNA contained in tissues or cells of human or non-human animal retina, placenta, etc. as a probe using the full length or a part of DNA represented by SEQ ID NO: 2 as a probe can be obtained by performing colony hybridization or plaque hybridization (molecular cloning second edition) on the library.
  • the DNA of the present invention is chemically synthesized using a DNA synthesizer such as Perkin 'Elma's DNA synthesizer model 392 using the phosphoramidite method. You can also get it.
  • a DNA synthesizer such as Perkin 'Elma's DNA synthesizer model 392 using the phosphoramidite method. You can also get it.
  • the obtained DNA is expressed by using a transformant obtained by introducing a recombinant vector containing the DNA into a host cell, or by expressing the amino acid sequence encoded by the DNA with hERR. 2. By comparing the homology with the amino acid sequence of hERR1 or hER, it can be confirmed that the DNA is a DNA encoding a protein having ligand-binding ability and DNA-binding ability.
  • an antisense having a partial sequence of the DNA of the present invention can be obtained by a conventional method or the above-mentioned DNA synthesizer.
  • Oligonucleotides such as oligonucleotides and sense oligonucleotides can be prepared. Oligonucleotides containing RNA can also be prepared.
  • oligonucleotide examples include DNA having the same sequence as the consecutive 5 to 60 bases in the nucleotide sequence of DNA, and DNA having a sequence complementary to the DNA.
  • RNA having a sequence complementary to these DNAs is also an oligonucleotide of the present invention.
  • oligonucleotide examples include DNA having the same sequence as 5 to 60 consecutive nucleotides in the nucleotide sequence shown in SEQ ID NO: 2 or DNA having a sequence complementary to the DNA.
  • Tm melting temperature
  • Specific examples include oligonucleotides having the nucleotide sequences shown in SEQ ID NOs: 3 to 6.
  • derivatives of these oligonucleotides hereinafter, referred to as oligonucleotide derivatives can also be used as the oligonucleotide of the present invention.
  • Oligonucleotide derivatives include oligonucleotide derivatives in which phosphodiester bonds in the oligonucleotide have been converted to phosphorothioate bonds, and phosphodiester bonds in the oligonucleotides having N3,1-P5, phosphoamidates.
  • the protein of the present invention can be prepared by the method described in Molecular Cloning, Second Edition, Current Protocol—Luzin, Molecular Biology, etc., for example, by the following method. Can be produced by expressing it in a host cell.
  • a DNA fragment of an appropriate length containing a portion encoding the protein is prepared. Further, by substituting the base sequence of the portion encoding the protein such that the base sequence becomes an optimal codon for expression in the host, the production rate of the protein can be improved.
  • a recombinant vector is prepared by inserting the DNA fragment or full-length cDNA into the downstream of a promoter in an appropriate expression vector.
  • a transformant producing the protein of the present invention By introducing the recombinant vector into a host cell suitable for the expression vector, a transformant producing the protein of the present invention can be obtained.
  • any cells that can express the target gene such as bacteria, yeast, animal cells, insect cells, and plant cells, can be used.
  • Expression vectors which are capable of autonomous replication in the above host cells or capable of being integrated into a chromosome, and which contain a promoter at a position where DNA encoding the protein of the present invention can be transcribed are used. Used.
  • the recombinant vector containing the DNA encoding the protein of the present invention can replicate autonomously in the prokaryote
  • the vector is preferably composed of a ribosome binding sequence, the ERR4 gene, a transcription termination sequence, and the like.
  • a gene that controls the promoter may be included.
  • expression vectors include pBTrp2, pBTac and pBTac2 (all commercially available from Behringer Garmanheim), PKK233-2 (Pharmacia), pSE280 (Invitrogen), pGEMEX-1 (Promega), pQE-8 (manufactured by QIAGEN), pKYPIO (Japanese Patent Publication No. 58-110600), pKYP200 CAgric. Biol. Chem., 48, 669 (1984)], pLSAl CAgric. Biol. Chem., 53, 277 (1989)] , PGELl [Proc. Natl. Acad. Sci.
  • the promoter may be any promoter that can be expressed in host cells such as Escherichia coli and Bacillus subtilis.
  • ⁇ Promo Isseki one Pfrp
  • lac promoter one P lac
  • P L promoter P L promoter
  • P R promoter Isseki one promoter derived from the P SE promoter Isseki one such as, Escherichia coli, phage, etc.
  • a promoter in which two P; ⁇ £ are connected in series P ⁇ £ x 2
  • a promoter that has been artificially designed and modified such as Promo Yuichi, lacT7 promoter, let I Promo Yuichi, etc. I can do it.
  • a plasmid in which the distance between the Shine-Dalgarno sequence, which is a ribosome binding sequence, and the initiation codon is adjusted to an appropriate distance (for example, 6 to 18 bases).
  • a transcription termination sequence is not necessarily required for expression of the DNA of the present invention, but it is preferable to arrange a transcription termination sequence immediately below a structural gene.
  • Host cells include microorganisms belonging to the genus Escherichia, Serratia, Bacillus, Brevibacterium, Corynebacterium, Microbacterium, Pseudomonas, etc., for example, Escherichia coli XLl-Blue Escherichia coli XL2-Blue XLEscherichia coli DH1, Escherichia coli MC1000, Escherichia coli KY3276, Escherichia coli W1485, Escherichia coli JM109 N Escherichia coli HB101, Escherichia coli No.49, Escherichia coli W3110, Escherichia coli NY49, Serratia ficaria, Serratia fonticola, Serratia liquefaciens N Serratia marcescens-, Bacillus subtilis, Bacillus amyloliquefaciens Brevibacterium ammoniagenes, Brevibacterium immariophilum ATCC
  • any method for introducing a recombinant vector any method can be used as long as it is a method for introducing DNA into the above host cells, and for example, a method using calcium ions [Pr0c.
  • YEpl3 ATCC37115
  • YEp24 ATCC37051
  • YCp50 ATCC37419
  • pHS19 pHS15 and the like
  • Any promoter can be used as long as it can be expressed in yeast strains.
  • PH05 promoter PGK promoter, GAP promoter, ADH promoter, gal 1 promoter, gal 10 Promoters such as promoter, heat shock polypeptide promoter, MF promoter and CUP promoter can be mentioned.
  • Saccharomyces cerevisiae yeast strains belonging to the genera Saccharomyces, Schizosaccharomyces, Kluybium mycetes, Trichosporone, Schizinomyces, Pichia, and the like.
  • any method can be used as long as it is a method for introducing DNA into yeast.
  • electroporation Methods in Enzymology, 194, 182 (1990)
  • spheroplast Law [p r0 c. Natl. Acad. Sci. USA, 81, 4889 (1984) ]
  • the lithium acetate method [Journal of Bacteriology, 153, 163 ( 1983) ] Proc. Natl. Acad. Sci . USA, 75, 1929 (1978).
  • examples of expression vectors include pcDNAI, pcDM8 (commercially available from Funakoshi), pAGE107 [JP-A-3-22979; Cytotechnology, 3, 133, (1990)], pAS3 -3 (JP-A-2-227075), pCDM8 CNature, 329, 840, (1987)], pcDNAI / Amp (manufactured by Invitrogen), REP4 (manufactured by Invitrogen), pAGE103 (J. Biochemistry, 101, 1307 (1987)) ], PAGE210, pAMo, pA oA, etc. Cut.
  • Any promoter can be used as long as it can be expressed in animal cells.
  • promoters include the cytomegalovirus (CMV) IE (immediate early) gene promoter, the SV40 early promoter, and the retro promoter.
  • CMV cytomegalovirus
  • Examples include virus promoters, meta-mouth choonein promoter, heat shock promoters, and SR promoter promoters.
  • the enhancer of the IE gene of human CMV may be used together with the promoter.
  • Host cells include mouse / myeloma cells, rat 'myeloma cells, mouse' hybridoma cells, Namalwa cells or Namalwa KJM-1 cells, human embryonic kidney cells, human leukemia cells, and African green monkeys. Kidney cells, Chinese hamster cells such as CH0 cells, HBT5637 (Japanese Patent Application Laid-Open No. 63-299), and the like.
  • Mouse / myeloma cells include SP2 / 0 and NS0, rat myeloma cells include YB2 / 0, etc., human fetal kidney cells include HEK293 (ATCC: CRL-1573), and human leukemia cells. Examples include BALL-1 and the like, and African green monkey kidney cells include COS-1 and COS-7.
  • any method can be used as long as it is a method for introducing DNA into animal cells.
  • baculovirus expression When an insect cell is used as a host, for example, baculovirus expression, vector sacrifice, lacto-radiation, etc. can be performed using Baculovirus Expression Vectors, A Laboratory Manual, WH Freeman and Company, New York (1992). )] ⁇ ⁇ Molecular Biology, Laboratory Laboratory Manual (Molecular Biology, A Laboratory Manual), Current 'Protocols' in' Molecular Noology, Bio / Technology, 6, 47 (1988), etc. Depending on the method, the protein can be expressed.
  • the recombinant gene transfer vector and the baculovirus are co-transfected into insect cells to obtain the recombinant virus in the culture supernatant of the insect cells, and then the recombinant virus is transferred to the insect cell. It can infect insect cells and express proteins.
  • Examples of the gene transfer vector used in the method include pVL1392, pVL1393, pBlueBacII (all manufactured by Invitorogen) and the like.
  • the baculovirus for example, as 0 insect cells that can use Rereru a burglar Gaka insects is a virus that infects out publicly available, power Nuclear one poly to Doroshisu virus (Autographa californica nuclear polyhedrosis virus) and the like
  • Doroshisu virus Autographa californica nuclear polyhedrosis virus
  • the ovarian cells of Spodoptera frugiperda, the ovarian cells of Tricoplia nia, and the cultured cells derived from the chick ovary can be used.
  • the ovarian cells of Spodoptera frugiperda include Sf9 and Sf21 (baculovirus expression 'vectors' and 'laboratory' manuals), and the like.
  • the ovarian cells of Trichoplusia include High 5 and BTI-TN-5B1-4 (manufactured by Invitrogen). Bombyx mori N4 and the like can be mentioned as cultured cells derived from ovaries.
  • Methods for co-transferring the above-described recombinant gene transfer vector and the baculovirus into insect cells to prepare a recombinant virus include, for example, a calcium phosphate method (Japanese Patent Laid-Open No. 2-227075), a lipofection method [Proc. Natl. Acad. Sci. USA, 84, 7413 (1987)].
  • examples of the expression vector include Ti plasmid and tobacco mosaic virus vector.
  • Any promoter can be used as long as it can be expressed in plant cells, and examples thereof include the 35S promoter of cauliflower mosaic virus (CaMV) and the geneactin 1 promoter.
  • CaMV cauliflower mosaic virus
  • Examples of the host cell include plant cells of tobacco, potato, tomato, carrot, soybean, abrana, alfa alfa, rice, wheat, wheat, and the like.
  • Any method for introducing a recombinant vector can be used so long as it is a method for introducing DNA into plant cells.
  • Agrobacteriim Agrobacteriim
  • an electroporation method Japanese Patent Publication No. 60-251887
  • a method using a particle gun Japanese Patent No. 2517813
  • Japanese Patent No. 2517813 Japanese Patent No. 2517813
  • a protein having a sugar chain or a sugar chain When expressed by yeast, animal cells, insect cells, or plant cells, a protein having a sugar chain or a sugar chain can be obtained.
  • the protein of the present invention can be produced by culturing the transformant obtained as described above in a medium, producing and accumulating the protein of the present invention in the culture, and collecting from the culture.
  • the method for culturing the transformant of the present invention in a medium can be performed according to a usual method used for culturing a host.
  • a culture medium for culturing a transformant obtained by using a prokaryote such as Escherichia coli or a eukaryote such as yeast as a host contains a carbon source, a nitrogen source, inorganic salts, and the like which can be used by the organism. Either a natural medium or a synthetic medium can be used as long as the medium can efficiently culture the cells.
  • the carbon source may be any one that can be assimilated by the organism; glucose, fructose, sucrose, molasses containing these, carbohydrates such as starch or starch hydrolysate, organic acids such as acetic acid and propionic acid, Alcohols such as ethanol and propanol can be used.
  • Nitrogen sources include ammonia, ammonium chloride, ammonium sulfate, ammonium acetate, ammonium phosphate and other ammonium-containing salts of inorganic or organic acids, other nitrogen-containing compounds, peptone, meat extract, yeast extract, corn steep liquor, Casein hydrolyzate, soybean meal and soybean meal hydrolyzate, various fermented bacterial cells and digests thereof can be used.
  • potassium (II) phosphate potassium (II) phosphate, magnesium phosphate, magnesium sulfate, sodium chloride, ferrous sulfate, man sulfate, copper sulfate, calcium carbonate, and the like can be used.
  • the cultivation is usually carried out under aerobic conditions such as shaking culture or deep aeration stirring culture.
  • the culture temperature is preferably 15 to 40 ° C, and the culture time is usually 16 hours to 7 days.
  • the pH during cultivation is maintained at 3.0 to 9.0.
  • the pH is adjusted using an inorganic or organic acid, an alkaline solution, urea, calcium carbonate, ammonia, or the like.
  • antibiotics such as ampicillin and tetracycline may be used during culture, if necessary. It may be added to the medium.
  • an inducer may be added to the medium, if necessary.
  • an inducer may be added to the medium, if necessary.
  • an inducer may be added to the medium.
  • indoleacrylic acid or the like may be added to the medium.
  • Culture is carried out usually p H 6 ⁇ 8, 2 5 ⁇ 4 0 ° 5% C_ ⁇ 2 under the conditions such as the presence 1-7 days.
  • antibiotics such as kanamycin, penicillin and streptomycin may be added to the medium during the culturing.
  • Culture media for transformants obtained using insect cells as a host include commonly used TNM-FH medium (Pharmingen), Sf-900 II SFM medium (Life Technologies), ExCell400, ExCe 11405 (all manufactured by JRH Biosciences), Grace's Insect Medium CGrace, TCC, Nature, 195, 788 (1962)] and the like can be used.
  • the cultivation is usually performed under conditions of pH 6 to 7 and 25 to 30 ° C for 1 to 5 days.
  • an antibiotic such as gentamicin may be added to the medium during the culture.
  • a transformant obtained using a plant cell as a host can be cultured as a cell or after being differentiated into a plant cell or organ.
  • a medium for culturing the transformant commonly used Murashige 'and' squeeg (MS) medium, white (White) medium, or a plant hormone such as auxin or cytokinin was added to these mediums.
  • MS Murashige 'and' squeeg
  • White white
  • a plant hormone such as auxin or cytokinin
  • Culture is usually performed at pH 5 to 9, 20 to 40 ° C for 3 to 60 days. If necessary, antibiotics such as kanamycin and hygromycin may be added to the medium during the culture.
  • a transformant derived from a microorganism, animal cell, or plant cell having the recombinant vector into which the DNA encoding the protein of the present invention is incorporated is cultured according to a usual culture method, and the protein By producing and accumulating, and collecting the protein from the culture, the protein can be produced.
  • a method for expressing a gene in addition to direct expression, secretory production, fusion protein expression, and the like can be performed according to the method described in Molecular Cloning, 2nd edition, and the like.
  • Methods for producing the protein of the present invention include a method of producing the protein in a host cell, a method of secreting the protein out of the host cell, and a method of producing the protein on the host cell outer membrane. The method can be selected by changing the structure.
  • the protein of the present invention is expressed in a form in which a signal peptide is added in front of the protein containing the active site of the protein of the present invention using a gene recombination technique, so that the protein of the present invention can be positively extracellularly outside the host cells. It can be secreted.
  • the production amount can be increased by using a gene amplification system using a dihydrofolate reductase gene or the like.
  • the transgenic animal or plant cells are redifferentiated to create an animal (transgenic non-human animal) or plant (transgenic plant) into which the gene has been introduced.
  • the protein of the present invention can also be produced using an individual.
  • the protein When the transformant is an individual animal or plant, the protein is raised or cultivated according to a usual method to produce and accumulate the protein, and the protein is collected from the individual animal or plant to obtain the protein. Can be manufactured.
  • Methods for producing the protein of the present invention using animal individuals include, for example, known methods American Journal of Clinical Nutrition, 63, 639S (1996), American Journal of Clinical Nutrition, 63, 627S (1996), Bio / Technology, 9, 830 (1991)] to produce the protein of the present invention in an animal constructed by introducing a gene.
  • the protein is produced and accumulated in the animal, and the protein is collected from the animal. By doing so, the protein can be produced.
  • the place of production and accumulation in the animal include milk (eg, JP-A-63-309192) and eggs of the animal.
  • the promoter used at this time any promoter that can be expressed in an animal can be used.
  • the casein promoter-1 and the casein promoter-1 which are breast cell-specific promoters can be used.
  • ⁇ -lactoglobulin promoter, whey acidic protein promoter, etc. are preferably used.
  • a transgenic plant into which a DNA encoding the protein of the present invention has been introduced can be prepared by a known method [tissue culture, 20 (1994); (1995), Trends in Biotechnology, 15, 45 (1997)], producing and accumulating the protein in the plant, and collecting the protein from the plant to produce the protein. There is a method.
  • the cells are collected by centrifugation after completion of the culture, suspended in an aqueous buffer, and then sonicated with a sonicator, french press, Mentongaurin. Crush cells with a homogenizer, dynomill, etc. to obtain a cell-free extract.
  • an ordinary enzyme isolation method that is, a solvent extraction method, a salting out method using ammonium sulfate, a desalting method, a precipitation method using an organic solvent, Getylaminoethyl (DEAE)-Sepharose, DIAI0N HPA-75 (Mitsubishi Chemical), anion exchange chromatography using resin, S-Sepharose FF
  • the cells When the protein is expressed in an insoluble form in the cells, the cells are similarly collected, crushed, and centrifuged to obtain a precipitate fraction obtained by a conventional method. After recovering the protein, the insoluble form of the protein is solubilized with a protein denaturant.
  • a purified sample can be obtained by the same isolation and purification method.
  • the protein of the present invention or a derivative such as a modified sugar thereof is secreted extracellularly, the protein or a derivative such as a sugar chain adduct thereof can be recovered in the culture supernatant.
  • a soluble fraction is obtained by treating the culture by a method such as centrifugation as described above, and a purified sample is obtained from the soluble fraction by using the same isolation and purification method as described above. Can be obtained.
  • Examples of the protein obtained in this manner include a protein having the amino acid sequence shown in SEQ ID NO: 1.
  • the polypeptide of the present invention can be produced as a fusion protein with another protein, and purified using affinity chromatography using a substance having an affinity for the fused protein. Natl. Acad. Sci. USA, 86, 8227 (1989), Genes Develop., 4, 1288 (1990), and the method described in JP-A-05-336963, W094 / 23021.
  • the polypeptide of the present invention can be produced as a fusion protein with protein A, and purified by affinity chromatography using immunoglobulin G.
  • the protein of the present invention is obtained by chemical synthesis methods such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the tBoc method (t-butyloxycarbonyl method). Can be manufactured. Chemical synthesis can also be carried out using a peptide synthesizer such as Advanced ChemTech, Nokin-Kenolema, Pharmacia, Protein Technology Instrument, Synthecell-Vega, PerSeptive, Shimadzu.
  • a peptide synthesizer such as Advanced ChemTech, Nokin-Kenolema, Pharmacia, Protein Technology Instrument, Synthecell-Vega, PerSeptive, Shimadzu.
  • the polypeptide is administered to an animal to give a polypeptide antibody.
  • a purified preparation of the protein of the present invention or a polypeptide fragment of the partial fragment of the protein, or a peptide having an amino acid sequence of a part of the protein of the present invention as an antigen the polypeptide is administered to an animal to give a polypeptide antibody. Can be made.
  • Animals such as rabbits, goats, rats, mice, hamsters and the like can be used for the administration.
  • the dose of the antigen is preferably 50 to 100 ⁇ g per animal.
  • a peptide When a peptide is used, it is desirable to use, as the antigen, a peptide covalently bonded to a carrier protein such as keyhole 1 impet haemocyanin or bovine tilogin purine.
  • a carrier protein such as keyhole 1 impet haemocyanin or bovine tilogin purine.
  • the peptide used as the antigen can also be synthesized using a peptide synthesizer.
  • the administration of the antigen is performed 3 to 10 times every 1 to 2 weeks after the first administration. Blood is collected from the fundus venous plexus on the 3rd to 7th days after each administration, and the reaction of the serum with the antigen used for immunization is determined by an enzyme immunoassay [enzyme immunoassay (ELISA): published by Medical Shoin (19) 76 years), Antibodies-A Laboratory Manual, Cold Spring Harbor Laboratory (1988)].
  • enzyme immunoassay enzyme immunoassay
  • Serum is obtained from a non-human mammal whose serum shows a sufficient antibody titer against the antigen used for immunization, and a polyclonal antibody can be separated and purified from the serum by the following method.
  • Methods for separating and purifying antibodies include centrifugation, salting out with 40-50% saturated ammonium sulfate, and prillic acid precipitation [Antibodies, A Laboratory manual, Cold Spring Harbor Laboratory, (1988)], or DEAE. —Sepharose column
  • Examples of the method include a method in which chromatography using an anion exchange column, a protein A-column, a protein G-column, a gel filtration column, or the like is used alone or in combination.
  • a rat whose serum shows a sufficient antibody titer against the antigen used for immunization is used as a source of antibody-producing cells.
  • the spleen is removed.
  • the spleen is shredded in a MEM medium (manufactured by Nissui Pharmaceutical Co., Ltd.), loosened with forceps, centrifuged at 1,200 rpm for 5 minutes, and the supernatant is discarded.
  • MEM medium manufactured by Nissui Pharmaceutical Co., Ltd.
  • the spleen cells in the resulting precipitate fraction are treated with Tris-ammonium chloride buffer (pH 7.65) for 1 to 2 minutes to remove red blood cells, and then washed three times with MEM medium. Used as
  • myeloma cells cell lines obtained from mice or rats are used.
  • 8-azaguanine-resistant mouse derived from BALB / c
  • myeloma cell line P3-X63Ag8-U1 (hereinafter abbreviated as P3-U1) [Curr. Topics. Microbiol. Immunol., 81, 1 (1978) , Europ.J. Immunol., 6, 511 (1976)), SP2 / 0-Agl4 (SP-2) CNature, 276, 269 (1978)), P3-X63-Ag8653 (653) (J.
  • the cells in the precipitate fraction obtained are gently sprinkled, and then slowly sucked and blown out with a female pipette.
  • the HAT medium in a normal medium, hypoxanthine (1 (T 4 mol / l), thymidine (1.5) x 10- 5 mol / l) and suspended aminopterin (4x 10-3 ⁇ 4ol eight) to the pressurizing example was medium] in 100 ml.
  • the suspension was dispensed by 100 ⁇ 1 / well to the plate for a 96-well culture, 5% C0 2 incuba- - evening - in cultured for 7-14 days at 37 ° C.
  • the partial fragment polypeptide of the protein of the present invention used as the antigen was coated on an appropriate plate, and reacted with the hybridoma culture supernatant or the purified antibody obtained in (d) described below as the first antibody. Further, after reacting an anti-rat or anti-mouse immunoglobulin antibody labeled with a biotin, an enzyme, a chemiluminescent substance or a radioactive compound as a second antibody, a reaction according to the labeling substance is carried out, and the protein of the present invention is reacted. Those that react specifically are selected as hybridomas producing the monoclonal antibody of the present invention.
  • hybridoma Using the hybridoma, cloning was repeated twice by the limiting dilution method (the first time was HT medium (medium obtained by removing aminopterin from the HAT medium), and the second time was A normal medium is used], and those with a stable and high antibody titer are selected as hybridoma strains producing the monoclonal antibody of the present invention.
  • Pristane-treated [0.5 ml of 2,6,10,14-tetramethylpenedecane (Pristane) was intraperitoneally administered and bred for 2 weeks].
  • c) Intraperitoneally inject 5 to 20 ⁇ 10 6 cells of the hybridoma cells producing the monoclonal antibody of the present invention obtained in the step c). In 10 to 21 days, Hypridoma develops ascites cancer.
  • the ascites is collected from the mouse with ascites tumor and centrifuged at 3000 rpm for 5 minutes to remove solids.
  • a monoclonal antibody can be purified and obtained in the same manner as the method used for the polyclonal antibody.
  • the antibody subclass is determined using a mouse monoclonal antibody evening kit or a rat monoclonal antibody typing kit.
  • the protein content is calculated by the Lowry method or from the absorbance at 280 nm.
  • An antibody class refers to an antibody isotype, and in humans, includes IgG, IgA, IgM, IgD, and IgE.
  • the subclass refers to an isotype within the class.
  • IgGK IgG2a ⁇ IgG2b, IgG3, and in a human, IgGl, IgG2, IgG3, and IgG4.
  • the RT-PCR method is a simple method, and is particularly useful as a detection method.
  • the detection method is also used for quantifying the expression level of a gene, and is used for inflammation, cancer, osteoporosis, It can be used for diagnosis of diseases caused by mutation of the gene encoding the protein of the present invention, such as diabetes and kidney disease.
  • the gene encoding the protein of the present invention can be obtained. Mutations can be detected.
  • the detection method can be used for diagnosis of diseases caused by mutation of the gene encoding the protein of the present invention, such as inflammation, cancer, osteoporosis, diabetes, and kidney disease.
  • the DNA of the present invention can be prepared by antisense RNA / DNA technology [Bioscience and Industry, 50, 322 (1992), Chemistry, 46, 681 (1991), Biotechnology, 9, 358 (1992), Trends in Biotechnology, 10, 87 (1992), Trends in Biotechnology, 10, 152 (1992), Cell Engineering, i £, 1463 (1997)], Triple Helix Technology !: Trends in Biotechnology, 10, 132 (1992)]
  • the transcription or translation of the gene encoding the protein of the present invention can be suppressed.
  • transcription of a gene encoding the protein of the present invention or translation of mRNA encoding the protein of the present invention can be suppressed, respectively. Production can be suppressed.
  • the method can be used for treating or preventing inflammation, cancer, osteoporosis, diabetes, kidney disease, and other diseases caused by mutations in the gene encoding the protein of the present invention.
  • a method for treating a disease caused by a mutation in the gene encoding the protein of the present invention cells obtained after introducing the recombinant vector of the present invention appropriately prepared for gene therapy into cells taken out of a patient, By returning the virus to the body, it is also possible to obtain appropriate retroviruses, adenoviruses, adeno-associated viruses, simple virus, A method for expressing the protein of the present invention in a patient's living body by administering it to a living body by carrying it on a viral vector such as an antivirus or by further encapsulating it in an artificial vesicle structure such as ribosome. Used.
  • ERR4 shows high homology with hERR2, but hERR2 may inhibit anti-inflammatory effects such as glucocorticoid (J. Biol. Chem., 271, 9879 (1996) Therefore, it is inferred that suppressing the expression of RR4 can enhance the action of anti-inflammatory substances.
  • ERR4 since ERR4 is expressed in the kidney, it can be an effective therapeutic agent for inflammation or renal diseases such as nephritis.
  • the promoter region include all promoter regions involved in transcription of a gene encoding a protein of the present invention in mammalian cells.
  • a promoter region involved in transcription of a gene encoding the protein of the present invention can be mentioned.
  • the promoter can be used for a screening method described later.
  • a method for identifying a substance that directly binds to the protein of the present invention and a method for designing a drug based on the three-dimensional structure of the protein [Charifson. PS, Practical Application of Computer-Aided Drug Design, Marcel Dekker (1997)]
  • a ligand that specifically binds to the protein can be screened.
  • the ligand can be used as a therapeutic agent for a disease associated with the protein of the present invention or a study on a signal transduction system or a biological function involving the protein of the present invention.
  • the transformant of the present invention is allowed to coexist with various test substances; A substance that controls the transcription of a gene encoding the protein of the present invention by analyzing the expression level of the offspring, a substance that participates in the transcription control function of the protein of the present invention, or a gene that is transcriptionally controlled by the protein of the present invention. Can be screened.
  • the interaction with them can be assayed, and at the same time, the interaction with these genes can be examined.
  • the drugs involved can be screened.
  • the CAT chloramphenicol acetyltransferase
  • GFP green fluorescent protein
  • the protein of the present invention or a tissue containing the protein can be immunologically detected.
  • the detection method can be used for diagnosis of diseases caused by mutation of the gene encoding the protein of the present invention, such as inflammation, cancer, osteoporosis, diabetes, and kidney disease.
  • the detection method can also be used for protein quantification.
  • Methods for immunological detection include the ELISA assay using a microtiter plate, a fluorescent antibody method, a stamp lot method, and an immunohistological staining method.
  • Methods for immunological quantification include sandwich ELISA using two types of monoclonal antibodies having different epitopes among antibodies that react with the protein of the present invention in the liquid phase, and a method of labeling with a radioisotope such as 12S I.
  • a radioimmunoassay method using the protein of the present invention and an antibody recognizing the protein of the present invention can be exemplified.
  • the antibody of the present invention can be used as a medicament, for example, a therapeutic agent for diseases caused by mutations in the gene encoding the protein of the present invention, such as inflammation, cancer, osteoporosis, diabetes, and kidney disease.
  • diseases caused by mutations in the gene encoding the protein of the present invention such as inflammation, cancer, osteoporosis, diabetes, and kidney disease.
  • the medicament containing the antibody of the present invention may be administered alone as a therapeutic agent. Although possible, it is usually admixed with one or more pharmacologically acceptable carriers to provide a pharmaceutical preparation produced by any method well known in the pharmaceutical arts. desirable.
  • Dosage forms include sprays, capsules, tablets, granules, syrups, emulsions, suppositories, injections, ointments, tapes and the like.
  • Formulations suitable for oral administration include emulsions, syrups, capsules, tablets, powders, granules and the like.
  • liquid preparations such as emulsions and syrups include water, sugars such as sucrose, sorbitol, fructose, glycols such as polyethylene glycol and propylene glycol, oils such as sesame oil, olive oil, soybean oil, p- It can be produced using preservatives such as hydroxybenzoic acid esters and flavors such as strawberry flavor and peppermint as additives.
  • preservatives such as hydroxybenzoic acid esters and flavors such as strawberry flavor and peppermint as additives.
  • excipients such as lactose, glucose, sucrose, mannitol, disintegrants such as starch, sodium alginate, lubricants such as magnesium stearate, talc, polyvinyl alcohol And a binder such as hydroxypropylcellulose and gelatin, a surfactant such as a fatty acid ester, and a plasticizer such as glycerin as additives.
  • Formulations suitable for parenteral administration include injections, suppositories, sprays and the like.
  • an injection is prepared using a carrier comprising a salt solution, a glucose solution, or a mixture of both.
  • Suppositories are prepared using carriers such as cocoa butter, hydrogenated fats or carboxylic acids.
  • Sprays are prepared using the compound itself or a carrier which does not irritate the oral and respiratory mucosa of the recipient and which disperses the compound as fine particles to facilitate absorption.
  • Specific examples of the carrier include lactose, glycerin and the like.
  • Formulations such as aerosols and dry powders are possible depending on the properties of the compound and the carrier used.
  • the components exemplified as additives for oral preparations can also be added.
  • the dose or frequency of administration varies depending on the desired therapeutic effect, administration method, treatment period, age, body weight, etc., but is usually 10 / g / kg to 8 mg / kg per day for an adult.
  • an objective non-human animal for example, an embryonic stem cell such as a sea lion, a sheep, a goat, a bush, a horse, a mouse, a chicken, etc.
  • an embryonic stem cell such as a sea lion, a sheep, a goat, a bush, a horse, a mouse, a chicken, etc.
  • the gene encoding the protein of the present invention on the chromosome is mutated by a known homologous recombination technique (eg, Nature, 326, 6110, 295 (1987), Cell, 51, 3, 503 (1987), etc.).
  • a mutant clone is prepared which is activated or substituted with an arbitrary sequence [for example, Nature, 350, 6315, 243 (1991)].
  • a chimeric individual consisting of an embryonic stem cell clone and normal cells is prepared by a method such as chimera method or chimera method in which an embryo fertilized egg of an animal is injected into a blastocyst (blastcyst). be able to.
  • an individual having an arbitrary mutation in the gene encoding the protein of the present invention on the chromosome of the whole body can be obtained.
  • a knockout non-human animal can be obtained as an individual in which the expression of the gene encoding the protein of the present invention is partially or completely suppressed from homozygous individuals in which both have mutations.
  • a knockout non-human animal by introducing a mutation into an arbitrary position of the gene encoding the protein of the present invention on the chromosome, a knockout non-human animal can be produced.
  • the activity of the product can be modified by introducing mutations by substituting, deleting, or inserting bases into the translation region of the gene encoding the protein of the present invention on the chromosome.
  • introducing a similar mutation into the expression control region it is possible to modify the degree, timing, tissue specificity, etc. of the expression.
  • Examples of such cases include the use of a promoter that is expressed in a specific region of the brain and deletion of the target gene only in that region (Cell, 87, 7, 1317 (1996)) or expression of Cre.
  • a promoter that is expressed in a specific region of the brain and deletion of the target gene only in that region (Cell, 87, 7, 1317 (1996)) or expression of Cre.
  • Cre expression of Cre.
  • a target gene is specifically deleted at an intended time using an adenovirus [Science, 278, 5335 (1997)].
  • the expression of the gene encoding the protein of the present invention on the chromosome can be controlled at any time or in any tissue, or the gene has any insertion, deletion, or substitution in its translation region or expression control region.
  • Knockout non-human animals can be produced. The knockout non-human animal can induce the symptoms of various diseases caused by the protein of the present invention at any time, any degree, or any site.
  • the knockout non-human animal of the present invention becomes an extremely useful animal model in the treatment and prevention of various diseases caused by the protein of the present invention.
  • it is very useful as a model for evaluating therapeutic drugs, preventive drugs, functional foods, and health foods.
  • FIG. 1 shows the results of agarose electrophoresis of RT-PCR products for single-stranded cDNA of human tissue.
  • Lane M is a marker of human / EcoT14I
  • lanes 1 to 10 are for heart, brain, placenta, lung, liver, skeletal muscle, kidney, renal, spleen, and thymus, respectively. This is an RT-PCR product obtained by converting cDNA into type II.
  • Lane C is the result of RT-PCR using pERR4 plasmid DNA as type III.
  • H82543 and H91842 show a high homology of 94.1%, and H91890 and H82542 show a high homology of 96.3%, which are considered to be almost identical.
  • human ERR2 with H82543 and human 1 ⁇ 12 with 1191842, it showed high homology at the 5 and 5 ends of human ERR2, with values of 84.1% and 86.0%, respectively.
  • human E The deletion of H82543 and H91842 from 82 bp to 85 bp of the RR 2 nucleotide sequence suggests that this EST group is part of a different gene from ERR2.
  • this EST group encodes a part of a different gene from ERR2, and the gene was named ERR4. It is thought that H82543 and H91842 represent the nucleotide sequence near the 5 'end of ERR4, and H82542 and H91890 represent the nucleotide sequence at the 3' side.
  • a Human Retina 5'-STRETCH cDNA library from Clontech was used according to the origin of the EST clone.
  • E. coli C600Hfl manufactured by Clontech
  • E. coli C600Hfl manufactured by Clontech
  • the solution was plated on a 150 mm Petri dish with pAgar.
  • the primer ERR4-01 corresponds to 84 bp to 107 bp of the EST sequence H82543, and the primer ERM-03 corresponds to 70 bp to 93 bp of the EST sequence H91890.
  • PCR was performed using the cDNA library fraction as type II.
  • the fraction amplified by the PCR was plated on a 15 Omm petri dish so as to generate about 30,000 plaques per sheet.
  • the resulting plaque phage was transferred onto a Hybond-N + membrane (manufactured by Amersham), and the plaque was hybridized to the membrane.
  • the probe used was digoxigenin labeled using the PCRDIG probe synthesis kit (Boehringer Mannheim) based on the primers ERR4-01 and EM4-03.
  • the reaction was performed using a hybridization solution (5 XSSC (75 Ommol / 1 sodium chloride, ⁇ 5 ol / l sodium citrate (pH 7.0)), 1.0% nucleic acid hybridization booking reagent (Boehringer Mannheim) in 0.1% N-lauroyl sarcosine, 0.02% SDS] at 65 ° C.
  • the filter was washed twice with 0.1% SSC containing 0.1% 803 at 68 ° C for 15 minutes, and then alkaline phosphatase-conjugated anti-DIG
  • the antibody is treated with a DIG luminescence detection kit (Boehringer Mannheim) comprising CSPD and a chemiluminescent film (Hyperfilm-ECL;
  • the plaque corresponding to the detected signal was suspended in SM buffer and again plated on a Petri dish so that a single plaque could be isolated.
  • a single plaque with a signal was suspended in an SM buffer, and phage DNA was isolated therefrom using a Lambda Midi Kit (QIAGEN).
  • the library used here uses the input gtlO vector, and the cDNA of the clone should have been inserted into the EcoRI site.Therefore, the phage DNA was cut with EcoRI, and the insert DNA was also converted into pBluescript. II SK (+) (manufactured by STRATAGENE) was ligated with a vector cut with Ec0RI and subcloned.
  • the nucleotide sequence from the 5 'end of cDNA of the clone obtained in Example 1 was determined using DNA Sequencer-377 of Perkin Pharma Co., Ltd.
  • the nucleotide sequence was determined using a kit from PerkinElmer Inc. and following the instructions for the kit. The homology of the determined nucleotide sequence to the nucleotide sequence of a known gene in the nucleotide sequence database GenBank was analyzed using the analysis program BLAST. Did not match.
  • ERR4 the protein encoded by this clone was designated as ERR4.
  • SEQ ID NO: 1 shows the amino acid sequence of ERR4 and SEQ ID NO: 2 shows its nucleotide sequence.
  • the research laboratory has been deposited at Tsukuba Tsukuba, Ibaraki, Japan, 1-3-1, Higashi (postal code 305-8566).
  • Each cDNA was synthesized using a Preamplification System (GIBC BRL). The specific procedure was as described in this kit, and dissolved in a final volume of 1 ml of water.
  • GIBC BRL Preamplification System
  • the solution was diluted 5-fold, and RT-PCR was performed using 5.0: 1 of the diluted solution.
  • the primer can be amplified in the region containing the open 'reading' frame of ERR4, and is a primer having the nucleotide sequence shown in SEQ ID NO: 5, which is an ERR4-specific sequence. Two types of sequencer, Bramer ERM-06, were used.
  • ERR4-07 corresponds to 807 bp to 829 bp of the nucleotide sequence of SEQ ID NO: 2
  • ERR4-06 corresponds to the complementary strand of 1470 bp to 1493 bp of the nucleotide sequence of SEQ ID NO: 2.
  • One cycle of denaturation at 94 ° C for 1 minute, annealing at 62 ° C for 1 minute, and extension at 72 ° C for 2 minutes was defined as one cycle, and the cycle was repeated 35 times to perform PCR. On the other hand, agarose gel electrophoresis was performed.
  • a protein having ligand-binding ability and DNA-binding ability useful for search and development of therapeutic agents for inflammation, cancer, osteoporosis, diabetes, kidney disease, etc., DNA encoding the protein, and the protein An antibody recognizing, and a method of using these can be provided.

Abstract

A protein having the amino acid sequence represented by SEQ ID N° 1 which has an ability to bind to a ligand and an ability to bind to a DNA and is useful in searching and developing remedies for inflammation, cancer, osteoporosis, diabetes, kidney diseases, etc.; a DNA encoding this protein; a recombinant vector containing this DNA; a transformant obtained by transferring this recombinant vector into host cells; an oligonucleotide corresponding to a part of the sequence of the above DNA; an antibody recognizing the above protein; and utilization thereof.

Description

新規蛋白質 技術分野  New protein technology
本発明は、 医薬、 診断薬として有用なリガンド結合能および D N A結合能を有 する蛋白質、 該蛋白質をコードする D N Aおよび該蛋白質を認識する抗体に関す る。  The present invention relates to a protein having ligand-binding ability and DNA-binding ability useful as a medicament and a diagnostic agent, DNA encoding the protein, and an antibody recognizing the protein.
明:  Ming:
 Fine
景技  Skill
核内レセプ夕一としては、 エストロゲンレセプ夕一、 グルココルチコイ ドレセ プター、 アンドロゲンレセプ夕一、 プロゲステロンレセプ夕一、 甲状腺ホルモン レセプ夕一、 ビタミン Dレセプ夕一、 電解質コルチコイ ドレセプ夕一、 レチノィ ン酸レセプ夕一、 9-cisレチノイン酸レセプ夕一等が知られており、 これらの一部 のものについてはそのサブタイプの存在が知られている。  Estrogen receptor, glucocorticoid receptor, androgen receptor, progesterone receptor, thyroid hormone receptor, vitamin D receptor, electrolyte corticoid receptor, retinoic acid Receptu Yuichi, 9-cis retinoic acid Receptu Yuichi, etc. are known, and the existence of subtypes of some of them is known.
これらの核内レセプ夕一は、 D N A結合領域、 リガンド結合領域、 転写制御領 域等の機能領域がよく保存されており、 スーパーファミリ一を形成して L、ること が知られている 〔核内レセプ夕一と情報伝達、加藤茂明著、 羊土社( 1 9 9 4年)〕 < 核内レセプ夕一は、 特定のリガンドが結合することにより、 標的遺伝子の転写を 活性化し、 種々の生体反応を発現させることが知られている。  These nuclear receptors have well-preserved functional regions such as DNA-binding region, ligand-binding region, and transcription control region, and are known to form a superfamily. Information communication with inner receptor Yuichi, Shigeaki Kato, Yodosha (1994)] <Nuclear receptor Yuichi activates the transcription of target genes by binding to specific ligands, It is known to develop a biological response.
核内レセプ夕一は極めて多数の疾患に関与するため、 核内レセプタ一と結合す るリガンドは種々の疾患の治療薬として用いられている。  Since nuclear receptors are involved in a large number of diseases, ligands that bind to nuclear receptors have been used as therapeutics for various diseases.
核内レセプ夕一と結合するリガンドを用いた治療薬の代表例としては、 グルコ コルチコィ ドを含有する抗炎症剤があげられる。 グルココルチコィ ドは異化作用 を持ち、 血糖の維持に働くが、 別の重要な作用として、 外界刺激に対して生体が 過剰な応答を示すことを抑制する。 炎症はこの過剰な応答による疾患であり、 グ ルココルチコィ ドはグルココルチコィ ドレセプ夕一と結合することにより、 炎症 反応を抑制するという薬効を示し、 古くから治療に用いられている。  Representative examples of therapeutic agents using ligands that bind to nuclear receptors include anti-inflammatory drugs containing glucocorticoid. Glucocorticoid has a catabolic effect and works to maintain blood glucose, but another important effect is to prevent the body from responding excessively to external stimuli. Inflammation is a disease caused by this excessive response. Glucocorticoid has been shown to bind to glucocorticoid drecept, thereby exhibiting a medicinal effect of suppressing the inflammatory response, and has been used for a long time in therapy.
またレチノイン酸レセプ夕一のリガンドであるレチノィン酸を急性前骨髄性白 血病患者に投与することにより、顕著に治療成績が上がったとする報告!; Blood, , 567-572, ( 1988)〕 がある。 さらに、 骨粗鬆症に対するエス トロゲンの作用もェ ストロゲンレセプ夕一を介するものである。 In addition, it was reported that administration of retinoic acid, a ligand for retinoic acid receptor, to patients with acute promyelocytic leukemia significantly improved the treatment results! Blood, 567-572, (1988)]. In addition, estrogen's effect on osteoporosis is It is through the strogen receptor Yuichi.
このように核内レセプ夕一は、 種々の疾患の検出や薬剤のスクリ一ニングのた めの標的分子として有用である。 発明の開示  Thus, nuclear receptors are useful as target molecules for detection of various diseases and screening of drugs. Disclosure of the invention
本発明の目的は、 炎症、 癌、 骨粗鬆症、 糖尿病、 腎疾患等の治療薬の探索、 開 発に有用なリガンド結合能および DN A結合能を有する蛋白質、 該蛋白質をコ一 ドする DNA、 該蛋白質を認識する抗体、 およびこれらの利用方法を提供するこ とにある。  An object of the present invention is to provide a protein having ligand-binding ability and DNA-binding ability useful for the search and development of therapeutic agents for inflammation, cancer, osteoporosis, diabetes, kidney disease, etc., a DNA encoding the protein, An object of the present invention is to provide an antibody that recognizes a protein, and a method for using the same.
本発明は、 以下の (1) ~ (41) に関する。  The present invention relates to the following (1) to (41).
( 1 ) 配列番号 1で示されるアミノ酸配列を有する蛋白質。  (1) A protein having the amino acid sequence represented by SEQ ID NO: 1.
( 2 ) 配列番号 1で示されるアミノ酸配列において 1以上のァミノ酸が欠失、 置換または付加されたアミノ酸配列からなり、 かつ配列番号 1で示されるァミノ 酸配列からなる蛋白質の有するリガンド結合能および DN A結合能を有する蛋白 質。  (2) a protein comprising an amino acid sequence represented by SEQ ID NO: 1 in which one or more amino acids have been deleted, substituted or added, and a protein comprising the amino acid sequence represented by SEQ ID NO: 1; and A protein having DNA binding ability.
( 3 ) 配列番号 1で示されるアミノ酸配列と 96 %以上の相同性を有するアミ ノ酸配列を含み、 かつ配列番号 1で示されるァミノ酸配列からなる蛋白質の有す るリガンド結合能および DN A結合能を有する蛋白質。  (3) Ligand-binding ability and DNA of a protein comprising an amino acid sequence having 96% or more homology with the amino acid sequence represented by SEQ ID NO: 1 and comprising the amino acid sequence represented by SEQ ID NO: 1 A protein having binding ability.
(4) (1) 〜 (3) のいずれかに記載の蛋白質をコードする DNA。  (4) A DNA encoding the protein according to any one of (1) to (3).
(5) 配列番号 2の 140〜1438番目の塩基配列を有する DNA。  (5) a DNA having the nucleotide sequence of positions 140 to 1438 of SEQ ID NO: 2;
(6) (4) または (5) 記載の DN Aとストリンジェン卜な条件下でハイブ リダイズし、 配列番号 2の 140〜 1438番目の塩基配列と 90 %以上の相同 性を有し、 かつ配列番号 1で示されるアミノ酸配列からなる蛋白質の有するリガ ンド結合能および DNA結合能を有する蛋白質をコードする DNA。  (6) It hybridizes with the DNA described in (4) or (5) under stringent conditions, has 90% or more homology with the nucleotide sequence at positions 140 to 1438 of SEQ ID NO: 2, and has a sequence A DNA encoding a protein having the ligand-binding ability and the DNA-binding ability of the protein having the amino acid sequence represented by No. 1.
(7) Escherichia coli DH5ひ/ pERR4 (FERM BP— 6613) に含有さ れるプラスミ ドに含まれ、 かつリガンド結合能および DN A結合能を有する蛋白 質をコードする DNA。  (7) DNA that is contained in the plasmid contained in Escherichia coli DH5 / pERR4 (FERM BP-6613) and encodes a protein having ligand binding ability and DNA binding ability.
(8) (4) 〜 (7) のいずれかに記載の DNAを含有してなる組換えべクタ  (8) A recombinant vector containing the DNA according to any one of (4) to (7).
(9) (8)記載の組換えべクタ一を宿主細胞に導入して得られる形質転換体 c (10) (9)記載の形質転換体を培地に培養し、 培養物中に ( 1 ) 〜 ( 3 ) のいずれかに記載の蛋白質を生成蓄積させ、 該培養物から該蛋白質を採取するこ とを特徴とする蛋白質の製造方法。 (9) Transformant c obtained by introducing the recombinant vector according to (8) into a host cell (10) The transformant according to (9) is cultured in a medium, the protein according to any one of (1) to (3) is produced and accumulated in the culture, and the protein is collected from the culture. And a method for producing a protein.
(11) ( 1 ) 〜 ( 3 ) のいずれかに記載の蛋白質を認識する抗体。  (11) An antibody that recognizes the protein according to any one of (1) to (3).
(12) (4) 〜 (7) のいずれかに記載の DNAの有する塩基配列中の連続 した 5〜 60塩基からなる配列に相当するオリゴヌクレオチドまたは該オリゴヌ クレオチドと相補的な配列に相当するオリゴヌクレオチド。  (12) An oligonucleotide corresponding to a sequence consisting of 5 to 60 consecutive nucleotides in the base sequence of the DNA according to any one of (4) to (7) or an oligonucleotide corresponding to a sequence complementary to the oligonucleotide. nucleotide.
(13) (4) 〜 (7) のいずれかに記載の DN Aを用い、 ハイブリダィゼ一 シヨン法により、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする遺伝子 の発現を検出する方法。  (13) Using the DNA described in any of (4) to (7), the expression of the gene encoding the protein described in any of (1) to (3) is detected by the hybridization method. Method.
(14) (12) のオリゴヌクレオチドを用い、 ポリメラーゼ ·チエイン - リ アクション法により、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする遺 伝子の発現を検出する方法。  (14) A method for detecting the expression of the gene encoding the protein according to any one of (1) to (3) by using the oligonucleotide of (12) by the polymerase chain reaction method.
(15) (4) 〜 (7) のいずれかに記載の DN Aを用い、 ハイブリダィゼ一 シヨン法により、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする遺伝子 の変異を検出する方法。  (15) Using the DNA described in any one of (4) to (7), detecting a mutation in the gene encoding the protein described in any one of (1) to (3) by a hybridization method. Method.
(16) (12)記載のオリゴヌクレオチドを用い、 ポリメラ一ゼ 'チェイン · リアクション法により、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする 遺伝子の変異を検出する方法。  (16) A method for detecting a mutation in the gene encoding the protein according to any one of (1) to (3) by using the oligonucleotide according to (12) by a polymerase 'chain reaction method.
(17) (13) 〜 (16) のいずれかに記載の方法を用いて炎症、 癌、 骨粗 鬆症、 糖尿病または腎疾患を検出する方法。  (17) A method for detecting inflammation, cancer, osteoporosis, diabetes or kidney disease using the method according to any one of (13) to (16).
(18) (4) 〜 (7) のいずれかに記載の DN Aを用いることを特徴とする、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする遺伝子の転写を抑制する 方法。  (18) A method for suppressing transcription of a gene encoding the protein according to any one of (1) to (3), which comprises using the DNA according to any one of (4) to (7). .
(19) (12)記載のオリゴヌクレオチドを用いることを特徴とする、 ( 1) ~ (3) のいずれかに記載の蛋白質をコードする遺伝子の転写を抑制する方法。 (19) A method for suppressing transcription of a gene encoding the protein according to any one of (1) to (3), which comprises using the oligonucleotide according to (12).
(20) (4) 〜 (7) のいずれかに記載の DNAを用いることを特徴とする、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする mRNAの翻訳を抑制す る方法。 (20) A method for suppressing translation of mRNA encoding the protein according to any one of (1) to (3), comprising using the DNA according to any one of (4) to (7). .
(21) (12) 記載のオリゴヌクレオチドを用いることを特徴とする、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする mRN Aの翻訳を抑制する方法。 (22) (4) 〜 (7) のいずれかに記載の DNAを用いることを特徴とする、 ( 1) 〜 (3) のいずれかに記載の蛋白質をコードする遺伝子のプロモーター領 域を取得する方法。 (21) Use of the oligonucleotide according to (12), (1) A method for inhibiting translation of mRNA encoding the protein according to any one of (3) to (3). (22) obtaining a promoter region of a gene encoding the protein according to any one of (1) to (3), wherein the DNA according to any one of (4) to (7) is used; Method.
(23) ( 12) 記載のオリゴヌクレオチドを用いることを特徴とする、 (1) 〜 (3) のいずれかに記載の蛋白質をコードする遺伝子のプロモーター領域を取 得する方法。  (23) A method for obtaining a promoter region of a gene encoding the protein according to any one of (1) to (3), which comprises using the oligonucleotide according to (12).
(24) (4) 〜 (7) のいずれかに記載の DN Aを含有する、 炎症、 癌、 骨 粗鬆症、 糖尿病または腎疾患の診断薬。  (24) A diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the DNA according to any one of (4) to (7).
(25) ( 12) 記載のオリゴヌクレオチドを含有する、 炎症、 癌、 骨粗鬆症、 糖尿病または腎疾患の診断薬。  (25) A diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the oligonucleotide according to (12).
(26) (4) ~ (7) のいずれかに記載の DN Aを含有する医薬。  (26) A medicament containing the DNA according to any one of (4) to (7).
(27) ( 12) 記載のオリゴヌクレオチドを含有する医薬。  (27) A medicament comprising the oligonucleotide according to (12).
(28) (4) 〜 (7) のいずれかに記載の DN Aを含有する炎症、 癌、 骨粗 鬆症、 糖尿病または賢疾患の治療薬。  (28) A therapeutic agent for inflammation, cancer, osteoporosis, diabetes or a sick disease, comprising the DNA according to any one of (4) to (7).
(29) ( 12) 記載のオリゴヌクレオチドを含有する炎症、 癌、 骨粗鬆症、 糖尿病または腎疾患の治療薬。  (29) A therapeutic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the oligonucleotide according to (12).
(30) ( 1) 〜 (3) のいずれかに記載の蛋白質を用いることを特徴とする、 該蛋白質と特異的に結合するリガンドのスクリーニング方法。  (30) A method for screening for a ligand that specifically binds to the protein according to any one of (1) to (3).
(31) (30) 記載の方法により取得されるリガンド。  (31) A ligand obtained by the method according to (30).
(32) ( 1 1) 記載の抗体を用いることを特徴とする、 ( 1) 〜 (3) のい ずれかに記載の蛋白質の免疫学的検出法。  (32) A method for immunologically detecting a protein according to any one of (1) to (3), comprising using the antibody according to (11).
(33) ( 1 1) 記載の抗体を用い、 ( 1 ) 〜 ( 3 ) のいずれかに記載の蛋白 質を検出することを特徴とする免疫組織染色法。  (33) An immunohistological staining method characterized by detecting the protein according to any one of (1) to (3) using the antibody according to (11).
(34) ( 1 1) 記載の抗体を含有する炎症、 癌、 骨粗鬆症、 糖尿病または腎 疾患の診断薬。  (34) A diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the antibody according to (11).
(35) ( 1 1) 記載の抗体を含有する医薬。  (35) A medicament containing the antibody according to (11).
(36) ( 1 1) 記載の抗体を含有する炎症、 癌、 骨粗鬆症、 糖尿病または腎 疾患の治療薬。  (36) A therapeutic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the antibody according to (11).
(37) (9) 記載の形質転換体を用いることを特徴とする、 ( 1) 〜 (3) のいずれかに記載の蛋白質をコードする遺伝子の転写を制御する物質のスクリ一 二ング方法。 (37) (1) to (3), characterized by using the transformant according to (9). A method for screening a substance that controls transcription of a gene encoding the protein according to any one of the above.
(38) (9) 記載の形質転換体を用いることを特徴とする、 ( 1) 〜 (3) のいずれかに記載の蛋白質による転写制御機能に関与する物質のスクリーニング 方法。  (38) A method for screening a substance involved in a transcriptional control function of a protein according to any one of (1) to (3), which comprises using the transformant according to (9).
(39) (9) 記載の形質転換体を用いることを特徴とする、 ( 1) 〜 (3) のいずれかに記載の蛋白質により転写制御を受ける遺伝子のスクリーニング方法。 (39) A method for screening a gene that is transcriptionally regulated by the protein according to any one of (1) to (3), comprising using the transformant according to (9).
(40) (1) 〜 (3) のいずれかに記載の蛋白質をコードする遺伝子の発現 がー部または完全に抑制されたノックァゥト非ヒト動物。 (40) A knockout non-human animal in which expression of the gene encoding the protein according to any one of (1) to (3) is partially or completely suppressed.
(41) ( 1) 〜 (3) のいずれかに記載の蛋白質の有する活性が一部または 完全に抑制されたノックアウト非ヒト動物。 本発明の蛋白質は、 プロゲステロン、 アンドロゲン、 ビタミン D、 グルココル チコィ ド、 電解質コルチコィ ド等のステロイ ドホルモン、 エストロゲン、 甲状腺 ホルモン、 レチノイン酸、 9- cisレチノイン、 またはこれらのァゴニスト、 アン夕 ゴニスト等のリガンドと結合する能力 (以下、 リガンド結合能と略す) を有し、 かつジンク ' フィンガー (zinc finger) 構造を持つことにより D N Aと結合する 能力 (以下、 DNA結合能と略す) を有する蛋白質である。  (41) A knockout non-human animal in which the activity of the protein according to any one of (1) to (3) is partially or completely suppressed. The protein of the present invention includes progesterone, androgen, vitamin D, glucocorticoid, electrolyte corticoid, etc., steroid hormones, estrogen, thyroid hormone, retinoic acid, 9-cis retinoin, or ligands such as agonist and anginoist. It is a protein that has the ability to bind to DNA (hereinafter abbreviated as ligand binding ability) and the ability to bind to DNA by having a zinc finger structure (hereinafter abbreviated as DNA binding ability).
本発明の蛋白質として、 例えば、 配列番号 1で示されるアミノ酸配列を有する 蛋白質 (以下、 該蛋白質を ERR4と呼ぶこともある) 、 該蛋白質をコードする ァミノ酸配列において 1以上のァミノ酸が欠失、 置換または付加されたアミノ酸 配列からなり、 かつ該蛋白質の有するリガンド結合能および DN A結合能を有す る蛋白質、配列番号 1で示されるアミノ酸配列と、 BLAST CJ- Mol. Biol. ,215, 403 (1990)〕 や F A S T A 〔Methods in Enzymology, 183, 63-98 (1990)〕 等を用 いて計算したときに、 96%以上の相同性を有するアミノ酸配列を含み、 かつリ ガンド結合能および DN A結合能を有する蛋白質等をあげることができる。  Examples of the protein of the present invention include a protein having an amino acid sequence represented by SEQ ID NO: 1 (hereinafter, the protein may be referred to as ERR4), and one or more amino acids are deleted in the amino acid sequence encoding the protein. A protein comprising a substituted or added amino acid sequence and having the ligand binding ability and the DNA binding ability of the protein, the amino acid sequence represented by SEQ ID NO: 1, and BLAST CJ-Mol. Biol., 215, 403 (1990)) and FASTA [Methods in Enzymology, 183, 63-98 (1990)], etc., contain an amino acid sequence with 96% or more homology, and have ligand binding ability and DN. A protein having A-binding ability can be used.
配列番号 1で示されるァミノ酸配列において 1以上のアミノ酸が欠失、 置換ま たは付加されたァミノ酸配列からなり、 かつ該蛋白質の有するリガンド結合能お よび DN A結合能を有する蛋白質は、 Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1989) (以下、 モレキ ユラ一 'クロ一ニング第 2版と略す) 、 Current Protocols in Molecular Biology, John Wiley & Sons (1987-1997) (以下、 カレント ' プロ トコ一ルズ ' イン 'モレ キユラ一 ·バイオロジーと略す) 、 Nucleic Acids Research, 10, 6487 (1982)、 Proc. Natl. Acad. Sci., USA, 79, 6409 (1982)、 Gene, 34, 315 (1985)、 Nucleic Acids Research, 13, 4431 (1985)、 Proc. Natl. Acad. Sci. USA, 82, 488 (1985) 等に記載の部位特異的変異導入法を用いて、 例えば配列番号 1で示されるァミノ 酸配列を有する蛋白質をコードする DN Aに部位特異的変異を導入することによ り取得することができる。 A protein comprising an amino acid sequence in which one or more amino acids have been deleted, substituted or added in the amino acid sequence represented by SEQ ID NO: 1 and which has the ligand-binding ability and the DNA-binding ability of the protein, Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press (1989) (Moleki Yura's' Cloning Second Edition '), Current Protocols in Molecular Biology, John Wiley & Sons (1987-1997) (hereinafter abbreviated as'Current'Protocols' in 'Mole Kiura Biology') Nucleic Acids Research, 10, 6487 (1982), Proc. Natl. Acad. Sci., USA, 79, 6409 (1982), Gene, 34, 315 (1985), Nucleic Acids Research, 13, 4431 (1985), Proc. Natl. Acad. Sci. USA, 82, 488 (1985), etc., using the site-specific mutagenesis method, for example, site-specific to DNA encoding a protein having an amino acid sequence represented by SEQ ID NO: 1. Can be obtained by introducing a target mutation.
欠失、 置換もしくは付加されるアミノ酸の数は特に限定されないが、 上記の部 位特異的変異法等の周知の方法により欠失、 置換もしくは付加できる程度の数で あり、 1個から数十個、 好ましくは 1〜20個、 より好ましくは 1〜10個、 さ らに好ましくは 1〜5個である。 また、 本発明の蛋白質が核内レセプ夕一として の機能を有するためには、 B LASTや FAS T A等の解析ソフトを用いて計算 したときに、 配列番号 1記載のアミノ酸配列と少なくとも 96%以上の相同性を 有していることが好ましい。  The number of amino acids to be deleted, substituted or added is not particularly limited. The number is preferably 1 to 20, more preferably 1 to 10, and even more preferably 1 to 5. Further, in order for the protein of the present invention to have a function as a nuclear receptor, at least 96% or more of the amino acid sequence described in SEQ ID NO: 1 when calculated using analysis software such as BLAST or FASTA. It is preferable to have the homology of
本発明の DNAとしては、 本発明の蛋白質をコ一ドする DNAをあげることが でき、 例えば、 配列番号 2の 140〜 1438番目に示される塩基配列を有する DNA、 該 DNAとストリンジェン卜な条件下でハイブリダィズし、 かつリガン ド結合能および DN A結合能を有する蛋白質をコ一ドする DN Aをあげることが できる。  Examples of the DNA of the present invention include a DNA encoding the protein of the present invention. For example, a DNA having the nucleotide sequence of nucleotides 140 to 1438 of SEQ ID NO: 2 and stringent conditions with the DNA Examples of the DNA include a DNA which hybridizes under the following conditions and encodes a protein having a ligand binding ability and a DNA binding ability.
ストリンジ工ントな条件下でハイブリダイズ可能な D N Aとは、 配列番号 2の 140〜 1438番目で表される塩基配列を有する DN Aをプローブとして、 コ ロニ一 'ハイブリダィゼーシヨン法、 プラーク 'ハイブリダィゼーシヨン法ある いはサザン ·プロット ·ハイプリダイゼーシヨン法等を用いることにより得られ る DNAを意味し、 具体的には、 コロニーあるいはプラーク由来の DNAを固定 化したフィルターを用いて、 0. 7〜 1. Omol八の塩化ナトリウム存在下、 65°C でハイブリダイゼーシヨンを行った後、 0. 1〜 2倍濃度の S S C溶液 (1倍濃 度の S S C溶液の組成は、 150 mmol/1塩化ナトリウム、 15 mmol/1 クェン 酸ナトリウムよりなる) を用い、 65 °C条件下でフィル夕一を洗浄することによ り同定できる D N Aをあげることができる。 ハイブリダィゼーシヨンは、 モレキュラー 'クロ一ニング第 2版、 カレント ' プロトコ一ルズ ·イン 'モレキュラー ·バイオロジー、 DNA Clonin 1: Core Techniques, A Practical Approach, Second Edition, Oxford University (1995) 等に記載されている方法に準じて行うことができる。 DNA that can be hybridized under stringent conditions is defined as DNA that has a nucleotide sequence represented by nucleotides 140 to 1438 in SEQ ID NO: 2 as a probe, and is used as a probe for hybridization and plaque. DNA obtained by using the hybridization method, Southern plot, hybridization method, etc., specifically, using a filter on which DNA derived from colonies or plaques is immobilized. After performing hybridization at 65 ° C in the presence of 0.7 mol of sodium chloride and 0.1 mol of SSC solution, the concentration of SSC solution of 0.1 to 2 times DNA that can be identified by washing the filter at 65 ° C under the conditions of 150 mmol / 1 sodium chloride and 15 mmol / 1 sodium citrate). Hybridization has been published in Molecular 'Clothing Second Edition, Current' Protocols in 'Molecular Biology, DNA Clonin 1: Core Techniques, A Practical Approach, Second Edition, Oxford University (1995), etc. It can be carried out according to the method described.
ハイブリダイズ可能な D N Aとして具体的には、 BLASTや FAST A等の 解析ソフトを用いて計算したときに、 配列番号 2の 140〜 1438番目で表さ れる塩基配列と少なくとも 90%以上の相同性を有する DNA、 好ましくは 9 5%以上の相同性を有する DN Aをあげることができる。  Specifically, as a hybridizable DNA, when calculated using analysis software such as BLAST or FAST A, it has at least 90% homology with the nucleotide sequence represented by nucleotides 140 to 1438 in SEQ ID NO: 2. DNA, preferably DNA having 95% or more homology.
以下、 本発明を詳細に説明する。  Hereinafter, the present invention will be described in detail.
1. 本発明の DN Aの調製  1. Preparation of DNA of the present invention
ヒ卜の組織より全 RNAあるいは mRNAを調製する。  Prepare total RNA or mRNA from human tissues.
ヒト組織から全 RN Aを調製する方法としては、 チォシアン酸グァニジン一ト リフルォロ酢酸セシウム法 〔Methods in Enzymology, 154, 3 (1987)〕 、 酸性チ オシアン酸グァニジン ·フエノール -クロ口ホルム (AG PC) 法 (Analytical Biochemistry, 162, 156 (1987)、 実験医学、 , 1937 (1991)〕 等をあげることが できる。  Methods for preparing total RNA from human tissues include guanidine thiocyanate-cesium trifluoroacetate method [Methods in Enzymology, 154, 3 (1987)], guanidine acid thiocyanate phenol-clonal form (AGPC) Method (Analytical Biochemistry, 162, 156 (1987), experimental medicine,, 1937 (1991)).
全 RNAから p o 1 y (A) +R N Aとして mR N Aを調製する方法としては、 オリゴ (dT)固定化セルロースカラム法 (モレキュラー ·クローニング第 2版)、 オリゴ dTラテックスを用いる方法 〔細胞工学 別冊 8 「新細胞工学実験プロト コール」 秀潤社 48- 52頁、 Nucleic Acids Res., Symposium Series, 19, 61 (1988)〕 等をあげることができる。 Methods for preparing mRNA as po 1 y (A) + RNA from total RNA include the oligo (dT) -immobilized cellulose column method (Molecular Cloning 2nd Edition) and the method using oligo dT latex [Cell Engineering Separate Volume 8 "New Cell Engineering Experimental Protocol" Shujunsha, pp. 48-52, Nucleic Acids Res., Symposium Series, 19, 61 (1988)].
ファース ト · トラック ' mRNA単離キット ast Track mRNA Isolation Kit; インビトロジェン ( Invitrogen) 社製〕 、 クイック ■プレップ · mR N A精製キ ット Quick Prep mRNA Purification Kit; フアルマシア (Pharmacia) 社製〕 等 のキットを用いて組織や細胞から直接 mRNAを調製することもできる。  Fast Track mRNA Isolation Kit; Fast Track mRNA Isolation Kit; manufactured by Invitrogen), Quick Prep mRNA Purification Kit Quick Prep mRNA Purification Kit; manufactured by Pharmacia It can also be used to prepare mRNA directly from tissues or cells.
また、 市販の mRNAを用いることができる。 市販のものとしては、 例えば、 Clontech社製のヒト網膜 mR N Aをあげることができる。  Also, commercially available mRNA can be used. Commercially available products include, for example, human retinal mRNA from Clontech.
調製した mRNA、 例えば、 ヒト網膜 mRNAから cDN Aライブラリ一を作 製する。  A cDNA library is prepared from the prepared mRNA, for example, human retinal mRNA.
cDNAライブラリ一作製法としては、 モレキュラー 'クローニング第 2版、 カレント 'プロトコ一ルズ 'イン 'モレキュラー 'バイオロジー、 A Laboratory Manual, 2nd Ed. (1989)N D N ACloning 1: Core Techniques, A Practical Approach, Second Edition, Oxford University Press (1995)等に記載された方法、 あるい は市販のキット、 例えば Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning (Life Technologies社製)、 ZAP-cDNA Synthesis Kit (ストラタ ジーン社製)を用いる方法等があげられる。 Methods for preparing a cDNA library include Molecular 'Cloning 2nd Edition, Current 'Protocols' in 'Molecular' Biology, A Laboratory Manual, 2nd Ed. (1989) N DN ACloning 1: Core Techniques, A Practical Approach, Second Edition, Oxford University Press (1995), etc. Or a method using a commercially available kit, for example, Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning (manufactured by Life Technologies), ZAP-cDNA Synthesis Kit (manufactured by Stratagene), or the like.
c D N Aライブラリ一を作製するためのクロ一ニングベクターとしては、 大腸 菌 K12株中で自立複製できるものであれば、 ファージベクタ一、 プラスミ ドベクタ 一等いずれも使用できる。  A phage vector, a plasmid vector, or the like can be used as a closing vector for preparing a cDNA library as long as it can replicate autonomously in the E. coli K12 strain.
具体的には、 ZAP Express 〔ストラ夕ジーン社製、 Strategies, 5, 58 (1992)〕、 pBluescript II SK (+) (Nucleic Acids Research, 17, 9494 (1989)〕、 Lambda ZAP II (ストラ夕ジーン社製)、 え gtlO、 Agtll CD N ACloning, A Practical Approach, 1, 49 (1985)〕、 ATriplEx (クローンテック社製) 、 え ExCell (フアルマシア社 製) 、 pT7T318U (フアルマシア社製) 、 pcD2 CMol. Cell. Biol., 3, 280 (1983)〕、 pUC 18 CGene, 33, 103 (1985)〕、 p AMo〔J. Biol. Chem. , 268, Z2782-22787 (1993)、 別名 pAMoPRC3Sc (特開平 05- 336963) 〕 等をあげることができる。  Specifically, ZAP Express (Strategene, Inc., Strategies, 5, 58 (1992)), pBluescript II SK (+) (Nucleic Acids Research, 17, 9494 (1989)), Lambda ZAP II (Strategene) GtlO, Agtll CDN ACloning, A Practical Approach, 1, 49 (1985)], ATriplEx (Clontech), EExCell (Pharmacia), pT7T318U (Pharmacia), pcD2 CMol. Cell. Biol., 3, 280 (1983)], pUC 18 CGene, 33, 103 (1985)], p AMo [J. Biol. Chem., 268, Z2782-22787 (1993), aka pAMoPRC3Sc (Japanese Unexamined Patent Publication No. -336963)].
宿主微生物としては、 大腸菌 Escherichia coliに属する微生物であればいずれ も用いることができる。 具体的には、 Escherichia coli XLl-Blue MRF, 〔ストラ 夕ジーン社製、 Strategies 5, 81 (1992)〕、 Escherichia coli C600 Genetics, 39, 440 ( 1954)〕、 Escherichia coli Y1088CScience, 222, 778 (1983)^ Escherichia coli Y1090 (Science, 222, 778 (1983)〕、 Escherichia coli NM522〔J. Mol. Biol., 166, 1 (1983)〕、 Escherichia coli K802 〔J. Mol. Biol., 16, 118 (1966)〕、 Escherichia coli JM105 CGene, 38, 275 (1985)〕、 Escherichia coli SOLRTM Strain (ストラ夕ジーン社製) 、 Escherichia coli LE392 (モレキュラー クロ一ニング 第 2版) 等を用いることができる。  As the host microorganism, any microorganism belonging to Escherichia coli can be used. Specifically, Escherichia coli XLl-Blue MRF, [Strategene Gene, Strategies 5, 81 (1992)], Escherichia coli C600 Genetics, 39, 440 (1954)], Escherichia coli Y1088CScience, 222, 778 (1983) ) ^ Escherichia coli Y1090 (Science, 222, 778 (1983)), Escherichia coli NM522 (J. Mol. Biol., 166, 1 (1983)), Escherichia coli K802 (J. Mol. Biol., 16, 118 ( 1966)], Escherichia coli JM105 CGene, 38, 275 (1985)], Escherichia coli SOLRTM Strain (manufactured by Straugene), Escherichia coli LE392 (Molecular Cloning 2nd Edition), and the like.
上記方法により作製した c D N Aライブラリ一に加え、 市販の c D N Aライブ ラリーも利用することができる。  In addition to the cDNA library prepared by the above method, a commercially available cDNA library can also be used.
市販の cDN Aライブラリ一として、 クロ一ンテック社、 ライフテックオリエ ン夕ル社等のヒト、 ゥシ、 マウス、 ラット、 ゥサギ等由来の各臓器 cDNAライ ブラリ一をあげることができる。 作製した cDNAライブラリーから各クローンを単離し、 それぞれのクローン について cDNAの塩基配列を末端から、 通常用いられる塩基配列解析方法、 例 えばサンガ一 (Sanger) らのジデォキシ法 〔Proc. Natl. Acad. Sci. USA, 74, 5463 (1977))あるいは 373 A · DNAシークェンサ一 (Perkin Elmer社製) 等の塩 基配列分析装置を用いて分析することにより、 該 DN Aの塩基配列を決定する。 それぞれの c D N Aの塩基配列が新規な配列かどうかは、 B L A S T等の相同 性検索プログラムを用いて、 GenBank、 EMB Lおよび DD B J等の塩基 配列データベースを検索することにより、 デ一夕ベース中の既存の遺伝子の塩基 配列と一致すると考えられるような明らかな相同性を示す塩基配列がないことに より確認できる。 Examples of commercially available cDNA libraries include cDNA libraries of various organs derived from human, mouse, mouse, rat, egret, etc., such as Clothtec and Lifetech Oriental. Each clone was isolated from the prepared cDNA library, and the nucleotide sequence of the cDNA for each clone was determined from the end using a commonly used nucleotide sequence analysis method, for example, the dideoxy method of Sanger et al. [Proc. Natl. Acad. Sci. USA, 74, 5463 (1977)) or a nucleotide sequence analyzer such as 373A DNA Sequencer (Perkin Elmer) is used to determine the nucleotide sequence of the DNA. Whether the nucleotide sequence of each cDNA is a novel sequence can be determined on a data base by searching base sequence databases such as GenBank, EMBL and DD BJ using a homology search program such as BLAST. This can be confirmed by the fact that there is no nucleotide sequence showing obvious homology that seems to match the nucleotide sequence of the existing gene.
上記の方法で得られる新規な DN Aの塩基配列として、 例えば配列番号 2で示 される塩基配列をあげることができる。  The nucleotide sequence of the novel DNA obtained by the above method includes, for example, the nucleotide sequence of SEQ ID NO: 2.
配列番号 2の 140〜 1438番目に示される塩基配列からなる DNAのコー ドする、 配列番号 1で示されるアミノ酸配列からなる蛋白質 (ERR 4) は、 ヒ トエストロゲン関連レセプ夕一 (human Estrogen Related Receptor) である hE RR2および hERR l CNature, 331, 91-94 (1988)〕 と、 それぞれ 95. 6% および 56. 9%の相同性を有する。  A protein consisting of the amino acid sequence represented by SEQ ID NO: 1 (ERR 4), which encodes a DNA consisting of the nucleotide sequence represented by nucleotides 140 to 1438 of SEQ ID NO: 2, is a human estrogen-related receptor. ) Are 95.6% and 56.9%, respectively, with hE RR2 and hERR l CNature, 331, 91-94 (1988)].
配列番号 1で示されるアミノ酸配列において、 103番目のシスティン残基〜 168番目のメチォニン残基の領域は、 hERR2と 100%、 hERRlと 9 1%、 hER 〔ヒトエス卜ロゲンレセプター (human Estrogen Receptor) 〕 と 7 3 %の相同性をそれぞれ示す。  In the amino acid sequence represented by SEQ ID NO: 1, the region from the cysteine residue at position 103 to the methionine residue at position 168 is hERR2 and 100%, hERRl and 91%, and hER (human estrogen receptor). And 73% homology, respectively.
hERの対応するアミノ酸配列からなる領域は、 ジンク . フィンガー構造を有 する DNA結合領域とされるが、 該領域は核内レセプ夕一フアミリーで相同性の 高い領域であることが知られている。 ERR 4においても、 該領域に特徴的なジ ンク 'フィンガ一構造を形成するために必須なシスティン残基の位置と数が完全 に保存されている。  The region consisting of the amino acid sequence corresponding to hER is a DNA binding region having a zinc finger structure, and this region is known to be a highly homologous region in nuclear receptors. Also in ERR4, the positions and numbers of cysteine residues essential for forming a characteristic finger structure in the region are completely conserved.
また、 配列番号 1で示されるアミノ酸配列において、 208番目のプロリン残 基〜 433番目のノ リン残基の領域は、 11£1^2と94%、 11£1^1と63%、 hERと 35%の相同性をそれぞれ示す。 hERの対応するアミノ酸配列からな る領域はリガンド結合領域とされるが、 該領域は核内レセプ夕一ファミ リ一で相 同性の高い領域であることが知られている。 In the amino acid sequence represented by SEQ ID NO: 1, the region from the proline residue at position 208 to the residue at position 433 is 94% in 11 £ 1 ^ 2, 63% in 11 £ 1 ^ 1 and 63% in hER. Each shows 35% homology. The region consisting of the corresponding amino acid sequence of hER is regarded as the ligand binding region, and this region corresponds to the nuclear receptor family. It is known that the region is highly homogenous.
従って、 E R R 4がリガンド結合能および D N A結合能を有することは明白で ある。  Therefore, it is clear that ERR4 has ligand binding ability and DNA binding ability.
さらに、 N末端領域において、 ERR 4は hERR 2とは高い相同性を示すが、 hERR 1および hERとは低い相同性を示す。核内レセプ夕一の N末端領域は、 転写制御領域であり、 かつ該領域はフアミリー間で相同性が比較的低い領域であ ることが知られている。  Furthermore, in the N-terminal region, ERR 4 shows high homology with hERR 2 but low homology with hERR 1 and hER. It is known that the N-terminal region of the nuclear receptor is a transcription control region, and that this region has relatively low homology between families.
このように、 ERR4のアミノ酸配列は、 DNA結合領域、 リガンド結合領域、 転写制御領域等、 核内レセプ夕一に特徴的な構造を有している。  Thus, the amino acid sequence of ERR4 has a unique structure in the nuclear receptor, such as a DNA binding region, a ligand binding region, and a transcription control region.
以上のように、 配列番号 2で示される塩基配列からなる D N Aが一旦取得され、 その塩基配列が決定された後は、 該塩基配列の 5' 端および 3' 端の塩基配列に 基づいたプライマーを調製し、 ヒトまたは非ヒト動物の網膜、 胎盤等の組織また は細胞に含まれる mRNAから合成した cDNAあるいは cDNAライブラリー を銪型として、 PCR法 〔PCR Protocols, Academic Press (1990)〕 を用いて D NAの増幅を行うことにより、 本発明の DNAを取得することができる。  As described above, once the DNA consisting of the nucleotide sequence of SEQ ID NO: 2 has been obtained and its nucleotide sequence has been determined, primers based on the nucleotide sequences at the 5 ′ and 3 ′ ends of the nucleotide sequence are used. Using a cDNA or cDNA library prepared from the mRNA contained in tissues or cells of the retina, placenta, etc. of human or non-human animals using the PCR method (PCR Protocols, Academic Press (1990)) The DNA of the present invention can be obtained by performing DNA amplification.
また、 配列番号 2で示される DN Aの全長あるいは一部をプロ一ブとして、 ヒ トまたは非ヒト動物の網膜、 胎盤等の組織または細胞に含まれる mRN Aから合 成した c DNAあるいは c DNAライブラリーに対してコロニ一ハイブリダイゼ —シヨンやプラークハイブリダイゼ一シヨン (モレキュラー ·クローニング第 2 版) を行うことにより、 本発明の DNAを取得することができる。  In addition, cDNA or cDNA synthesized from mRNA contained in tissues or cells of human or non-human animal retina, placenta, etc. as a probe using the full length or a part of DNA represented by SEQ ID NO: 2 as a probe The DNA of the present invention can be obtained by performing colony hybridization or plaque hybridization (molecular cloning second edition) on the library.
決定された DN Aの塩基配列に基づいて、 フォスフォアミダイ ト法を利用した パーキン 'エルマ一社の DN A合成機 model 392等の D N A合成機で化学合成する ことにより、 本発明の DNAを取得することもできる。  Based on the determined nucleotide sequence of DNA, the DNA of the present invention is chemically synthesized using a DNA synthesizer such as Perkin 'Elma's DNA synthesizer model 392 using the phosphoramidite method. You can also get it.
取得した DN Aについて、 該 DN Aを含む組換えべクタ一を宿主細胞に導入し て得られる形質転換体を用いて蛋白質を発現させることにより、 または該 DNA がコ一ドするアミノ酸配列と hERR 2、 hERR 1または hERのアミノ酸配 列との相同性を比較することにより、 該 D N Aがリガンド結合能および D N A結 合能を有する蛋白質をコードする DN Aであることを確認することができる。 上述の方法で取得した本発明の D N Aおよび D N A断片を用いて、 常法あるい は上記の DN A合成機により、 本発明の DN Aの一部の配列を有するアンチセン ス ·オリゴヌクレオチド、 センス ·オリゴヌクレオチド等のオリゴヌクレオチド を調製することができる。 また R N Aを含むオリゴヌクレオチドを調製すること もできる。 The obtained DNA is expressed by using a transformant obtained by introducing a recombinant vector containing the DNA into a host cell, or by expressing the amino acid sequence encoded by the DNA with hERR. 2. By comparing the homology with the amino acid sequence of hERR1 or hER, it can be confirmed that the DNA is a DNA encoding a protein having ligand-binding ability and DNA-binding ability. Using the DNA and DNA fragment of the present invention obtained by the above method, an antisense having a partial sequence of the DNA of the present invention can be obtained by a conventional method or the above-mentioned DNA synthesizer. Oligonucleotides such as oligonucleotides and sense oligonucleotides can be prepared. Oligonucleotides containing RNA can also be prepared.
該ォリゴヌクレオチドとしては、 上記 D N Aの有する塩基配列中の連続した 5 〜6 0塩基と同じ配列を有する D N Aまたは該 D N Aと相補的な配列を有する D N Aをあげることができる。 またこれらの D N Aと相補的な配列を有する R N A も本発明のオリゴヌクレオチドである。  Examples of the oligonucleotide include DNA having the same sequence as the consecutive 5 to 60 bases in the nucleotide sequence of DNA, and DNA having a sequence complementary to the DNA. RNA having a sequence complementary to these DNAs is also an oligonucleotide of the present invention.
該オリゴヌクレオチドとして、 例えば、 配列番号 2に示される塩基配列中の連 続した 5〜6 0塩基と同じ配列を有する D N Aまたは該 D N Aと相補的な配列を 有する D N Aをあげることができる。 センスプライマーおよびアンチセンスブラ イマ一として用いる場合には、 両者の融解温度 (T m) および塩基数が極端に変 わることのない上記のオリゴヌクレオチドが好ましい。 具体的には、 配列番号 3 〜 6に示された塩基配列を有するオリゴヌクレオチドをあげることができる。 更に、 これらオリゴヌクレオチドの誘導体 (以下、 オリゴヌクレオチド誘導体 という) も本発明のオリゴヌクレオチドとして利用することができる。  Examples of the oligonucleotide include DNA having the same sequence as 5 to 60 consecutive nucleotides in the nucleotide sequence shown in SEQ ID NO: 2 or DNA having a sequence complementary to the DNA. When used as a sense primer and an antisense primer, the above oligonucleotides in which the melting temperature (Tm) and the number of bases of both do not extremely change are preferable. Specific examples include oligonucleotides having the nucleotide sequences shown in SEQ ID NOs: 3 to 6. Furthermore, derivatives of these oligonucleotides (hereinafter, referred to as oligonucleotide derivatives) can also be used as the oligonucleotide of the present invention.
オリゴヌクレオチド誘導体としては、 オリゴヌクレオチド中のリン酸ジエステ ル結合がホスフォロチォェ一ト結合に変換されたォリゴヌクレオチド誘導体、 ォ リゴヌクレオチド中のリン酸ジエステル結合が N 3,一P 5,ホスフォアミデート 結合に変換されたオリゴヌクレオチド誘導体、 オリゴヌクレオチド中のリボース とリン酸ジエステル結合がぺプチド核酸結合に変換されたォリゴヌクレオチド誘 導体、 ォリゴヌクレオチド中のゥラシルが C— 5プロピニルゥラシルで置換され たオリゴヌクレオチド誘導体、 オリゴヌクレオチド中のゥラシルが C— 5チアゾ —ルゥラシルで置換されたオリゴヌクレオチド誘導体、 オリゴヌクレオチド中の シトシンが C一 5プロピニルシトシンで置換されたオリゴヌクレオチド誘導体、 オリゴヌクレオチド中のシトシンがフエノキサジン修飾シトシン (phenoxazine- modified cytosine)で置換されたォリゴヌクレオチド誘導体、 ォリゴヌクレオチ ド中のリボースが 2,一 0—プロピルリボースで置換されたォリゴヌクレオチド 誘導体、 あるいはオリゴヌクレオチド中のリボースが 2 '—メ トキシェトキシリボ —スで置換されたオリゴヌクレオチド誘導体等があげられる 〔細胞工学,!^, 1463 ( 1997)〕 。 2. 本発明の蛋白質の製造 Oligonucleotide derivatives include oligonucleotide derivatives in which phosphodiester bonds in the oligonucleotide have been converted to phosphorothioate bonds, and phosphodiester bonds in the oligonucleotides having N3,1-P5, phosphoamidates. Oligonucleotide derivative in which the ribose and phosphodiester bonds in the oligonucleotide have been converted to peptide nucleic acid bonds, and peracyl in the oligonucleotide has been replaced with C-5 propynyl peracyl Oligonucleotide derivatives in which peracyl in the oligonucleotide is substituted with C-5 thiazo-peruracil, oligonucleotide derivatives in which cytosine in the oligonucleotide is substituted with C-5-propynylcytosine, oligonucleotides Oligonucleotide derivatives in which the cytosine in the oligonucleotide has been replaced with phenoxazine-modified cytosine, oligoribonucleotide derivatives in which the ribose in the oligonucleotide has been replaced by 2,10-propylribose, or oligonucleotides in the oligonucleotide Oligonucleotide derivatives in which ribose is substituted with 2'-methoxetoxyribose, etc. [Cell engineering,! ^, 1463 (1997)]. 2. Production of the protein of the present invention
本発明の蛋白質は、 モレキュラー 'クロ一ニング第 2版、 カレント .プロトコ —ルズ ·ィン 'モレキュラー ·バイオロジー等に記載された方法等を用い、 例え ば以下の方法により、 本発明の DN Aを宿主細胞中で発現させて、 製造すること ができる。  The protein of the present invention can be prepared by the method described in Molecular Cloning, Second Edition, Current Protocol—Luzin, Molecular Biology, etc., for example, by the following method. Can be produced by expressing it in a host cell.
全長 cDNAをもとにして、 必要に応じて、 該蛋白質をコードする部分を含む 適当な長さの DNA断片を調製する。 また、 該蛋白質をコードする部分の塩基配 列を、 宿主の発現に最適なコドンとなるように、 塩基を置換することにより、 該 蛋白質の生産率を向上させることができる。  Based on the full-length cDNA, if necessary, a DNA fragment of an appropriate length containing a portion encoding the protein is prepared. Further, by substituting the base sequence of the portion encoding the protein such that the base sequence becomes an optimal codon for expression in the host, the production rate of the protein can be improved.
該 DNA断片、 または全長 cDNAを適当な発現べクタ一のプロモーターの下 流に揷入することにより、 組換えベクターを作製する。  A recombinant vector is prepared by inserting the DNA fragment or full-length cDNA into the downstream of a promoter in an appropriate expression vector.
該組換えベクターを、 該発現ベクターに適合した宿主細胞に導入することによ り、 本発明の蛋白質を生産する形質転換体を得ることができる。  By introducing the recombinant vector into a host cell suitable for the expression vector, a transformant producing the protein of the present invention can be obtained.
宿主細胞としては、 細菌、 酵母、 動物細胞、 昆虫細胞、 植物細胞等、 目的とす る遺伝子を発現できるものであればいずれも用いることができる。  As the host cell, any cells that can express the target gene, such as bacteria, yeast, animal cells, insect cells, and plant cells, can be used.
発現ベクターとしては、 上記宿主細胞において自立複製可能ないしは染色体中 への組込が可能で、 本発明の蛋白質をコ一ドする D N Aを転写できる位置にプロ モ—夕一を含有しているものが用いられる。  Expression vectors which are capable of autonomous replication in the above host cells or capable of being integrated into a chromosome, and which contain a promoter at a position where DNA encoding the protein of the present invention can be transcribed are used. Used.
細菌等の原核生物を宿主細胞として用いる場合は、 本発明の蛋白質をコ一ドす る DNAを含有してなる組換えベクターは原核生物中で自立複製可能であると同 時に、 プロモー夕一、 リボソーム結合配列、 ERR 4遺伝子、 転写終結配列、 よ り構成されたベクターであることが好ましい。 プロモーターを制御する遺伝子が 含まれていてもよい。  When a prokaryote such as a bacterium is used as a host cell, the recombinant vector containing the DNA encoding the protein of the present invention can replicate autonomously in the prokaryote, The vector is preferably composed of a ribosome binding sequence, the ERR4 gene, a transcription termination sequence, and the like. A gene that controls the promoter may be included.
発現べクタ一としては、 例えば、 pBTrp2、 pBTac pBTac2 (いずれもべ一リン ガーマンハイム社より市販) 、 PKK233- 2 (フアルマシア社) 、 pSE280 (インビト ロジェン社) 、 pGEMEX- 1 〔プロメガ社製〕 、 pQE- 8 (キアゲン社製) 、 pKYPIO (特 開昭 58- 110600)、 pKYP200 CAgric. Biol. Chem. , 48, 669 (1984)〕、 pLSAl CAgric. Biol. Chem. , 53, 277 (1989)〕 、 pGELl 〔Proc. Natl. Acad. Sci. USA, 82, 4306 (1985)〕、 pBluescript II SK (-) (ストラ夕ジーン社製)、 pTrs30 (FERM BP-5407)、 pTrs32(FERM BP- 5408)、 pGHA2(FERM BP- 400)、 pGKA2(FERM B- 6798)、 pTerm2 (特 開平 3- 22979、 US4686191、 US4939094, US5160735) 、 pEG400 . Bacteriol . , 172, 2392 ( 1990)〕 、 pGEX (フアルマシア社製) 、 pET- 3 (ノバジェン社製) 、 pSupex、 pUB110、 pTP5、 pC194、 pTrxFus ( Invitrogen社製)、 p AL-c2 (New England Biolabs 社製) 、 pETシステム (ノバジェン社製)等をあげることができる。 Examples of expression vectors include pBTrp2, pBTac and pBTac2 (all commercially available from Behringer Garmanheim), PKK233-2 (Pharmacia), pSE280 (Invitrogen), pGEMEX-1 (Promega), pQE-8 (manufactured by QIAGEN), pKYPIO (Japanese Patent Publication No. 58-110600), pKYP200 CAgric. Biol. Chem., 48, 669 (1984)], pLSAl CAgric. Biol. Chem., 53, 277 (1989)] , PGELl [Proc. Natl. Acad. Sci. USA, 82, 4306 (1985)], pBluescript II SK (-) (manufactured by Straugene), pTrs30 (FERM BP-5407), pTrs32 (FERM BP-5408) , PGHA2 (FERM BP-400), pGKA2 (FERM B-6798), pTerm2 (Special Kaihei 3-22979, US4686191, US4939094, US5160735), pEG400.Bacteriol., 172, 2392 (1990)), pGEX (Pharmacia), pET-3 (Novagen), pSupex, pUB110, pTP5, pC194, pTrxFus (Invitrogen), pAL-c2 (New England Biolabs), pET system (Novagen) and the like.
プロモー夕—としては、 大腸菌や枯草菌等の宿主細胞中で発現できるものであ ればいかなるものでもよい。 例えば、 ^プロモ一夕一 (Pfrp)、 lacプロモータ 一 ( P lac)、 P Lプロモーター、 P Rプロモ一夕一、 P SEプロモ一夕一等の、 大腸菌 やファージ等に由来するプロモーター、 S P 0 1プロモー夕一、 S P 0 2プロモ 一夕一、 p e n Pプロモー夕一等をあげることができる。 また P;^£を 2つ直列さ せたプロモーター (P^£x 2 ) 、 ; プロモー夕一、 lacT7プロモーター、 let I プロモー夕一のように人為的に設計改変されたプロモータ一等も用いることがで きる。 The promoter may be any promoter that can be expressed in host cells such as Escherichia coli and Bacillus subtilis. For example, ^ Promo Isseki one (Pfrp), lac promoter one (P lac), P L promoter, P R promoter Isseki one, promoter derived from the P SE promoter Isseki one such as, Escherichia coli, phage, etc., SP 0 1 Promo Yuichi, SP 0 2 Promo Yuichi, pen P Promo Yuichi, etc. Also, use a promoter in which two P; ^ £ are connected in series (P ^ £ x 2); a promoter that has been artificially designed and modified, such as Promo Yuichi, lacT7 promoter, let I Promo Yuichi, etc. I can do it.
リボソーム結合配列であるシャイン—ダルガノ (Shine- Dalgarno) 配列と開始 コドンとの間を適当な距離 (例えば 6 ~ 1 8塩基) に調節したプラスミ ドを用い ることが好ましい。  It is preferable to use a plasmid in which the distance between the Shine-Dalgarno sequence, which is a ribosome binding sequence, and the initiation codon is adjusted to an appropriate distance (for example, 6 to 18 bases).
本発明の組換えべクタ一においては、 本発明の D N Aの発現には転写終結配列 は必ずしも必要ではないが、 構造遺伝子の直下に転写終結配列を配置することが 好ましい。  In the recombinant vector of the present invention, a transcription termination sequence is not necessarily required for expression of the DNA of the present invention, but it is preferable to arrange a transcription termination sequence immediately below a structural gene.
宿主細胞としては、 ェシエリヒア属、 セラチア属、 バチルス属、 ブレビバクテ リウム属、 コリネバクテリウム属、 ミクロバクテリウム属、 シユードモナス属等 に属する微生物、 例えば、 Escherichia coli XLl-Blue Escherichia coli XL2- Blueヽ Escherichia coli DH1、 Escherichia coli MC1000, Escherichia coli KY3276、 Escherichia coli W1485、 Escherichia coli JM109N Escherichia coli HB101、 Escherichia coli No.49、 Escherichia coli W3110、 Escherichia coli NY49、 Serratia ficaria、 Serratia fonticola、 Serratia liquefaciensN Serratia marcescens―、 Bacillus subtilis、 Bacillus amyloliquefaciens Brevibacterium ammoniagenes、 Brevibacterium immariophilum ATCC14068、 Brevibacterium saccharolyticum ATCC14066、 Corynebacterium glutamicum ATCC13032、 Host cells include microorganisms belonging to the genus Escherichia, Serratia, Bacillus, Brevibacterium, Corynebacterium, Microbacterium, Pseudomonas, etc., for example, Escherichia coli XLl-Blue Escherichia coli XL2-Blue XLEscherichia coli DH1, Escherichia coli MC1000, Escherichia coli KY3276, Escherichia coli W1485, Escherichia coli JM109 N Escherichia coli HB101, Escherichia coli No.49, Escherichia coli W3110, Escherichia coli NY49, Serratia ficaria, Serratia fonticola, Serratia liquefaciens N Serratia marcescens-, Bacillus subtilis, Bacillus amyloliquefaciens Brevibacterium ammoniagenes, Brevibacterium immariophilum ATCC14068, Brevibacterium saccharolyticum ATCC14066, Corynebacterium glutamicum ATCC13032,
Corynebacterium glutamicum ATCC14067、 Corynebacterium glutamicum ATCC13869、 Corynebacterium acetoacidoDhi lum ATCC13870、 Microbacterium ammoniaphiliun ATCC15354, Pseudomonas sp. D-0110等をあげることができる。 Corynebacterium glutamicum ATCC14067, Corynebacterium glutamicum ATCC13869, Corynebacterium acetoacidoDhi lum ATCC13870, Microbacterium ammoniaphiliun ATCC15354, Pseudomonas sp. D-0110 and the like.
組換えベクターの導入方法としては、 上記宿主細胞へ DN Aを導入する方法で あればいずれも用いることができ、例えば、カルシウムイオンを用いる方法〔 P r 0 c . As a method for introducing a recombinant vector, any method can be used as long as it is a method for introducing DNA into the above host cells, and for example, a method using calcium ions [Pr0c.
Natl. Acad. Sci. USA, 69, 2110 (1972)〕 、 プロトプラスト法 (特開昭 63- 248394)、 エレクトロポレーシヨン法〔Gene, , 107 (1982)、 Molecular & General Genetics,Natl. Acad. Sci. USA, 69, 2110 (1972)], protoplast method (JP-A-63-248394), electroporation method (Gene, 107 (1982), Molecular & General Genetics,
168, 111 (1979)〕 等をあげることができる。 168, 111 (1979)].
酵母菌株を宿主細胞として用いる場合には、発現ベクターとして、例えば、 YEpl3 (ATCC37115)、 YEp24 (ATCC37051)、 YCp50 (ATCC37419)、 pHS19、 pHS15等を用 いることができる。  When a yeast strain is used as a host cell, for example, YEpl3 (ATCC37115), YEp24 (ATCC37051), YCp50 (ATCC37419), pHS19, pHS15 and the like can be used as expression vectors.
プロモー夕一としては、 酵母菌株中で発現できるものであればいずれのものを 用いてもよく、 例えば、 PH05プロモー夕一、 PGKプロモーター、 GAPプロモーター、 ADHプロモーター、 gal 1プロモ一夕一、 gal 10プロモーター、 ヒートショックポ リペプチドプロモータ一、 MFひ 1プロモーター、 CUP 1プロモータ一等のプロモ一 夕一をあげることができる。  Any promoter can be used as long as it can be expressed in yeast strains. For example, PH05 promoter, PGK promoter, GAP promoter, ADH promoter, gal 1 promoter, gal 10 Promoters such as promoter, heat shock polypeptide promoter, MF promoter and CUP promoter can be mentioned.
宿主細胞としては、 サッカロマイセス属、 シゾサヅカロマイセス属、 クルイべ 口ミセス属、 トリコスポロン属、 シヮニォミセス属、 ピチア属等に属する酵母菌 株をあげることができ、 具体的には、 Saccharomyces cerevisiae,  Examples of the host cell include yeast strains belonging to the genera Saccharomyces, Schizosaccharomyces, Kluybium mycetes, Trichosporone, Schizinomyces, Pichia, and the like.Specifically, Saccharomyces cerevisiae ,
Schizosaccharomyces pombe、 Kluyveromyces lactis、 Trichosporon pullulans、 Schwanniomyces alluvius -、 Pichia pastoris等をあげることができる。 Schizosaccharomyces pombe, Kluyveromyces lactis, Trichosporon pullulans, Schwanniomyces alluvius-, Pichia pastoris and the like.
組換えベクターの導入方法としては、 酵母に DN Aを導入する方法であればい ずれも用いることができ、 例えば、 エレクトロポレーシヨン法 〔Methods in Enzymology, 194, 182 (1990)〕 、 スフエロプラスト法 〔pr0c. Natl. Acad. Sci. USA, 81, 4889 (1984)〕 、 酢酸リチウム法 〔Journal of Bacteriology, 153, 163 (1983)〕 、 Proc. Natl. Acad. Sci. USA, 75, 1929 ( 1978)記載の方法等をあげる ことができる。 As a method for introducing a recombinant vector, any method can be used as long as it is a method for introducing DNA into yeast. For example, electroporation [Methods in Enzymology, 194, 182 (1990)], spheroplast Law [p r0 c. Natl. Acad. Sci. USA, 81, 4889 (1984) ], the lithium acetate method [Journal of Bacteriology, 153, 163 ( 1983) ], Proc. Natl. Acad. Sci . USA, 75, 1929 (1978).
動物細胞を宿主として用いる場合には、 発現べクタ一として、 例えば、 pcDN AI、 p c DM8 (フナコシ社より市販)、 pAGE107〔特開平 3- 22979;Cytotechnology, 3, 133, (1990)〕、 pAS3-3 (特開平 2- 227075)、 pCDM8 CNature, 329, 840,(1987)〕 、 pcDNAI/Amp (Invitrogen社製)、 REP4 (Invitrogen社製) 、 pAGE103 〔J. Biochemistry, 101, 1307 (1987)〕、 pAGE210, pAMo、 pA oA等をあげることがで ぎる。 When an animal cell is used as a host, examples of expression vectors include pcDNAI, pcDM8 (commercially available from Funakoshi), pAGE107 [JP-A-3-22979; Cytotechnology, 3, 133, (1990)], pAS3 -3 (JP-A-2-227075), pCDM8 CNature, 329, 840, (1987)], pcDNAI / Amp (manufactured by Invitrogen), REP4 (manufactured by Invitrogen), pAGE103 (J. Biochemistry, 101, 1307 (1987)) ], PAGE210, pAMo, pA oA, etc. Cut.
プロモーターとしては、 動物細胞中で発現できるものであればいずれも用いる ことができ、 例えば、 サイ トメガロウィルス (CMV) の IE ( immediate early) 遺 伝子のプロモー夕一、 SV40の初期プロモーター、 レトロウイルスのプロモー夕一、 メタ口チォネインプロモーター、 ヒートショックプロモー夕一、 S Rひプロモー 夕一等をあげることができる。 また、 ヒト C M Vの I E遺伝子のェンハンサーを プロモ一夕一と共に用いてもよい。  Any promoter can be used as long as it can be expressed in animal cells. Examples of such promoters include the cytomegalovirus (CMV) IE (immediate early) gene promoter, the SV40 early promoter, and the retro promoter. Examples include virus promoters, meta-mouth choonein promoter, heat shock promoters, and SR promoter promoters. Alternatively, the enhancer of the IE gene of human CMV may be used together with the promoter.
宿主細胞としては、 マウス · ミエローマ細胞、 ラット ' ミエローマ細胞、 マウ ス'ハイプリ ドーマ細胞、ヒトの細胞であるナマルバ(Namalwa)細胞または Namalwa KJM- 1細胞、 ヒト胎児腎臓細胞、 ヒト白血病細胞、 アフリカミ ドリザル腎臓細胞、 チャイニーズ 'ハムスターの細胞である CH0細胞、 HBT5637 (特開昭 63-299) 等を あげることができる。  Host cells include mouse / myeloma cells, rat 'myeloma cells, mouse' hybridoma cells, Namalwa cells or Namalwa KJM-1 cells, human embryonic kidney cells, human leukemia cells, and African green monkeys. Kidney cells, Chinese hamster cells such as CH0 cells, HBT5637 (Japanese Patent Application Laid-Open No. 63-299), and the like.
マウス · ミエローマ細胞としては、 SP2/0、 NS0等、 ラヅ ト ' ミエローマ細胞と しては YB2/0等、 ヒト胎児腎臓細胞としては HEK293(ATCC: CRL- 1573)等、 ヒト白血 病細胞としては、 BALL- 1等、 アフリカミ ドリザル腎臓細胞としては COS- 1、 COS- 7 等をあげることができる。  Mouse / myeloma cells include SP2 / 0 and NS0, rat myeloma cells include YB2 / 0, etc., human fetal kidney cells include HEK293 (ATCC: CRL-1573), and human leukemia cells. Examples include BALL-1 and the like, and African green monkey kidney cells include COS-1 and COS-7.
組換えベクターの導入方法としては、 動物細胞に D N Aを導入する方法であれ ばいずれも用いることができ、 例えば、 エレクトロボレ一シヨン法  As a method for introducing a recombinant vector, any method can be used as long as it is a method for introducing DNA into animal cells.
CCytotechnology, 3, 133 ( 1990)〕 、 リン酸カルシウム法 (特開平 2- 227075) 、 リポフエクシヨン法 Proc. Natl . Acad. Sci . USA, 84, 7413 ( 1987)〕 、 Virology, 52, 456 ( 1973)に記載の方法等をあげることができる。  CCytotechnology, 3, 133 (1990)], calcium phosphate method (Japanese Patent Laid-Open No. 2-227075), lipofection method Proc. Natl. Acad. Sci. USA, 84, 7413 (1987)), and Virology, 52, 456 (1973). And the like.
昆虫細胞を宿主として用いる場合には、 例えばバキュロウィルス ·イクスプレ ヅシヨン ·ベクタ一ス ァ · ラ不ラ トリ—— 'マ二ユア レ 〔Baculovirus Expression Vectors, A Laboratory Manual, W. H. Freeman and Company, NewYork ( 1992)〕 ヽ モレキュラー'バイオロジー ァ 'ラボラトリー'マニュアル(Molecular Biology, A Laboratory Manual) 、 カレント 'プロトコールズ 'イン 'モレキュラー ·ノ イ ォロジ一 、 Bio/Technology, 6, 47 ( 1988)等に記載された方法によって、 蛋白質 を発現することができる。  When an insect cell is used as a host, for example, baculovirus expression, vector sacrifice, lacto-radiation, etc. can be performed using Baculovirus Expression Vectors, A Laboratory Manual, WH Freeman and Company, New York (1992). )] さ れ Molecular Biology, Laboratory Laboratory Manual (Molecular Biology, A Laboratory Manual), Current 'Protocols' in' Molecular Noology, Bio / Technology, 6, 47 (1988), etc. Depending on the method, the protein can be expressed.
即ち、 組換え遺伝子導入ベクターおよびバキュロウィルスを昆虫細胞に共導入 して昆虫細胞培養上清中に組換えウィルスを得た後、 さらに組換えウィルスを昆 虫細胞に感染させ、 蛋白質を発現させることができる。 That is, the recombinant gene transfer vector and the baculovirus are co-transfected into insect cells to obtain the recombinant virus in the culture supernatant of the insect cells, and then the recombinant virus is transferred to the insect cell. It can infect insect cells and express proteins.
該方法において用いられる遺伝子導入ベクターとしては、 例えば、 pVL1392、 pVL1393、 pBlueBacI I I (ともに Invitorogen社製)等をあげることができる。  Examples of the gene transfer vector used in the method include pVL1392, pVL1393, pBlueBacII (all manufactured by Invitorogen) and the like.
バキュロウィルスとしては、 例えば、 夜盗蛾科昆虫に感染するウィルスである アウトグラファ ·カリフォルニ力 ·ヌクレア一 ·ポリへドロシス ·ウィルス (Autographa californica nuclear polyhedrosis virus)等を用レヽることができる 0 昆虫細胞としては、 Spodoptera frugiperdaの卵巣細胞、 Tric opl^ia n丄の卵 巣細胞、 力ィコ卵巣由来の培養細胞等を用いることができる。 The baculovirus, for example, as 0 insect cells that can use Rereru a burglar Gaka insects is a virus that infects out publicly available, power Nuclear one poly to Doroshisu virus (Autographa californica nuclear polyhedrosis virus) and the like The ovarian cells of Spodoptera frugiperda, the ovarian cells of Tricoplia nia, and the cultured cells derived from the chick ovary can be used.
Spodoptera frugiperdaの卵巣細胞としては Sf9、 Sf21 (バキュロウィルス ·ィ クスプレヅシヨン 'ベクターズ ァ 'ラボラトリー'マニュアル)等、 Trichoplusia の卵巣細胞としては High 5、 BTI-TN-5B1-4 (インビトロジェン社製) 等、 カイ コ卵巣由来の培養細胞としては Bombyx mori N4等をあげることができる。  The ovarian cells of Spodoptera frugiperda include Sf9 and Sf21 (baculovirus expression 'vectors' and 'laboratory' manuals), and the like. The ovarian cells of Trichoplusia include High 5 and BTI-TN-5B1-4 (manufactured by Invitrogen). Bombyx mori N4 and the like can be mentioned as cultured cells derived from ovaries.
組換えゥィルスを調製するための、 昆虫細胞への上記組換え遺伝子導入べクタ 一と上記バキュロウィルスの共導入方法としては、 例えば、 リン酸カルシウム法 (特開平 2- 227075) 、 リポフエクシヨン法 〔Proc. Natl . Acad. Sci . USA, 84, 7413 ( 1987)〕 等をあげることができる。  Methods for co-transferring the above-described recombinant gene transfer vector and the baculovirus into insect cells to prepare a recombinant virus include, for example, a calcium phosphate method (Japanese Patent Laid-Open No. 2-227075), a lipofection method [Proc. Natl. Acad. Sci. USA, 84, 7413 (1987)].
植物細胞を宿主細胞として用いる場合には、 発現べクタ一として、 例えば、 T iプラスミ ド、 タバコモザイクウィルスベクター等をあげることができる。  When a plant cell is used as a host cell, examples of the expression vector include Ti plasmid and tobacco mosaic virus vector.
プロモーターとしては、 植物細胞中で発現できるものであればいずれのものを 用いてもよく、 例えば、 カリフラワーモザイクウィルス (CaMV) の 35Sプロモー夕 一、 ィネアクチン 1プロモーター等をあげることができる。  Any promoter can be used as long as it can be expressed in plant cells, and examples thereof include the 35S promoter of cauliflower mosaic virus (CaMV) and the geneactin 1 promoter.
宿主細胞としては、 タバコ、 ジャガイモ、 トマト、 ニンジン、 ダイズ、 ァブラ ナ、 アルフアルファ、 イネ、 コムギ、 ォォムギ等の植物細胞等をあげることがで きる。  Examples of the host cell include plant cells of tobacco, potato, tomato, carrot, soybean, abrana, alfa alfa, rice, wheat, wheat, and the like.
組換えベクターの導入方法としては、 植物細胞に D N Aを導入する方法であれ ばいずれも用いることができ、例えば、 ァグロパクテリゥム (Agrobacteriim) (特 開昭 59- 140885、 特閧昭 60-70080、 W094/00977) 、 エレクトロポレーシヨン法 (特 開昭 60- 251887) 、 パーティクルガン (遺伝子銃) を用いる方法 (特許第 2606856、 特許第 2517813) 等をあげることができる。  Any method for introducing a recombinant vector can be used so long as it is a method for introducing DNA into plant cells. For example, Agrobacteriim (Agrobacteriim) (JP-A-59-140885, JP-A-60-160885) 70080, W094 / 00977), an electroporation method (Japanese Patent Publication No. 60-251887), a method using a particle gun (gene gun) (Japanese Patent No. 2606856, Japanese Patent No. 2517813), and the like.
遺伝子の発現方法としては、 直接発現以外に、 モレキュラー ·クロ一ニング第 2版に記載されている方法等に準じて、 分泌生産、 融合蛋白質発現等を行うこと ができる。 In addition to direct expression, there are molecular expression methods Secretory production, fusion protein expression, etc. can be performed according to the method described in the second edition.
酵母、 動物細胞、 昆虫細胞または植物細胞により発現させた場合には、 糖ある レ、は糖鎖が付加された蛋白質を得ることができる。  When expressed by yeast, animal cells, insect cells, or plant cells, a protein having a sugar chain or a sugar chain can be obtained.
以上のようにして得られる形質転換体を培地に培養し、 培養物中に本発明の蛋 白質を生成蓄積させ、 該培養物から採取することにより、 本発明の蛋白質を製造 することができる。  The protein of the present invention can be produced by culturing the transformant obtained as described above in a medium, producing and accumulating the protein of the present invention in the culture, and collecting from the culture.
本発明の形質転換体を培地に培養する方法は、 宿主の培養に用いられる通常の 方法に従って行うことができる。  The method for culturing the transformant of the present invention in a medium can be performed according to a usual method used for culturing a host.
大腸菌等の原核生物あるいは酵母等の真核生物を宿主として得られた形質転換 体を培養する培地としては、 該生物が資化し得る炭素源、 窒素源、 無機塩類等を 含有し、 形質転換体の培養を効率的に行える培地であれば天然培地、 合成培地の いずれを用いてもよい。  A culture medium for culturing a transformant obtained by using a prokaryote such as Escherichia coli or a eukaryote such as yeast as a host contains a carbon source, a nitrogen source, inorganic salts, and the like which can be used by the organism. Either a natural medium or a synthetic medium can be used as long as the medium can efficiently culture the cells.
炭素源としては、 該生物が資化し得るものであればよく、 グルコース、 フラク トース、 スクロース、 これらを含有する糖蜜、 デンプンあるいはデンプン加水分 解物等の炭水化物、 酢酸、 プロピオン酸等の有機酸、 エタノール、 プロパノール 等のアルコール類等を用いることができる。  The carbon source may be any one that can be assimilated by the organism; glucose, fructose, sucrose, molasses containing these, carbohydrates such as starch or starch hydrolysate, organic acids such as acetic acid and propionic acid, Alcohols such as ethanol and propanol can be used.
窒素源としては、 アンモニア、 塩化アンモニゥム、 硫酸アンモニゥム、 酢酸ァ ンモニゥム、 リン酸アンモニゥム等の無機酸もしくは有機酸のアンモニゥム塩、 その他の含窒素化合物、 ならびに、 ペプトン、 肉エキス、 酵母エキス、 コーンス チープリカー、 カゼイン加水分解物、 大豆粕および大豆粕加水分解物、 各種発酵 菌体およびその消化物等を用レ、ることができる。  Nitrogen sources include ammonia, ammonium chloride, ammonium sulfate, ammonium acetate, ammonium phosphate and other ammonium-containing salts of inorganic or organic acids, other nitrogen-containing compounds, peptone, meat extract, yeast extract, corn steep liquor, Casein hydrolyzate, soybean meal and soybean meal hydrolyzate, various fermented bacterial cells and digests thereof can be used.
無機塩としては、 リン酸第一カリウム、 リン酸第二カリウム、 リン酸マグネシ ゥム、 硫酸マグネシウム、 塩化ナトリウム、 硫酸第一鉄、 硫酸マン癌、 硫酸銅、 炭酸カルシウム等を用いることができる。  As the inorganic salt, potassium (II) phosphate, potassium (II) phosphate, magnesium phosphate, magnesium sulfate, sodium chloride, ferrous sulfate, man sulfate, copper sulfate, calcium carbonate, and the like can be used.
培養は、 通常振盪培養または深部通気攪拌培養等の好気的条件下で行う。 培養温度は 1 5〜4 0 °Cがよく、 培養時間は、 通常 1 6時間〜 7日間である。 培 養中の p Hは 3 . 0〜9 . 0に保持する。 p Hの調整は、 無機または有機の酸、 アルカリ溶液、 尿素、 炭酸カルシウム、 アンモニア等を用いて行う。  The cultivation is usually carried out under aerobic conditions such as shaking culture or deep aeration stirring culture. The culture temperature is preferably 15 to 40 ° C, and the culture time is usually 16 hours to 7 days. The pH during cultivation is maintained at 3.0 to 9.0. The pH is adjusted using an inorganic or organic acid, an alkaline solution, urea, calcium carbonate, ammonia, or the like.
また、 培養中必要に応じて、 アンピシリンやテトラサイクリン等の抗生物質を 培地に添加してもよい。 In addition, antibiotics such as ampicillin and tetracycline may be used during culture, if necessary. It may be added to the medium.
プロモーターとして誘導性のプロモータ一を用いた組換えベクターで形質転換 した微生物を培養するときには、 必要に応じてィンデューサ一を培地に添加して もよい。 例えば、 プロモ一夕一を用いた組換えべクタ一で形質転換した微生物 を培養するときにはイソプロビル一 5— D—チォガラクトビラノシド等を、 ; プ 口モータ—を用いた組換えべクタ—で形質転換した微生物を培養するときにはィ ンドールァクリル酸等を培地に添加してもよい。  When culturing a microorganism transformed with a recombinant vector using an inducible promoter as a promoter, an inducer may be added to the medium, if necessary. For example, when culturing a microorganism transformed with a recombinant vector using a promoter, isoprovir-15-D-thiogalactovyranoside, etc .; When culturing the microorganism transformed with-, indoleacrylic acid or the like may be added to the medium.
動物細胞を宿主として得られた形質転換体を培養する培地としては、 一般に使 用されている RPMI 1640培地 The Journal of the American Medical Association, 199, 519 ( 1967)〕、EagleのMEM培地〔Science, 122, 501 ( 1952)〕、DMEM培地〔Virology, 8, 396 ( 1959)〕、 199培地 proceeding of the Society for the Biological Medicine, 73, 1 ( 1950)〕 またはこれら培地に牛胎児血清等を添加した培地等を用いること ができる。  As culture media for culturing transformants obtained using animal cells as hosts, commonly used RPMI 1640 medium The Journal of the American Medical Association, 199, 519 (1967)], Eagle's MEM medium [Science, 122, 501 (1952)), DMEM medium (Virology, 8, 396 (1959)), 199 medium proceeding of the Society for the Biological Medicine, 73, 1 (1950)), or fetal calf serum or the like was added to these mediums. A medium or the like can be used.
培養は、 通常 p H 6〜8、 2 5〜4 0 ° 5 % C〇2存在下等の条件下で 1〜 7 日間行う。 Culture is carried out usually p H 6~8, 2 5~4 0 ° 5% C_〇 2 under the conditions such as the presence 1-7 days.
また、 培養中必要に応じて、 カナマイシン、 ペニシリン、 ストレプトマイシン 等の抗生物質を培地に添加してもよい。  If necessary, antibiotics such as kanamycin, penicillin and streptomycin may be added to the medium during the culturing.
昆虫細胞を宿主として得られた形質転換体を培養する培地としては、 一般に使 用されている TNM-FH培地(Pharmingen社製)、 Sf-900 I I SFM培地(Life Technologies 社製)、 ExCell400、 ExCe 11405 (いずれも JRH Biosciences社製)、 Grace' s Insect Medium CGrace, T. C. C. , Nature, 195, 788 ( 1962)〕 等を用いることができる。 培養は、 通常 p H 6〜7、 2 5〜3 0 °C等の条件下で、 1〜5日間行う。  Culture media for transformants obtained using insect cells as a host include commonly used TNM-FH medium (Pharmingen), Sf-900 II SFM medium (Life Technologies), ExCell400, ExCe 11405 (all manufactured by JRH Biosciences), Grace's Insect Medium CGrace, TCC, Nature, 195, 788 (1962)] and the like can be used. The cultivation is usually performed under conditions of pH 6 to 7 and 25 to 30 ° C for 1 to 5 days.
また、 培養中必要に応じて、 ゲンタマイシン等の抗生物質を培地に添加しても よい。  If necessary, an antibiotic such as gentamicin may be added to the medium during the culture.
植物細胞を宿主として得られた形質転換体は、 細胞として、 または植物の細胞 や器官に分化させて培養することができる。 該形質転換体を培養する培地として は、一般に使用されているムラシゲ'アンド 'スク一グ (MS)培地、ホワイ ト(White) 培地、 またはこれら培地にオーキシン、 サイ トカイニン等、 植物ホルモンを添加 した培地等を用いることができる。  A transformant obtained using a plant cell as a host can be cultured as a cell or after being differentiated into a plant cell or organ. As a medium for culturing the transformant, commonly used Murashige 'and' squeeg (MS) medium, white (White) medium, or a plant hormone such as auxin or cytokinin was added to these mediums. A medium or the like can be used.
培養は、 通常 p H 5〜9、 2 0〜4 0 °Cの条件下で 3〜 6 0日間行う。 また、 培養中必要に応じて、 カナマイシン、 ハイグロマイシン等の抗生物質を 培地に添加してもよい。 Culture is usually performed at pH 5 to 9, 20 to 40 ° C for 3 to 60 days. If necessary, antibiotics such as kanamycin and hygromycin may be added to the medium during the culture.
上記のとおり、 本発明の蛋白質をコードする D N Aを組み込んだ組換え体べク 夕一を保有する微生物、 動物細胞、 あるいは植物細胞由来の形質転換体を、 通常 の培養方法に従って培養し、 該蛋白質を生成蓄積させ、 該培養物より該蛋白質を 採取することにより、 該蛋白質を製造することができる。  As described above, a transformant derived from a microorganism, animal cell, or plant cell having the recombinant vector into which the DNA encoding the protein of the present invention is incorporated is cultured according to a usual culture method, and the protein By producing and accumulating, and collecting the protein from the culture, the protein can be produced.
遺伝子の発現方法としては、 直接発現以外に、 モレキュラー ·クローニング第 2版に記載されている方法等に準じて、 分泌生産、 融合蛋白質発現等を行うこと ができる。  As a method for expressing a gene, in addition to direct expression, secretory production, fusion protein expression, and the like can be performed according to the method described in Molecular Cloning, 2nd edition, and the like.
本発明の蛋白質の生産方法としては、 宿主細胞内に生産させる方法、 宿主細胞 外に分泌させる方法、 あるいは宿主細胞外膜上に生産させる方法があり、 使用す る宿主細胞や、 生産させる蛋白質の構造を変えることにより、 該方法を選択する ことができる。  Methods for producing the protein of the present invention include a method of producing the protein in a host cell, a method of secreting the protein out of the host cell, and a method of producing the protein on the host cell outer membrane. The method can be selected by changing the structure.
本発明の蛋白質が宿主細胞内あるいは宿主細胞外膜上に生産される場合、 ポー ルソンらの方法〔J. Biol . Chem. , 264, 17619 ( 1989 )〕、 ロウらの方法!; Proc . Natl . Acad. Sci ., USA, 86, 8227 ( 1989 )、 Genes Develop. , 4, 1288 ( 1990 )〕 、 または 特開平 05-336963、 W094/23021等に記載の方法を準用することにより、 該蛋白質を 宿主細胞外に積極的に分泌させることができる。  When the protein of the present invention is produced in a host cell or on a host cell outer membrane, the method of Paulson et al. [J. Biol. Chem., 264, 17619 (1989)], the method of Lowe et al.!; Proc. Natl. Acad. Sci., USA, 86, 8227 (1989), Genes Develop., 4, 1288 (1990)], or the method described in JP-A-05-336963, W094 / 23021, etc. Can be actively secreted out of the host cell.
即ち、 遺伝子組換えの手法を用いて、 本発明の蛋白質の活性部位を含む蛋白質 の手前にシグナルべプチドを付加した形で発現させることにより、 本発明の蛋白 質を宿主細胞外に積極的に分泌させることができる。  That is, the protein of the present invention is expressed in a form in which a signal peptide is added in front of the protein containing the active site of the protein of the present invention using a gene recombination technique, so that the protein of the present invention can be positively extracellularly outside the host cells. It can be secreted.
また、 特開平 2- 227075に記載されている方法に準じて、 ジヒドロ葉酸還元酵素 遺伝子等を用いた遺伝子増幅系を利用して生産量を上昇させることもできる。 さらに、 遺伝子導入した動物または植物の細胞を再分化させることにより、 遺 伝子が導入された動物個体(トランスジヱニック非ヒト動物)または植物個体(ト ランスジエニック植物) を造成し、 これらの個体を用いて本発明の蛋白質を製造 することもできる。  Further, according to the method described in Japanese Patent Application Laid-Open No. 2-227075, the production amount can be increased by using a gene amplification system using a dihydrofolate reductase gene or the like. Furthermore, the transgenic animal or plant cells are redifferentiated to create an animal (transgenic non-human animal) or plant (transgenic plant) into which the gene has been introduced. The protein of the present invention can also be produced using an individual.
形質転換体が動物個体または植物個体の場合は、 通常の方法に従って、 飼育ま たは栽培し、 該蛋白質を生成蓄積させ、 該動物個体または植物個体より該蛋白質 を採取することにより、 該蛋白質を製造することができる。 動物個体を用いて本発明の蛋白質を製造する方法としては、 例えば公知の方法 American Journal of Clinical Nutrition, 63, 639S ( 1996 )、 American Journal of Clinical Nutrition, 63, 627S ( 1996 )、 Bio/Technology, 9, 830 ( 1991 )〕 に 準じて遺伝子を導入して造成した動物中に本発明の蛋白質を生産する方法があげ られる。 When the transformant is an individual animal or plant, the protein is raised or cultivated according to a usual method to produce and accumulate the protein, and the protein is collected from the individual animal or plant to obtain the protein. Can be manufactured. Methods for producing the protein of the present invention using animal individuals include, for example, known methods American Journal of Clinical Nutrition, 63, 639S (1996), American Journal of Clinical Nutrition, 63, 627S (1996), Bio / Technology, 9, 830 (1991)] to produce the protein of the present invention in an animal constructed by introducing a gene.
動物個体の場合は、 例えば、 本発明の蛋白質をコードする D N Aを導入したト ランスジエニック非ヒト動物を飼育し、 該蛋白質を該動物中に生成 ·蓄積させ、 該動物中より該蛋白質を採取することにより、 該蛋白質を製造することができる。 該動物中の生成'蓄積場所としては、例えば、該動物のミルク (特開昭 63- 309192)、 卵等をあげることができる。 この際に用いられるプロモー夕一としては、 動物で 発現できるものであればいずれも用いることができるが、 例えば、 乳腺細胞特異 的なプロモー夕一であるひカゼインプロモータ一、 ?カゼインプロモ一夕一、 β ラクトグロブリンプロモーター、 ホエー酸性プロテインプロモ一夕一等が好適に 用いられる。  In the case of an animal individual, for example, a transgenic non-human animal into which the DNA encoding the protein of the present invention has been introduced is bred, the protein is produced and accumulated in the animal, and the protein is collected from the animal. By doing so, the protein can be produced. Examples of the place of production and accumulation in the animal include milk (eg, JP-A-63-309192) and eggs of the animal. As the promoter used at this time, any promoter that can be expressed in an animal can be used. For example, the casein promoter-1 and the casein promoter-1 which are breast cell-specific promoters can be used. , Β-lactoglobulin promoter, whey acidic protein promoter, etc. are preferably used.
植物個体を用いて本発明の蛋白質を製造する方法としては、 例えば本発明の蛋 白質をコードする D N Aを導入したトランスジエニック植物を公知の方法 〔組織 培養, 20 ( 1994)、 組織培養, 21 ( 1995 )、 Trends in Biotechnology, 15, 45 ( 1997)〕 に準じて栽培し、 該蛋白質を該植物中に生成 ·蓄積させ、 該植物中より該蛋白質 を採取することにより、 該蛋白質を生産する方法があげられる。  As a method for producing the protein of the present invention using a plant individual, for example, a transgenic plant into which a DNA encoding the protein of the present invention has been introduced can be prepared by a known method [tissue culture, 20 (1994); (1995), Trends in Biotechnology, 15, 45 (1997)], producing and accumulating the protein in the plant, and collecting the protein from the plant to produce the protein. There is a method.
本発明の形質転換体により製造された蛋白質を単離 ·精製する方法としては、 通常の酵素の単離、 精製法を用いることができる。  As a method for isolating and purifying the protein produced by the transformant of the present invention, a usual enzyme isolation and purification method can be used.
例えば、 本発明の蛋白質が、 細胞内に溶解状態で発現した場合には、 培養終了 後、 細胞を遠心分離により回収し、 水系緩衝液にけん濁後、 超音波破砕機、 フレ ンチプレス、 マントンガウリンホモゲナイザー、 ダイノミル等により細胞を破砕 し、 無細胞抽出液を得る。  For example, when the protein of the present invention is expressed in a dissolved state in cells, the cells are collected by centrifugation after completion of the culture, suspended in an aqueous buffer, and then sonicated with a sonicator, french press, Mentongaurin. Crush cells with a homogenizer, dynomill, etc. to obtain a cell-free extract.
該無細胞抽出液を遠心分離することにより得られる上清から、 通常の酵素の単 離精製法、 即ち、 溶媒抽出法、 硫安等による塩析法、 脱塩法、 有機溶媒による沈 殿法、 ジェチルアミノエチル (DEAE) —セファロース、 DIAI0N HPA- 75 (三菱化成 社製)等レジンを用いた陰イオン交換クロマトグラフィー法、 S- Sepharose FF From the supernatant obtained by centrifuging the cell-free extract, an ordinary enzyme isolation method, that is, a solvent extraction method, a salting out method using ammonium sulfate, a desalting method, a precipitation method using an organic solvent, Getylaminoethyl (DEAE)-Sepharose, DIAI0N HPA-75 (Mitsubishi Chemical), anion exchange chromatography using resin, S-Sepharose FF
(Pharmacia社製)等のレジンを用いた陽イオン交換クロマトグラフィ一法、 プチ ルセファロ一ス、 フエ二ルセファロ一ス等のレジンを用いた疎水性クロマトグラ フィ一法、 分子篩を用いたゲルろ過法、 ァフィ二ティークロマトグラフィー法、 クロマトフォーカシング法、 等電点電気泳動等の電気泳動法等の手法を単独ある レ、は組み合わせて用い、 精製標品を得ることができる。 (Pharmacia), etc. Hydrophobic chromatography using resins such as lucephalos and phenylsephalos, gel filtration using molecular sieve, affinity chromatography, chromatofocusing, electrofocusing, etc. Purified samples can be obtained by using techniques such as electrophoresis alone, in combination.
また、 該蛋白質が細胞内に不溶体を形成して発現した場合は、 同様に細胞を回 収後破砕し、 遠心分離を行うことにより得られた沈殿画分より、 通常の方法によ り該蛋白質を回収後、 該蛋白質の不溶体を蛋白質変性剤で可溶化する。  When the protein is expressed in an insoluble form in the cells, the cells are similarly collected, crushed, and centrifuged to obtain a precipitate fraction obtained by a conventional method. After recovering the protein, the insoluble form of the protein is solubilized with a protein denaturant.
該可溶化液を、 蛋白質変性剤を含まないあるいは蛋白質変性剤の濃度が蛋白質 が変性しない程度に希薄な溶液に希釈、 あるいは透析し、 該蛋白質を正常な立体 構造に構成させた後、 上記と同様の単離精製法により精製標品を得ることができ る。  After diluting or dialyzing the solubilized solution to a solution containing no protein denaturing agent or a diluting concentration of the protein denaturing agent so that the protein is not denatured, the protein is formed into a normal three-dimensional structure. A purified sample can be obtained by the same isolation and purification method.
本発明の蛋白質あるいはその糖修飾体等の誘導体が細胞外に分泌された場合に は、 培養上清に該蛋白質あるいはその糖鎖付加体等の誘導体を回収することがで きる。  When the protein of the present invention or a derivative such as a modified sugar thereof is secreted extracellularly, the protein or a derivative such as a sugar chain adduct thereof can be recovered in the culture supernatant.
即ち、 該培養物を上記と同様の遠心分離等の手法により処理することにより可 溶性画分を取得し、 該可溶性画分から、 上記と同様の単離精製法を用いることに より、 精製標品を得ることができる。  That is, a soluble fraction is obtained by treating the culture by a method such as centrifugation as described above, and a purified sample is obtained from the soluble fraction by using the same isolation and purification method as described above. Can be obtained.
このようにして取得される蛋白質として、 例えば、 配列番号 1に示されるアミ ノ酸配列を有する蛋白質をあげることができる。  Examples of the protein obtained in this manner include a protein having the amino acid sequence shown in SEQ ID NO: 1.
また、 本発明のポリペプチドを他の蛋白質との融合蛋白質として生産し、 融合 した蛋白質に親和性をもつ物質を用いたァフィニティーク口マトグラフィ一を利 用して精製することもできる。例えば、ロウらの方法〔Proc . Natl . Acad. Sci . USA, 86, 8227 ( 1989 )、 Genes Develop. , 4, 1288 ( 1990 )〕、特開平 05- 336963、 W094/23021 に記載の方法に準じて、 本発明のポリべプチドをプロティン Aとの融合蛋白質と して生産し、 ィムノグロプリン Gを用いるァフィ二ティ一クロマトグラフィ一に より精製することができる。  Alternatively, the polypeptide of the present invention can be produced as a fusion protein with another protein, and purified using affinity chromatography using a substance having an affinity for the fused protein. Natl. Acad. Sci. USA, 86, 8227 (1989), Genes Develop., 4, 1288 (1990), and the method described in JP-A-05-336963, W094 / 23021. Similarly, the polypeptide of the present invention can be produced as a fusion protein with protein A, and purified by affinity chromatography using immunoglobulin G.
また、 本発明のポリべプチドを F 1 a gぺプチドとの融合タンパク質として生 産し、 抗 F 1 a g抗体を用いるァフィ二ティ一クロマトグラフィ一により精製す ることができる CProc . Natl . Acad. Sci . USA, 86, 8227 ( 1989 )、 Genes Develop. , 4, 1288 ( 1990 )〕 。 更に、 該ポリペプチド自身に対する抗体を用いたァフィニテ ィ一クロマトグラフィーで精製することもできる。 Natl. Acad. Sci., Which produces the polypeptide of the present invention as a fusion protein with an F1ag peptide and can be purified by affinity chromatography using an anti-F1ag antibody. USA, 86, 8227 (1989), Genes Develop., 4, 1288 (1990)]. Further, an affinity using an antibody against the polypeptide itself is used. It can also be purified by HPLC.
上記で取得された蛋白質のアミノ酸情報を基に、 F m o c法 (フルォレニルメ チルォキシカルボニル法) 、 t B o c法 (t一ブチルォキシカルボニル法) 等の 化学合成法により、 本発明の蛋白質を製造することができる。 また、 Advanced ChemTech社、ノ 一キン -ェノレマ一社、 Pharmacia社、 Protein Technology Instrument 社、 Synthecell- Vega社、 PerSeptive社、 島津製作所等のペプチド合成機を利用し て化学合成することもできる。  Based on the amino acid information of the protein obtained above, the protein of the present invention is obtained by chemical synthesis methods such as the Fmoc method (fluorenylmethyloxycarbonyl method) and the tBoc method (t-butyloxycarbonyl method). Can be manufactured. Chemical synthesis can also be carried out using a peptide synthesizer such as Advanced ChemTech, Nokin-Kenolema, Pharmacia, Protein Technology Instrument, Synthecell-Vega, PerSeptive, Shimadzu.
3 . 本発明の蛋白質を認識する抗体の調製  3. Preparation of antibody recognizing the protein of the present invention
( 1 ) ポリクローナル抗体の作製  (1) Preparation of polyclonal antibody
本発明の蛋白質または該蛋白質の部分断片ポリべプチドの精製標品、 あるいは 本発明の蛋白質の一部のアミノ酸配列を有するぺプチドを抗原として用い、 動物 に投与することによりポリク口一ナル抗体を作製することができる。  Using a purified preparation of the protein of the present invention or a polypeptide fragment of the partial fragment of the protein, or a peptide having an amino acid sequence of a part of the protein of the present invention as an antigen, the polypeptide is administered to an animal to give a polypeptide antibody. Can be made.
投与する動物として、 ゥサギ、 ャギ、 ラット、 マウス、 ハムスター等を用いる ことができる。  Animals such as rabbits, goats, rats, mice, hamsters and the like can be used for the administration.
該抗原の投与量は動物 1匹当たり 5 0〜1 0 0〃gが好ましい。  The dose of the antigen is preferably 50 to 100 μg per animal.
ペプチドを用いる場合は、 ペプチドをスカシガイへモシァニン(keyhole 1 impet haemocyanin) ゃ牛チログ口プリン等のキヤリア蛋白に共有結合させたものを抗原 とするのが望ましい。 抗原とするペプチドは、 ペプチド合成機で合成することも できる。  When a peptide is used, it is desirable to use, as the antigen, a peptide covalently bonded to a carrier protein such as keyhole 1 impet haemocyanin or bovine tilogin purine. The peptide used as the antigen can also be synthesized using a peptide synthesizer.
該抗原の投与は、 1回目の投与の後 1〜2週間おきに 3〜1 0回行う。 各投与後、 3〜 7日目に眼底静脈叢より採血し、 該血清が免疫に用いた抗原と反応すること を酵素免疫測定法〔酵素免疫測定法 (E L I S A法) :医学書院刊 ( 1 9 7 6年)、 Antibodies - A Laboratory Manual, Cold Spring Harbor Laboratory ( 1988)〕 等 で確認する。  The administration of the antigen is performed 3 to 10 times every 1 to 2 weeks after the first administration. Blood is collected from the fundus venous plexus on the 3rd to 7th days after each administration, and the reaction of the serum with the antigen used for immunization is determined by an enzyme immunoassay [enzyme immunoassay (ELISA): published by Medical Shoin (19) 76 years), Antibodies-A Laboratory Manual, Cold Spring Harbor Laboratory (1988)].
免疫に用いた抗原に対し、 その血清が充分な抗体価を示した非ヒト哺乳動物よ り血清を取得し、 該血清より、 下記方法によりポリクロ一ナル抗体を分離、 精製 することができる。  Serum is obtained from a non-human mammal whose serum shows a sufficient antibody titer against the antigen used for immunization, and a polyclonal antibody can be separated and purified from the serum by the following method.
抗体を分離、 精製する方法としては、 遠心分離、 4 0〜5 0 %飽和硫酸アンモ ニゥムによる塩析、 力プリル酸沈殿 〔Antibodies, A Laboratory manual, Cold Spring Harbor Laboratory, ( 1988)〕 、 または D E A E—セファロースカラム、 陰イオン交換カラム、 プロテイン A—カラム、 プロテイン G—カラムあるいはゲ ル濾過カラム等を用いるクロマトグラフィー等を、 単独または組み合わせて処理 する方法があげられる。 Methods for separating and purifying antibodies include centrifugation, salting out with 40-50% saturated ammonium sulfate, and prillic acid precipitation [Antibodies, A Laboratory manual, Cold Spring Harbor Laboratory, (1988)], or DEAE. —Sepharose column, Examples of the method include a method in which chromatography using an anion exchange column, a protein A-column, a protein G-column, a gel filtration column, or the like is used alone or in combination.
(2) モノクローナル抗体の作製  (2) Preparation of monoclonal antibody
(a)抗体産性細胞の調製  (a) Preparation of antibody-producing cells
上記(1)において、 免疫に用いた抗原に対し、 その血清が十分な抗体価を示した ラッ トを抗体産生細胞の供給源として供する。  In the above (1), a rat whose serum shows a sufficient antibody titer against the antigen used for immunization is used as a source of antibody-producing cells.
該抗体価を示したラッ卜に抗原物質を最終投与した後 3〜7日目に、 脾臓を摘 出する。  Three to seven days after the final administration of the antigenic substance to the rat showing the antibody titer, the spleen is removed.
該脾臓を MEM培地 (日水製薬社製) 中で細断し、 ピンセットでほぐし、 1 , 200 r pmで 5分間遠心分離した後、 上清を捨てる。  The spleen is shredded in a MEM medium (manufactured by Nissui Pharmaceutical Co., Ltd.), loosened with forceps, centrifuged at 1,200 rpm for 5 minutes, and the supernatant is discarded.
得られた沈殿画分の脾細胞をトリスー塩化アンモニゥム緩衝液 (pH7. 65) で 1〜2分間処理し赤血球を除去した後、 MEM培地で 3回洗浄し、 得られた脾 細胞を抗体産生細胞として用いる。  The spleen cells in the resulting precipitate fraction are treated with Tris-ammonium chloride buffer (pH 7.65) for 1 to 2 minutes to remove red blood cells, and then washed three times with MEM medium. Used as
(b)骨髄腫細胞の調製  (b) Preparation of myeloma cells
骨髄腫細胞としては、 マウスまたはラッ卜から取得した株化細胞を使用する。 例えば、 8—ァザグァニン耐性マウス(BALB/c由来)骨髄腫細胞株 P3- X63Ag8- U1 (以 下、 P 3— U 1と略す) 〔Curr. Topics. Microbiol. Immunol., 81, 1 (1978)、 Europ. J. Immunol., 6, 511 (1976)〕、 SP2/0-Agl4(SP-2) CNature, 276, 269 (1978)〕、 P3-X63-Ag8653(653)〔J. Immunol., 123, 1548 (1979)〕、 P3-X63-Ag8(X63) (Nature, 256, 495 (1975)〕 等を用いることができる。 これらの細胞株は、 8—ァザグァニ ン培地 〔RPMI— 1640培地にグルタミン ( 1. 5麵 ol/l) 、 2—メルカプ トエタノール (5 x 10-5mol/l) 、 ジェン夕マイシン ( 10〃g/ml) および 牛胎児血清 (FCS) (CSL社製、 10%) を加えた培地 (以下、 正常培地と いう) に、 さらに 8—ァザグァニン ( 15〃g/ml) を加えた培地〕 で継代す るが、 細胞融合の 3〜4日前に正常培地で培養し、 融合には該細胞を 2 X 107個 以上用いる。 As myeloma cells, cell lines obtained from mice or rats are used. For example, 8-azaguanine-resistant mouse (derived from BALB / c) myeloma cell line P3-X63Ag8-U1 (hereinafter abbreviated as P3-U1) [Curr. Topics. Microbiol. Immunol., 81, 1 (1978) , Europ.J. Immunol., 6, 511 (1976)), SP2 / 0-Agl4 (SP-2) CNature, 276, 269 (1978)), P3-X63-Ag8653 (653) (J. Immunol., 123, 1548 (1979)], P3-X63-Ag8 (X63) (Nature, 256, 495 (1975)), etc. These cell lines can be used in an 8-azaguanine medium [RPMI-1640 medium]. glutamine (1.5 noodles ol / l), 2-mercaptoethanol (5 x 10- 5 mol / l ), Jen evening puromycin (10〃G / ml) and fetal calf serum (FCS) (CSL Ltd., 10 %) And a medium supplemented with 8-azaguanine (15 μg / ml)], but 3-4 days before cell fusion, Culture and use 2 × 10 7 or more of the cells for fusion.
(c)ハイプリ ドーマの作製  (c) Preparation of Hypri-Doma
(a)で取得した抗体産生細胞と(b)で取得した骨髄腫細胞を MEM培地または P B S (リン酸ニナトリウム 1. 83 g、 リン酸一カリウム 0. 21 g、 食塩 7. 65 g、 蒸留水 1リットル、 pH7. 2) でよく洗浄し、 細胞数が、 抗体産生細 胞:骨髄腫細胞 =5〜: L 0 : 1になるよう混合し、 1, 200 rpmで 5分間遠 心分離した後、 上清を捨てる。 The antibody-producing cells obtained in (a) and the myeloma cells obtained in (b) were added to a MEM medium or PBS (1.83 g of disodium phosphate, 0.21 g of monopotassium phosphate, salt 7. Wash well with 65 g, 1 liter of distilled water, pH 7.2), mix the cells so that the number of antibody-producing cells: myeloma cells = 5-: L 0: 1, and mix at 1,200 rpm for 5 minutes. After centrifugation, discard the supernatant.
得られた沈澱画分の細胞群をよくほぐし、 該細胞群に、 攪拌しながら、 37°C で、 108抗体産生細胞あたり、 ポリエチレングライコ一ルー 1000 (PEG— 1000) 2 g、 MEM 2mlおよびジメチルスルホキシド (DM SO) 0. 7 mlを混合した溶液を 0. 2〜 lml添加し、 さらに 1〜 2分間毎に MEM培地 1〜2mlを数回添加する。 Thoroughly loosened The resulting precipitated fraction of cell groups, the said group of cells, with stirring, at 37 ° C, 10 8 antibody-producing cells per polyethylene glyco one Roux 1000 (PEG- 1000) 2 g, MEM 2ml and Add 0.2 to 1 ml of a mixture of 0.7 ml of dimethyl sulfoxide (DMSO), and add 1 to 2 ml of MEM medium several times every 1 to 2 minutes.
添加後、 MEM培地を加えて全量が 50mlになるように調製する。 該調製液 を 900 r p mで 5分間遠心分離後、 上清を捨てる。  After addition, add MEM medium to adjust the total volume to 50 ml. After centrifuging the preparation at 900 rpm for 5 minutes, discard the supernatant.
得られた沈殿画分の細胞を、 ゆるやかに ίまぐした後、 メスピペットによる吸込 み、 吹出しでゆるやかに HAT培地〔正常培地にヒポキサンチン ( 1 (T4mol/l)、 チミジン ( 1. 5 x 10—5mol/l) およびアミノプテリン (4x 10—¾ol八) を加 えた培地〕 100ml中に懸濁する。 The cells in the precipitate fraction obtained are gently sprinkled, and then slowly sucked and blown out with a female pipette. The HAT medium [in a normal medium, hypoxanthine (1 (T 4 mol / l), thymidine (1.5) x 10- 5 mol / l) and suspended aminopterin (4x 10-¾ol eight) to the pressurizing example was medium] in 100 ml.
該懸濁液を 96穴培養用プレートに 100〃1/穴ずつ分注し、 5%C02イン キュベ—夕—中、 37°Cで 7~ 14日間培養する。 The suspension was dispensed by 100〃1 / well to the plate for a 96-well culture, 5% C0 2 incuba- - evening - in cultured for 7-14 days at 37 ° C.
培養後、 培養上清の一部をとりアンチボディィズ 〔Antibodies, A Laboratory manual , Cold Spring Harbor Laboratory, Chapter 14 (1988)〕 等に述べられて いる酵素免疫測定法により、 本発明の蛋白質の部分断片ポリべプチドに特異的に 反応するハイプリ ドーマを選択する。  After the culturing, a part of the culture supernatant is taken to obtain the protein of the present invention by an enzyme immunoassay described in Antibodies, A Laboratory manual, Cold Spring Harbor Laboratory, Chapter 14 (1988). Select hybridomas that react specifically with partial fragment polypeptides.
酵素免疫測定法の具体的例として、 以下の方法をあげることができる。  The following method can be given as a specific example of the enzyme immunoassay.
免疫の際、 抗原に用いた本発明の蛋白質の部分断片ポリべプチドを適当なプレ ートにコートし、 ハイプリ ドーマ培養上清もしくは後述の(d)で得られる精製抗体 を第一抗体として反応させ、 さらに第二抗体としてピオチン、 酵素、 化学発光物 質あるいは放射線化合物等で標識した抗ラットまたは抗マウスィムノグロブリン 抗体を反応させた後に標識物質に応じた反応を行い、 本発明の蛋白質に特異的に 反応するものを本発明のモノクローナル抗体を生産するハイプリ ドーマとして選 択する。  At the time of immunization, the partial fragment polypeptide of the protein of the present invention used as the antigen was coated on an appropriate plate, and reacted with the hybridoma culture supernatant or the purified antibody obtained in (d) described below as the first antibody. Further, after reacting an anti-rat or anti-mouse immunoglobulin antibody labeled with a biotin, an enzyme, a chemiluminescent substance or a radioactive compound as a second antibody, a reaction according to the labeling substance is carried out, and the protein of the present invention is reacted. Those that react specifically are selected as hybridomas producing the monoclonal antibody of the present invention.
該ハイプリ ドーマを用いて、限界希釈法によりクローニングを 2回繰り返し〔 1 回目は、 HT培地 (HAT培地からアミノプテリンを除いた培地) 、 2回目は、 正常培地を使用する〕 、 安定して強い抗体価の認められたものを本発明のモノク 口一ナル抗体を産生するハイプリ ド一マ株として選択する。 Using the hybridoma, cloning was repeated twice by the limiting dilution method (the first time was HT medium (medium obtained by removing aminopterin from the HAT medium), and the second time was A normal medium is used], and those with a stable and high antibody titer are selected as hybridoma strains producing the monoclonal antibody of the present invention.
(d)モノクローナル抗体の調製  (d) Preparation of monoclonal antibody
プリスタン処理 〔2, 6, 10, 14—テトラメチルペン夕デカン (P r i s t ane) 0. 5mlを腹腔内投与し、 2週間飼育する〕 した 8〜10週令のマ ウスまたはヌードマウスに、 ( c )で取得した本発明の蛋白質モノクロ一ナル抗体産 生ハイプリ ドーマ細胞 5〜20 X 106細胞ノ匹を腹腔内に注射する。 10〜21 日間でハイプリ ドーマは腹水癌化する。 Pristane-treated [0.5 ml of 2,6,10,14-tetramethylpenedecane (Pristane) was intraperitoneally administered and bred for 2 weeks]. c) Intraperitoneally inject 5 to 20 × 10 6 cells of the hybridoma cells producing the monoclonal antibody of the present invention obtained in the step c). In 10 to 21 days, Hypridoma develops ascites cancer.
該腹水癌化したマウスから腹水を採取し、 3000 rpmで 5分間遠心分離し て固形分を除去する。  The ascites is collected from the mouse with ascites tumor and centrifuged at 3000 rpm for 5 minutes to remove solids.
得られた上清より、 ポリクローナル抗体で用いた方法と同様の方法でモノクロ —ナル抗体を精製、 取得することができる。  From the obtained supernatant, a monoclonal antibody can be purified and obtained in the same manner as the method used for the polyclonal antibody.
抗体のサブクラスの決定は、 マウスモノクローナル抗体夕イビングキットまた はラットモノクローナル抗体タイピングキットを用いて行う。 蛋白質量は、 ロー リー法あるいは 280 nmでの吸光度より算出する。  The antibody subclass is determined using a mouse monoclonal antibody evening kit or a rat monoclonal antibody typing kit. The protein content is calculated by the Lowry method or from the absorbance at 280 nm.
抗体のクラスとは抗体のアイソタイプのことで、 ヒトでは、 IgG、 IgA、 IgM、 IgD、 IgEがあげられる。 サブクラスとは、 クラス内のアイソタイプのことで、 マウスで は、 IgGK IgG2aヽ IgG2b、 IgG3、 ヒトでは、 IgGl、 IgG2、 IgG3、 IgG4があげられ る。  An antibody class refers to an antibody isotype, and in humans, includes IgG, IgA, IgM, IgD, and IgE. The subclass refers to an isotype within the class. In a mouse, IgGK IgG2a ヽ IgG2b, IgG3, and in a human, IgGl, IgG2, IgG3, and IgG4.
4. 本発明の DNA、 蛋白質または抗体の利用  4. Use of DNA, protein or antibody of the present invention
(1) 本発明の DN Aまたはオリゴヌクレオチドを用いた、 ハイプリダイゼー シヨン法、 またはポリメラ一ゼ ·チエイン · リアクション (P CR) 法による遺 伝子発現の検出  (1) Detection of gene expression by the hybridization method or the polymerase chain reaction (PCR) method using the DNA or oligonucleotide of the present invention.
本発明の D N Aまたはオリゴヌクレオチドを用い、 ノーザンハイブリダィゼ一 シヨン法 (モレキュラー 'クロ一ニング第 2版)、 PCR法および RT (reverse- transcribed) 一 PCR法 〔ともに PCR Protocols, Academic Press (1990)〕 (以 上、 併せて PCR法ともいう) 等を行うことにより、 本発明の蛋白質をコードす る mRN Aの発現を検出することができる。 このうち、 RT— PCR法は簡便な 方法であるため、 検出方法として特に有用である。  Using the DNA or oligonucleotide of the present invention, the Northern hybridization method (Molecular 'Cloning 2nd edition), the PCR method and the RT (reverse-transcribed) -PCR method (both PCR Protocols, Academic Press (1990) )] (Hereinafter, also referred to as PCR method), etc., to detect the expression of mRNA encoding the protein of the present invention. Of these, the RT-PCR method is a simple method, and is particularly useful as a detection method.
該検出方法は、 遺伝子の発現量の定量にも用いられ、 また炎症、 癌、 骨粗鬆症、 糖尿病、 腎疾患等、 本発明の蛋白質をコードする遺伝子の変異が原因となってい る疾患の診断に利用することができる。 The detection method is also used for quantifying the expression level of a gene, and is used for inflammation, cancer, osteoporosis, It can be used for diagnosis of diseases caused by mutation of the gene encoding the protein of the present invention, such as diabetes and kidney disease.
(2) 本発明の DN Aまたはオリゴヌクレオチドを用いた、 ハイブリダィゼ一 シヨン法または P C R法による、 本発明の蛋白質をコードする遺伝子の変異の検 出  (2) Detection of mutation of the gene encoding the protein of the present invention by the hybridization method or the PCR method using the DNA or the oligonucleotide of the present invention.
本発明のオリゴヌクレオチドをプロ一ブとして、 ゲノム DN Aに対してサザン ハイブリダィゼ一シヨン法 (モレキュラー 'クロ一ニング第 2版) 、 PCR法等 を行うことにより、 本発明の蛋白質をコードする遺伝子の変異を検出することが できる。  By using the oligonucleotide of the present invention as a probe and subjecting the genome DNA to a Southern hybridization method (Molecular 'Cloning 2nd edition), a PCR method, or the like, the gene encoding the protein of the present invention can be obtained. Mutations can be detected.
該検出方法は、 炎症、 癌、 骨粗鬆症、 糖尿病、 腎疾患等、 本発明の蛋白質をコ ードする遺伝子の変異が原因となっている疾患の診断に利用することができる。  The detection method can be used for diagnosis of diseases caused by mutation of the gene encoding the protein of the present invention, such as inflammation, cancer, osteoporosis, diabetes, and kidney disease.
(3) 本発明の DN Aまたはオリゴヌクレオチドを用いた本発明の蛋白質をコ 一ドする遺伝子の転写または翻訳の抑制  (3) Suppression of transcription or translation of a gene encoding the protein of the present invention using the DNA or oligonucleotide of the present invention
本発明の DNAは、 アンチセンス RNA/DNA技術 〔バイオサイエンスとィ ンダストリー, 50, 322 (1992)、 化学, 46, 681 (1991)、 Biotechnology, 9, 358 (1992)、 Trends in Biotechnology, 10, 87 (1992)、 Trends in Biotechnology, 10, 152 (1992)、細胞工学, i£, 1463 (1997)〕、 トリプル.ヘリックス技術 !: Trends in Biotechnology, 10, 132 (1992)〕 等を用い、 本発明の蛋白質をコードする遺 伝子の転写または翻訳を抑制することができる。  The DNA of the present invention can be prepared by antisense RNA / DNA technology [Bioscience and Industry, 50, 322 (1992), Chemistry, 46, 681 (1991), Biotechnology, 9, 358 (1992), Trends in Biotechnology, 10, 87 (1992), Trends in Biotechnology, 10, 152 (1992), Cell Engineering, i £, 1463 (1997)], Triple Helix Technology !: Trends in Biotechnology, 10, 132 (1992)] The transcription or translation of the gene encoding the protein of the present invention can be suppressed.
例えば、 本発明の DN Aまたはオリゴヌクレオチドを投与することにより、 本 発明の蛋白質をコードする遺伝子の転写、 または本発明の蛋白質をコードする m RNAの翻訳を、 それぞれ抑制でき、 本発明の蛋白質の生産を抑制することがで きる。  For example, by administering the DNA or oligonucleotide of the present invention, transcription of a gene encoding the protein of the present invention or translation of mRNA encoding the protein of the present invention can be suppressed, respectively. Production can be suppressed.
該抑制方法は、 炎症、 癌、 骨粗鬆症、 糖尿病、 腎疾患等、 本発明の蛋白質をコ —ドする遺伝子の変異が原因となっている疾患の治療または予防に利用すること ができる。  The method can be used for treating or preventing inflammation, cancer, osteoporosis, diabetes, kidney disease, and other diseases caused by mutations in the gene encoding the protein of the present invention.
本発明の蛋白質をコードする遺伝子の変異が原因となっている疾患の治療方法 としては、 患者から取り出した細胞に、 遺伝子治療用に適切に調製した本発明の 組換えベクターを導入した後、 細胞を生体内に戻すことにより、 また適当なレト ロウィルス、 アデノウイルス、 アデノ随伴ウィルス、 単純へルぺスウィルス、 レ ンチウイルス等のウイルスベクターに乗せて生体に投与することにより、 さらに リボソーム等の人工的なべシクル構造に封入して生体に投与することにより、 本 発明の蛋白質を患者の生体内で発現させる方法が用いられる。 As a method for treating a disease caused by a mutation in the gene encoding the protein of the present invention, cells obtained after introducing the recombinant vector of the present invention appropriately prepared for gene therapy into cells taken out of a patient, By returning the virus to the body, it is also possible to obtain appropriate retroviruses, adenoviruses, adeno-associated viruses, simple virus, A method for expressing the protein of the present invention in a patient's living body by administering it to a living body by carrying it on a viral vector such as an antivirus or by further encapsulating it in an artificial vesicle structure such as ribosome. Used.
なお、 本発明の蛋白質、 例えば ERR4は hERR2と高い相同性を示すが、 hERR 2はグルココルチコィ ド等の抗炎症作用を阻害する可能性がある 〔J. Biol. Chem. , 271, 9879 (1996)〕 ことから、 Ε R R 4の発現を抑制すると抗炎症 物質の作用を増強できることが推察される。 また、 ERR4は腎臓で発現してい ることから、 炎症または腎疾患、 例えば腎炎の有効な治療薬となり得る。  In addition, the protein of the present invention, for example, ERR4 shows high homology with hERR2, but hERR2 may inhibit anti-inflammatory effects such as glucocorticoid (J. Biol. Chem., 271, 9879 (1996) Therefore, it is inferred that suppressing the expression of RR4 can enhance the action of anti-inflammatory substances. In addition, since ERR4 is expressed in the kidney, it can be an effective therapeutic agent for inflammation or renal diseases such as nephritis.
(4) 本発明の DNAまたはオリゴヌクレオチドを用いた本発明の蛋白質をコ ―ドする遺伝子のプロモー夕一領域の取得  (4) Obtaining the promoter region of the gene encoding the protein of the present invention using the DNA or oligonucleotide of the present invention
本発明の DN Aまたはオリゴヌクレオチドをプローブとして、 公知の方法 〔東 京大学医科学研究所制癌研究部編、 新細胞工学実験プロトコール、 秀潤社 ( 19 93年) 〕 を用いて、 該遺伝子のプロモ一夕一領域を取得することが可能である。 プロモータ—領域としては、 哺乳動物細胞において本発明の蛋白質をコ一ドす る遺伝子の転写に関与するすべてのプロモーター領域があげられる。 例えば、 ヒ 卜の腎臓、 胎盤、 心臓、 脳、 腎臓、 骨格筋または脬臓、 特に腎臓または胎盤で、 本発明の蛋白質をコードする遺伝子の転写に関与するプロモー夕一領域をあげる ことができる。 該プロモータ一は後述のスクリ一ニング方法に利用することがで きる。  Using the DNA or oligonucleotide of the present invention as a probe and the known method [New Cell Engineering Experimental Protocol, Shujunsha (1993), edited by the Cancer Research Division, Institute of Medical Science, The University of Tokyo, Shujunsha (1993)] It is possible to get the Promos overnight area. Examples of the promoter region include all promoter regions involved in transcription of a gene encoding a protein of the present invention in mammalian cells. For example, in human kidney, placenta, heart, brain, kidney, skeletal muscle or kidney, in particular, kidney or placenta, a promoter region involved in transcription of a gene encoding the protein of the present invention can be mentioned. The promoter can be used for a screening method described later.
(5) 本発明の蛋白質を用いた該蛋白質と特異的に結合するリガンドのスクリ —ニング  (5) Screening of a ligand which specifically binds to the protein using the protein of the present invention
本発明の蛋白質と直接結合する物質を同定する方法、 該蛋白質の立体構造に基 づく薬剤デザインの方法等の方法 〔Charifson. P. S., Practical Application of Computer-Aided Drug Design, Marcel Dekker (1997)〕 を用いることにより、 該 蛋白質と特異的に結合するリガンドをスクリーニングすることができる。 該リガ ンドは本発明の蛋白質が関与する疾患の治療薬、 または本発明の蛋白質が関与す る情報伝達系や生体機能に関する研究に利用することができる。  A method for identifying a substance that directly binds to the protein of the present invention and a method for designing a drug based on the three-dimensional structure of the protein [Charifson. PS, Practical Application of Computer-Aided Drug Design, Marcel Dekker (1997)] By using it, a ligand that specifically binds to the protein can be screened. The ligand can be used as a therapeutic agent for a disease associated with the protein of the present invention or a study on a signal transduction system or a biological function involving the protein of the present invention.
( 6 ) 本発明の形質転換体を用いた本発明の蛋白質をコ一ドする遺伝子の発現 の解析  (6) Analysis of expression of a gene encoding the protein of the present invention using the transformant of the present invention
本発明の形質転換体を種々の被験物質と共存させ、 該形質転換体における遺伝 子の発現レベルを解析することにより、 本発明の蛋白質をコードする遺伝子の転 写を制御する物質、 本発明の蛋白質による転写制御機能に関与する物質、 または 本発明の蛋白質により転写制御を受ける遺伝子をスクリーニングすることができ る。 The transformant of the present invention is allowed to coexist with various test substances; A substance that controls the transcription of a gene encoding the protein of the present invention by analyzing the expression level of the offspring, a substance that participates in the transcription control function of the protein of the present invention, or a gene that is transcriptionally controlled by the protein of the present invention. Can be screened.
また、 既存の核内レセプ夕一をコ一ドする遺伝子やその他の転写因子遺伝子と 共発現させることにより、 それらとの相互作用を検定することができ、 同時にこ れら遺伝子との相互作用に関与する薬剤をスクリーニングすることができる。 これらの方法において、 既存の核内レセプ夕一や転写因子の標的 D N A配列を 転写制御領域に有する C A T (クロラムフエニコールァセチルトランスフェラ一 ゼ)遺伝子、 G F P (グリーンフルオレセントプロテイン)遺伝子、 ルシフェラ一ゼ 遺伝子、 ? -ガラクトシダ一ゼ遺伝子等々のレポ一夕一遺伝子を用いると、 スクリ —ニング効率をあげることができる。  In addition, by co-expressing existing nuclear receptor genes and other transcription factor genes, the interaction with them can be assayed, and at the same time, the interaction with these genes can be examined. The drugs involved can be screened. In these methods, the CAT (chloramphenicol acetyltransferase) gene, which has the target DNA sequence of the existing nuclear receptor or a transcription factor in the transcription control region, the GFP (green fluorescent protein) gene, Screening efficiency can be increased by using repo alleles such as luciferase gene and? -Galactosidase gene.
( 7 ) 本発明の抗体を用いた本発明の蛋白質の免疫学的検出  (7) Immunological detection of the protein of the present invention using the antibody of the present invention
本発明の抗体を用い、 抗原抗体反応を行わせることにより、 本発明の蛋白質ま たは該蛋白質を含む組織を免疫学的に検出することができる。  By performing an antigen-antibody reaction using the antibody of the present invention, the protein of the present invention or a tissue containing the protein can be immunologically detected.
該検出法は、 炎症、 癌、 骨粗鬆症、 糖尿病、 腎疾患等、 本発明の蛋白質をコー ドする遺伝子の変異が原因となっている疾患の診断に利用することができる。 また、 該検出方法は、 蛋白質の定量にも用いることができる。  The detection method can be used for diagnosis of diseases caused by mutation of the gene encoding the protein of the present invention, such as inflammation, cancer, osteoporosis, diabetes, and kidney disease. The detection method can also be used for protein quantification.
免疫学的に検出する方法としては、 マイクロタイ夕一プレートを用いる E Lェ S A法、 蛍光抗体法、 ゥヱスタンプロット法、 免疫組織染色法等をあげることが できる。  Methods for immunological detection include the ELISA assay using a microtiter plate, a fluorescent antibody method, a stamp lot method, and an immunohistological staining method.
免疫学的に定量する方法としては、 液相中で本発明の蛋白質と反応する抗体の うちェピトープが異なる 2種類のモノクローナル抗体を用いたサンドイッチ E L I S A法、 12SI等の放射性同位体で標識した本発明の蛋白質と本発明の蛋白質を認 識する抗体とを用いるラジオィムノアツセィ法等があげられる。 Methods for immunological quantification include sandwich ELISA using two types of monoclonal antibodies having different epitopes among antibodies that react with the protein of the present invention in the liquid phase, and a method of labeling with a radioisotope such as 12S I. A radioimmunoassay method using the protein of the present invention and an antibody recognizing the protein of the present invention can be exemplified.
( 8 ) 本発明の抗体を含有する医薬  (8) a medicine containing the antibody of the present invention
本発明の抗体は、 医薬、 例えば炎症、 癌、 骨粗鬆症、 糖尿病、 腎疾患等、 本発 明の蛋白質をコードする遺伝子の変異が原因となっている疾患の治療薬として用 いることができる。  The antibody of the present invention can be used as a medicament, for example, a therapeutic agent for diseases caused by mutations in the gene encoding the protein of the present invention, such as inflammation, cancer, osteoporosis, diabetes, and kidney disease.
本発明の抗体を含有する医薬は、 治療薬として該化合物単独で投与することも 可能ではあるが、 通常は薬理学的に許容される一つあるいはそれ以上の担体と一 緒に混合し、 製剤学の技術分野においてよく知られる任意の方法により製造した 医薬製剤として提供するのが望ましい。 The medicament containing the antibody of the present invention may be administered alone as a therapeutic agent. Although possible, it is usually admixed with one or more pharmacologically acceptable carriers to provide a pharmaceutical preparation produced by any method well known in the pharmaceutical arts. desirable.
投与経路は、 治療に際して最も効果的なものを使用するのが望ましく、 経口投 与、 または口腔内、 気道内、 直腸内、 皮下、 筋肉内および静脈内等の非経口投与 をあげることができる。 投与形態としては、 噴霧剤、 カプセル剤、 錠剤、 顆粒剤、 シロップ剤、 乳剤、 座剤、 注射剤、 軟膏、 テープ剤等があげられる。  It is desirable to use the most effective route for treatment, including oral administration or parenteral administration such as buccal, respiratory, rectal, subcutaneous, intramuscular and intravenous. Dosage forms include sprays, capsules, tablets, granules, syrups, emulsions, suppositories, injections, ointments, tapes and the like.
経口投与に適当な製剤としては、 乳剤、 シロップ剤、 カプセル剤、 錠剤、 散剤、 顆粒剤等があげられる。 例えば乳剤およびシロップ剤のような液体調製物は、 水、 ショ糖、 ソルビトール、 果糖等の糖類、 ポリエチレングリコール、 プロピレング リコール等のグリコール類、 ごま油、 ォリーブ油、 大豆油等の油類、 p—ヒドロ キシ安息香酸エステル類等の防腐剤、 ストロベリーフレーバー、 ペパーミント等 のフレーバー類等を添加剤として用いて製造できる。 カプセル剤、 錠剤、 散剤、 顆粒剤等は、 乳糖、 ブドウ糖、 ショ糖、 マンニトール等の賦形剤、 デンプン、 ァ ルギン酸ナトリゥム等の崩壊剤、 ステアリン酸マグネシウム、 タルク等の滑沢剤、 ポリビニルアルコール、 ヒドロキシプロピルセルロース、 ゼラチン等の結合剤、 脂肪酸エステル等の界面活性剤、 グリセリン等の可塑剤等を添加剤として用いて 製造できる。  Formulations suitable for oral administration include emulsions, syrups, capsules, tablets, powders, granules and the like. For example, liquid preparations such as emulsions and syrups include water, sugars such as sucrose, sorbitol, fructose, glycols such as polyethylene glycol and propylene glycol, oils such as sesame oil, olive oil, soybean oil, p- It can be produced using preservatives such as hydroxybenzoic acid esters and flavors such as strawberry flavor and peppermint as additives. Capsules, tablets, powders, granules, etc. are excipients such as lactose, glucose, sucrose, mannitol, disintegrants such as starch, sodium alginate, lubricants such as magnesium stearate, talc, polyvinyl alcohol And a binder such as hydroxypropylcellulose and gelatin, a surfactant such as a fatty acid ester, and a plasticizer such as glycerin as additives.
非経口投与に適当な製剤としては、 注射剤、 座剤、 噴霧剤等があげられる。 例 えば、 注射剤は、 塩溶液、 ブドウ糖溶液、 あるいは両者の混合物からなる担体等 を用いて調製する。 座剤はカカオ脂、 水素化脂肪またはカルボン酸等の担体を用 いて調製される。 また、 噴霧剤は該化合物そのもの、 ないしは受容者の口腔およ び気道粘膜を刺激せず、 かつ該化合物を微細な粒子として分散させ吸収を容易に させる担体等を用いて調製する。 担体として具体的には乳糖、 グリセリン等が例 示される。 該化合物および用いる担体の性質により、 エアロゾル、 ドライバウダ 一等の製剤が可能である。 また、 これらの非経口剤においても経口剤で添加剤と して例示した成分を添加することもできる。  Formulations suitable for parenteral administration include injections, suppositories, sprays and the like. For example, an injection is prepared using a carrier comprising a salt solution, a glucose solution, or a mixture of both. Suppositories are prepared using carriers such as cocoa butter, hydrogenated fats or carboxylic acids. Sprays are prepared using the compound itself or a carrier which does not irritate the oral and respiratory mucosa of the recipient and which disperses the compound as fine particles to facilitate absorption. Specific examples of the carrier include lactose, glycerin and the like. Formulations such as aerosols and dry powders are possible depending on the properties of the compound and the carrier used. In these parenteral preparations, the components exemplified as additives for oral preparations can also be added.
投与量または投与回数は、 目的とする治療効果、 投与方法、 治療期間、 年齢、 体重等により異なるが、 通常成人 1日当たり 10 / g/kg〜8 mg/kgである。  The dose or frequency of administration varies depending on the desired therapeutic effect, administration method, treatment period, age, body weight, etc., but is usually 10 / g / kg to 8 mg / kg per day for an adult.
( 9 ) 本発明の D N Aを用いたノックァゥト非ヒト動物の作製 本発明の D N Aを含有してなる組換えベクターを用い、 目的とする非ヒト動物、 例えばゥシ、ヒヅジ、ャギ、ブ夕、ゥマ、マウス、ニヮトリ等の胚性幹細胞(embryonic stem cell )において、 染色体上の本発明の蛋白質をコードする遺伝子を公知の相 同組換えの手法〔例えば、 Nature, 326, 6110, 295 ( 1987)、 Cell, 51, 3, 503 ( 1987) 等〕 により不活化または任意の配列と置換した変異クローンを作製する 〔例えば、 Nature, 350, 6315, 243 ( 1991 )〕。 (9) Preparation of knockout non-human animal using DNA of the present invention Using a recombinant vector containing the DNA of the present invention, an objective non-human animal, for example, an embryonic stem cell such as a sea lion, a sheep, a goat, a bush, a horse, a mouse, a chicken, etc. In the above, the gene encoding the protein of the present invention on the chromosome is mutated by a known homologous recombination technique (eg, Nature, 326, 6110, 295 (1987), Cell, 51, 3, 503 (1987), etc.). A mutant clone is prepared which is activated or substituted with an arbitrary sequence [for example, Nature, 350, 6315, 243 (1991)].
該胚性幹細胞の変異クローンを用い、 動物の受精卵の胚盤胞 (blastcyst)への注 入キメラ法または集合キメラ法等の手法により、 胚性幹細胞クローンと正常細胞 からなるキメラ個体を調製することができる。  Using the mutant clone of the embryonic stem cell, a chimeric individual consisting of an embryonic stem cell clone and normal cells is prepared by a method such as chimera method or chimera method in which an embryo fertilized egg of an animal is injected into a blastocyst (blastcyst). be able to.
該キメラ個体と正常個体の掛け合わせにより、 全身の細胞の染色体上の本発明 の蛋白質をコードする遺伝子に任意の変異を有する個体を得ることができ、 さら にその個体の掛け合わせにより相同染色体の双方に変異が入ったホモ個体の中か ら、 本発明の蛋白質をコードする遺伝子の発現が一部または完全に抑制された個 体としてノックアウト非ヒト動物を得ることができる。  By crossing the chimeric individual with a normal individual, an individual having an arbitrary mutation in the gene encoding the protein of the present invention on the chromosome of the whole body can be obtained. A knockout non-human animal can be obtained as an individual in which the expression of the gene encoding the protein of the present invention is partially or completely suppressed from homozygous individuals in which both have mutations.
また、 染色体上の本発明の蛋白質をコ一ドする遺伝子の任意の位置へ変異を導 入することにより、 ノックァゥト非ヒト動物を作製することも可能である。 例え ば染色体上の本発明の蛋白質をコードする遺伝子の翻訳領域中へ塩基を置換、 欠 失、 挿入等させて変異を導入することにより、 その産物の活性を改変させること も可能である。 また、 その発現制御領域への同様な変異を導入することにより、 発現の程度、 時期、組織特異性等を改変させることも可能である。さらに Cre- ΙοχΡ 系との組合せにより、 より積極的に発現時期、 発現部位、 発現量等を制御するこ とも可能である。  In addition, by introducing a mutation into an arbitrary position of the gene encoding the protein of the present invention on the chromosome, a knockout non-human animal can be produced. For example, the activity of the product can be modified by introducing mutations by substituting, deleting, or inserting bases into the translation region of the gene encoding the protein of the present invention on the chromosome. Further, by introducing a similar mutation into the expression control region, it is possible to modify the degree, timing, tissue specificity, etc. of the expression. In addition, it is possible to more positively control the expression timing, expression site, expression amount, and the like by combining with the Cre-ΙοΙ system.
このような例として、 脳のある特定の領域で発現されるプロモーターを利用し て、 その領域でのみ目的遺伝子を欠失させた例 〔Cell, 87, 7, 1317 ( 1996 )〕 や Creを発現するアデノウイルスを用いて、 目的の時期に、 臓器特異的に目的遺伝子 を欠失させた例 〔Science, 278, 5335 ( 1997)〕 が知られている。  Examples of such cases include the use of a promoter that is expressed in a specific region of the brain and deletion of the target gene only in that region (Cell, 87, 7, 1317 (1996)) or expression of Cre. There is known an example in which a target gene is specifically deleted at an intended time using an adenovirus [Science, 278, 5335 (1997)].
従って、 染色体上の本発明の蛋白質をコードする遺伝子についても、 このよう に任意の時期や組織で発現を制御できる、 または任意の挿入、 欠失、 置換をその 翻訳領域や発現制御領域に有する、 ノックァゥト非ヒト動物を作製することがで きる。 ノックアウト非ヒト動物は、 任意の時期、 任意の程度または任意の部位で、 本 発明の蛋白質に起因する種々の疾患の症状を誘導することができる。 Therefore, the expression of the gene encoding the protein of the present invention on the chromosome can be controlled at any time or in any tissue, or the gene has any insertion, deletion, or substitution in its translation region or expression control region. Knockout non-human animals can be produced. The knockout non-human animal can induce the symptoms of various diseases caused by the protein of the present invention at any time, any degree, or any site.
このように、 本発明のノックアウト非ヒト動物は、 本発明の蛋白質に起因する 種々の疾患の治療や予防において極めて有用な動物モデルとなる。 特にその治療 薬、 予防薬、 また機能性食品、 健康食品等の評価用モデルとして非常に有用であ 。 図面の簡単な説明  Thus, the knockout non-human animal of the present invention becomes an extremely useful animal model in the treatment and prevention of various diseases caused by the protein of the present invention. In particular, it is very useful as a model for evaluating therapeutic drugs, preventive drugs, functional foods, and health foods. BRIEF DESCRIPTION OF THE FIGURES
第 1図 は、 ヒト組織の 1本鎖 cDNAに対する RT— PCR産物のァガロース 電気泳動の結果を示す。 レーン Mは、 人/ E c oT 14 Iのマーカ一であり、 レ —ン 1から 10までは順に、 それぞれ心臓、 脳、 胎盤、 肺、 肝臓、 骨格筋、 腎臓、 臈臓、 脾臓、 胸腺の cDNAを鍊型にした RT— P CR産物である。 また、 レ一 ン Cは、 pERR4プラスミ ド DNAを銪型にした RT— P CRの結果である。 発明を実施するための最良の形態  FIG. 1 shows the results of agarose electrophoresis of RT-PCR products for single-stranded cDNA of human tissue. Lane M is a marker of human / EcoT14I, and lanes 1 to 10 are for heart, brain, placenta, lung, liver, skeletal muscle, kidney, renal, spleen, and thymus, respectively. This is an RT-PCR product obtained by converting cDNA into type II. Lane C is the result of RT-PCR using pERR4 plasmid DNA as type III. BEST MODE FOR CARRYING OUT THE INVENTION
以下、 本発明の実施例を示す。  Hereinafter, examples of the present invention will be described.
実施例 1 エストロゲン関連レセプ夕一 (Estrogen Related Recepter) 遺伝子産 物の探索 Example 1 Search for Estrogen Related Recepter Gene Product
( 1 ) GenBankデータベースによる ESTクローンの探索  (1) Search for EST clones using GenBank database
ヒト ERR2 (ァクセッション番号: X51417) の塩基配列をもとに、 塩基配列 デ—夕ベース G e n B a n k中の塩基配列との相同性を解析プログラム B L A S Tを用いて解析した。 その結果、 ヒト ERR 2と高い相同性のある E S T  Based on the nucleotide sequence of human ERR2 (accession number: X51417), the homology with the nucleotide sequence in the nucleotide sequence GenBank was analyzed using an analysis program BLAST. As a result, E ST with high homology to human ERR 2
(Expressed Sequence Tag) が 4つ見出された。 そのクロ一ン名とァクセッショ ン番号は、 それぞれ、 ys69hll.rl(H82542), ys69hll.sl(H82543)N ys81d07.sl (H91842) 、 ys81d07.rl(H91890)であった。 また、 これらのクローンの mRN Aソ —スはすべて、 ヒト網膜であった。 (Expressed Sequence Tag) was found 4 times. Its black Ichin name and Akusessho version number, respectively, ys69hll.rl (H82542), ys69hll.sl ( H82543) N ys81d07.sl (H91842), was ys81d07.rl (H91890). Also, the mRNA sources of these clones were all human retina.
H82543と H91842とは 94. 1 %、 H91890と H82542とは 96. 3%と、 それぞれ 高い相同性を示し、 ほとんど一致していると考えられる。 一方、 ヒト ERR2と H82543, ヒト 1^1 2と1191842とを比較すると、 ヒト E R R 2の 5,端で高い相同 性を示し、 その値はそれぞれ 84. 1%、 86. 0%であった。 しかも、 ヒト E RR 2塩基配列の 82 bpから 85 bpにかけて、 H82543、 H91842では欠失して いることから、 この E S T群は ERR 2とは別の遺伝子の一部であると考えられ た。 H82543 and H91842 show a high homology of 94.1%, and H91890 and H82542 show a high homology of 96.3%, which are considered to be almost identical. On the other hand, comparing human ERR2 with H82543 and human 1 ^ 12 with 1191842, it showed high homology at the 5 and 5 ends of human ERR2, with values of 84.1% and 86.0%, respectively. Moreover, human E The deletion of H82543 and H91842 from 82 bp to 85 bp of the RR 2 nucleotide sequence suggests that this EST group is part of a different gene from ERR2.
また、 ヒト £1^2と1191890の相補鎖、 およびヒト E R R 2と H82542の相補鎖 とを比較すると、 ヒト ERR2の 350bp〜700 b p付近で高い相同性を示 し、 その値はそれぞれ 88. 6%、 87. 1%であった。  Comparing the complementary strand of human £ 1 ^ 2 and 1191890 and the complementary strand of human ERR2 and H82542 show high homology near 350 bp to 700 bp of human ERR2, and the value is 88.6 each. %, 87.1%.
このことから、 この E S T群は ERR 2とは別の遺伝子の一部をコ一ドしてい ると考えられ、 その遺伝子を ERR4と命名した。 H82543、 H91842は ERR4の 5'端付近、 H82542、 H91890はそれより 3 '側の塩基配列を表わしていると考えら れる。  From this, it is considered that this EST group encodes a part of a different gene from ERR2, and the gene was named ERR4. It is thought that H82543 and H91842 represent the nucleotide sequence near the 5 'end of ERR4, and H82542 and H91890 represent the nucleotide sequence at the 3' side.
(2) c DN Aライブラリ一からの取得  (2) Acquisition from the cDNA library
実際に E R R 4遺伝子を取得するための c D N Aライブラリ一は、 E S Tクロ —ンの由来に従い、 Clontech社の Human Retina 5' -STRETCH cDNA libraryを使用 した。  As a cDNA library for actually obtaining the ERR4 gene, a Human Retina 5'-STRETCH cDNA library from Clontech was used according to the origin of the EST clone.
対数増殖期の大腸菌 C600Hfl株 (Clontech社製)を 10腿 ol/l硫酸マグネシウム 溶液中に懸濁し、 約 3万プラークのファージを添加して、 37°Cで 15分間静置 後、 NZ YT o pAga rで 150 mmシャーレに該溶液をプレーティングした。 計 12枚のシャーレ分を作製して、 それぞれ SM緩衝液 〔50画 ol/lトリス塩酸 (pH 7. 5) 、 100醒 ol八 塩化ナトリウム、 8腿 ol八 硫酸マグネシウム、 0. 01 %ゼラチン〕 を用いてファージ粒子をシャーレごとに回収し、 cDNA ライブラリーフラクションとした。  E. coli C600Hfl (manufactured by Clontech) in the logarithmic growth phase was suspended in a 10 l / l magnesium sulfate solution, about 30,000 plaques of phage were added, and the mixture was allowed to stand at 37 ° C for 15 minutes. The solution was plated on a 150 mm Petri dish with pAgar. Prepare a total of 12 Petri dishes, each with SM buffer solution (50 fractions ol / l Tris-HCl (pH 7.5), 100 liters ol sodium octachloride, 8 thighs ol magnesium octasulfate, 0.01% gelatin) The phage particles were collected for each petri dish using, and used as a cDNA library fraction.
ERR 2と比較し、 ERR 4に特異的な塩基配列である、 配列番号 3に示す塩 基配列を有するプライマ一 ERR4-01および配列番号 4に示す塩基配列を有するプ ラィマ一 03を設計した。 プライマ一 ERR4- 01は、 E S T配列 H82543の 84b pから 107bpに、 また、 プライマ一 ERM-03は、 E S T配列 H91890の 70 b p から 93 bpに、 それぞれ対応している。  A primer ERR4-01 having a base sequence shown in SEQ ID NO: 3 and a primer 03 having a base sequence shown in SEQ ID NO: 4, which are base sequences specific to ERR 4, were designed in comparison with ERR 2. The primer ERR4-01 corresponds to 84 bp to 107 bp of the EST sequence H82543, and the primer ERM-03 corresponds to 70 bp to 93 bp of the EST sequence H91890.
このプライマ一 ERR4- 01と ERR4- 03とを用いて、 cDNAライブラリーフラクシ ヨンを錶型として P CRを行った。  Using the primers ERR4-01 and ERR4-03, PCR was performed using the cDNA library fraction as type II.
該 P CRにより増幅がみられたフラクションを、 1枚あたり約 3万プラークを 生じるように、 15 Ommシャーレにプレーティングした。 生じたプラークのファージを Hybond- N+メンブレン( Amersham社製)に卜ランスフ ァ一し、 このメンブレンに対しプラーク 'ハイブリダィゼ一シヨンを行った。 プローブは、 プライマー ERR4-01とプライマー EM4-03をもとに、 PCRDIGプロ一 ブ合成キット(Boehringer Mannheim社製)を用いてジゴキシゲニン標識したものを 用いた。 反応は、 ハイプリダイゼ一ション溶液 〔 5 X S S C (75 Ommol/1塩化 ナトリウム、 Ί 5誦 ol/l クェン酸ナトリウム (pH7. 0) )、 1. 0 %核酸ハ ィブリダイゼ一ション用ブ口ッキング試薬(Boehringer Mannheim社製)、 0. 1 % N—ラウロイルザルコシン、 0. 02%SDS〕 中、 65 °Cでー晚行った。 The fraction amplified by the PCR was plated on a 15 Omm petri dish so as to generate about 30,000 plaques per sheet. The resulting plaque phage was transferred onto a Hybond-N + membrane (manufactured by Amersham), and the plaque was hybridized to the membrane. The probe used was digoxigenin labeled using the PCRDIG probe synthesis kit (Boehringer Mannheim) based on the primers ERR4-01 and EM4-03. The reaction was performed using a hybridization solution (5 XSSC (75 Ommol / 1 sodium chloride, Ί5 ol / l sodium citrate (pH 7.0)), 1.0% nucleic acid hybridization booking reagent (Boehringer Mannheim) in 0.1% N-lauroyl sarcosine, 0.02% SDS] at 65 ° C.
ハイブリダィゼーシヨン後のフィルターを 0. 1% 803を含む0. 1倍濃度 の SSCにより、 68° ( 、 15分間の条件で 2回洗浄した後、 アルカリフォスフ ァ夕一ゼ結合抗 D I G抗体、 化学発光基質 CSPDからなる D I G発光検出キッ h (Boehringer Mannheim社製)で処理し、 化学発光フィルム(Hyperf ilm-ECL;  After hybridization, the filter was washed twice with 0.1% SSC containing 0.1% 803 at 68 ° C for 15 minutes, and then alkaline phosphatase-conjugated anti-DIG The antibody is treated with a DIG luminescence detection kit (Boehringer Mannheim) comprising CSPD and a chemiluminescent film (Hyperfilm-ECL;
Amersham社製)を感光させてシグナルの検出を行つた。 (Amersham) to detect the signal.
検出されたシグナルに相当するプラークを SM緩衝液に懸濁し、 再度、 シング ルプラークが単離できるようにシャーレにプレーティングした。  The plaque corresponding to the detected signal was suspended in SM buffer and again plated on a Petri dish so that a single plaque could be isolated.
生じたプラークに対して、 上記と同様の操作を行い、 ハイブリダィゼーシヨン によるシグナルの検出を行つた。  The same operation as described above was performed on the resulting plaque, and the signal was detected by hybridization.
シグナルのあったシングルプラークを S M緩衝液に懸濁し、 そこから Lambda Midi Kit(QIAGEN社製)を用いてファージ DNAを単離した。  A single plaque with a signal was suspended in an SM buffer, and phage DNA was isolated therefrom using a Lambda Midi Kit (QIAGEN).
ここで用いたライブラリーは入 gtlOベクターを使用しており、 クローンの cD NAは E c o R Iサイ トに揷入されているはずなので、 ファージ DNAを E c o R Iで切断しィンサート DNAを、 同じく pBluescript II SK( + )(STRATAGENE社製) を E c 0 RIで切断したベクターとライゲーシヨンしてサブクローニングを行つ た。 実施例 2 塩基配列の決定  The library used here uses the input gtlO vector, and the cDNA of the clone should have been inserted into the EcoRI site.Therefore, the phage DNA was cut with EcoRI, and the insert DNA was also converted into pBluescript. II SK (+) (manufactured by STRATAGENE) was ligated with a vector cut with Ec0RI and subcloned. Example 2 Determination of base sequence
実施例 1で得たクローンの cDN Aの 5, 末端からの塩基配列をパーキン ·ェ ルマ一社の DNAシークェンサ一377を用いて決定した。  The nucleotide sequence from the 5 'end of cDNA of the clone obtained in Example 1 was determined using DNA Sequencer-377 of Perkin Pharma Co., Ltd.
塩基配列の決定は、 パーキンエルマ一社のキットを使用し、 該キッ卜の指示に つて行った o 決定された塩基配列について、 塩基配列データベース G e nB an k中の既知 遺伝子の塩基配列との相同性を解析プログラム B L A S Tを用いて解析したとこ ろ、 ESTと一致する以外、 他の既知遺伝子とは一致しなかった。 The nucleotide sequence was determined using a kit from PerkinElmer Inc. and following the instructions for the kit. The homology of the determined nucleotide sequence to the nucleotide sequence of a known gene in the nucleotide sequence database GenBank was analyzed using the analysis program BLAST. Did not match.
このクローンは、 核内レセプ夕一、 特にヒト ERR 1、 ERR 2, ERR3と 相同性をもつので、 該クローンにコードされる蛋白質を ERR 4とした。 配列番 号 1に ERR4のァミノ酸配列を配列番号 2にその塩基配列を示す。  Since this clone has homology to nuclear receptors, especially human ERR1, ERR2 and ERR3, the protein encoded by this clone was designated as ERR4. SEQ ID NO: 1 shows the amino acid sequence of ERR4 and SEQ ID NO: 2 shows its nucleotide sequence.
ERR 4をコードする cDNAを含有するプラスミ ド pERR4を保有する大 腸菌 Escherichia coli DH5ひ/ pE皿 4は、 FERM BP— 6613として、 平成 11年 1月 7日付けで工業技術院生命工学工業技術研究所、 日本国茨城県つくば 巿東 1丁目 1番 3号 (郵便番号 305-8566) に寄託されている。 実施例 3 ERR4遺伝子の各種臓器での発現  Plasmid containing cDNA encoding ERR4 Escherichia coli DH5 / pE4, which has pERR4, was designated as FERM BP-6613 on January 7, 1999 The research laboratory has been deposited at Tsukuba Tsukuba, Ibaraki, Japan, 1-3-1, Higashi (postal code 305-8566). Example 3 Expression of ERR4 gene in various organs
ヒト各種組織 (心臓、 脳、 胎盤、 肺、 肝臓、 骨格筋、 腎臓、 滕臓、 脾臓、 胸腺) の po ly (A) +RNA(Clontech社製) 1〃gから、 Superscript  Superscript from 1〃g of poly (A) + RNA (Clontech) of various human tissues (heart, brain, placenta, lung, liver, skeletal muscle, kidney, ligens, spleen, thymus)
Preamplification System(GIBC0 BRL社製)を用いて、 それぞれの cDN Aを合成 した。 具体的な手法は、 このキットの指示に従い、 最終量 lmlの水に溶解させ た。 Each cDNA was synthesized using a Preamplification System (GIBC BRL). The specific procedure was as described in this kit, and dissolved in a final volume of 1 ml of water.
該溶液を 5倍に希釈し、 該希釈液 5. 0〃1を用い、 RT— PCRを行った。 プライマ一は、 ERR4のオープン ' リーディング ' フレームを含む領域が増 幅でき、 ERR4特異的な配列である、 配列番号 5に示す塩基配列を有するプラ ィマ一 ERR4- 07、 配列番号 6に示す塩基配列を有するブラィマー ERM- 06の 2種類 を用いた。  The solution was diluted 5-fold, and RT-PCR was performed using 5.0: 1 of the diluted solution. The primer can be amplified in the region containing the open 'reading' frame of ERR4, and is a primer having the nucleotide sequence shown in SEQ ID NO: 5, which is an ERR4-specific sequence. Two types of sequencer, Bramer ERM-06, were used.
ERR4-07は、 配列番号 2の塩基配列の 807 b pから 829 b pに対応しており、 ERR4-06は、 配列番号 2の塩基配列の 1470bpから 1493 b pの相補鎖に対 応している。  ERR4-07 corresponds to 807 bp to 829 bp of the nucleotide sequence of SEQ ID NO: 2, and ERR4-06 corresponds to the complementary strand of 1470 bp to 1493 bp of the nucleotide sequence of SEQ ID NO: 2.
94 °Cの変性を 1分間、 62 °Cのァニーリングを 1分間、 72 °Cの伸長を 2分 間の反応を 1サイクルとし、 該サイクルを 35回繰り返して P CRを行い、 その 増幅産物に対し、 ァガロースゲル電気泳動を行った。  One cycle of denaturation at 94 ° C for 1 minute, annealing at 62 ° C for 1 minute, and extension at 72 ° C for 2 minutes was defined as one cycle, and the cycle was repeated 35 times to perform PCR. On the other hand, agarose gel electrophoresis was performed.
結果を図 1に示す。  The results are shown in Figure 1.
肝臓と腎臓由来の c DNAから、 約 0. 7 kbの特異的なバンドが検出された。 従って、 ERR 4遺伝子は、 主に肝臓と腎臓で特異的に発現していることが判明 した。 産業上の利用可能性 A specific band of about 0.7 kb was detected in cDNA from liver and kidney. Therefore, it was found that the ERR4 gene was specifically expressed mainly in the liver and kidney. Industrial applicability
本発明によれば、 炎症、 癌、 骨粗鬆症、 糖尿病、 腎疾患等の治療薬の探索、 開 発に有用なリガンド結合能および DN A結合能を有する蛋白質、 該蛋白質をコー ドする DNA、 該蛋白質を認識する抗体、 およびこれらの利用方法を提供するこ とができる。  According to the present invention, a protein having ligand-binding ability and DNA-binding ability useful for search and development of therapeutic agents for inflammation, cancer, osteoporosis, diabetes, kidney disease, etc., DNA encoding the protein, and the protein An antibody recognizing, and a method of using these can be provided.

Claims

請求の範囲 The scope of the claims
1. 配列番号 1で示されるアミノ酸配列からなる蛋白質。  1. A protein consisting of the amino acid sequence represented by SEQ ID NO: 1.
2. 配列番号 1で示されるァミノ酸配列において 1以上のアミノ酸が欠失、 置換または付加されたアミノ酸配列からなり、 かつ配列番号 1で示されるァミノ 酸配列からなる蛋白質の有するリガンド結合能および DNA結合能を有する蛋白  2. Ligand-binding ability and DNA of a protein comprising an amino acid sequence represented by SEQ ID NO: 1 in which one or more amino acids are deleted, substituted or added, and comprising the amino acid sequence represented by SEQ ID NO: 1 Protein with binding ability
3. 配列番号 1で示されるアミノ酸配列と 96%以上の相同性を有するアミ ノ酸配列を含み、 かつ配列番号 1で示されるアミノ酸配列からなる蛋白質の有す るリガンド結合能および DN A結合能を有する蛋白質。 3. Ligand-binding ability and DNA-binding ability of a protein comprising an amino acid sequence having 96% or more homology with the amino acid sequence represented by SEQ ID NO: 1 and having the amino acid sequence represented by SEQ ID NO: 1 A protein having
4. 請求項 1〜3のいずれかに記載の蛋白質をコードする DNA。  4. A DNA encoding the protein according to any one of claims 1 to 3.
5. 配列番号 2の 140〜 1438番目の塩基配列を有する DNA。  5. DNA having a nucleotide sequence at positions 140 to 1438 in SEQ ID NO: 2.
6. 請求項 4または 5記載の DNAとストリンジェン卜な条件下でハイプリ ダイズし、 配列番号 2の 140〜 1438番目の塩基配列と 90 %以上の相同性 を有し、 かつ配列番号 1で示されるアミノ酸配列からなる蛋白質の有するリガン ド結合能および DN A結合能を有する蛋白質をコードする DNA。  6. It hybridizes with the DNA according to claim 4 or 5 under stringent conditions, has 90% or more homology with the nucleotide sequence at positions 140 to 1438 of SEQ ID NO: 2, and is represented by SEQ ID NO: 1. DNA encoding a protein having a ligand-binding ability and a DNA-binding ability of a protein comprising an amino acid sequence represented by
7. Escherichia coli DH5ひ/ pERR4 (FERM BP— 6613) の保有す るプラスミ ドに含まれ、 かつリガンド結合能および DNA結合能を有する蛋白質 をコードする DNA。  7. DNA that is contained in the plasmid of Escherichia coli DH5 / pERR4 (FERM BP-6613) and encodes a protein having ligand binding ability and DNA binding ability.
8. 請求項 4〜 7のいずれかに記載の DNAを含有してなる組換えベクター c 8. A recombinant vector c containing the DNA according to any one of claims 4 to 7.
9. 請求項 8記載の組換えべクタ一を宿主細胞に導入して得られる形質転換 体。 9. A transformant obtained by introducing the recombinant vector according to claim 8 into a host cell.
10. 請求項 9記載の形質転換体を培地に培養し、 培養物中に請求項 1〜 3 のいずれかに記載の蛋白質を生成蓄積させ、 該培養物から該蛋白質を採取するこ とを特徴とする蛋白質の製造方法。  10. The transformant according to claim 9 is cultured in a medium, the protein according to any one of claims 1 to 3 is produced and accumulated in the culture, and the protein is collected from the culture. A method for producing a protein.
11. 請求項 1〜3のいずれかに記載の蛋白質を認識する抗体。  11. An antibody that recognizes the protein according to any one of claims 1 to 3.
12. 請求項 4 ~ 7のいずれかに記載の DN Aの有する塩基配列中の連続し た 5〜 60塩基からなる配列に相当するオリゴヌクレオチドまたは該ォリゴヌク レオチドと相補的な配列に相当するォリゴヌクレオチド。  12. An oligonucleotide corresponding to a sequence consisting of 5 to 60 consecutive nucleotides in the nucleotide sequence of the DNA according to any one of claims 4 to 7, or an oligonucleotide corresponding to a sequence complementary to the oligonucleotide. nucleotide.
13. 請求項 4 ~ 7のいずれかに記載の DN Aを用い、 ハイブリダィゼ一シ ョン法により、 請求項 1〜 3のいずれかに記載の蛋白質をコ一ドする遺伝子の発 現を検出する方法。 13. Use of the DNA of any one of claims 4 to 7 to generate a gene encoding the protein of any one of claims 1 to 3 by a hybridization method. How to detect the reality.
14. 請求項 1 2記載のオリゴヌクレオチドを用い、 ポリメラーゼ 'チエイ ン ' リアクション法により、 請求項 1〜3のいずれかに記載の蛋白質をコードす る遺伝子の発現を検出する方法。  14. A method for detecting the expression of a gene encoding the protein according to any one of claims 1 to 3, using the oligonucleotide according to claim 12 by a polymerase 'chain' reaction method.
15. 請求項 4 ~ 7のいずれかに記載の D N Aを用い、 ハイブリダィゼーシ ヨン法により、 請求項 1〜3のいずれかに記載の蛋白質をコ一ドする遺伝子の変 異を検出する方法。  15. Using the DNA according to any one of claims 4 to 7 to detect a mutation in a gene encoding the protein according to any one of claims 1 to 3 by a hybridization method. Method.
16. 請求項 1 2記載のオリゴヌクレオチドを用い、 ポリメラ一ゼ *チエイ ン ' リアクション法により、 請求項 1〜3のいずれかに記載の蛋白質をコードす る遺伝子の変異を検出する方法。  16. A method for detecting a mutation in a gene encoding the protein according to any one of claims 1 to 3, using the oligonucleotide according to claim 12 by a polymerase * chain 'reaction method.
17. 請求項 1 3〜 1 6のいずれかに記載の方法を用いて炎症、 癌、 骨粗鬆 症、 糖尿病または腎疾患を検出する方法。  17. A method for detecting inflammation, cancer, osteoporosis, diabetes or kidney disease using the method according to any one of claims 13 to 16.
18. 請求項 4〜 7のいずれかに記載の D N Aを用いることを特徴とする、 請求項 1 ~ 3のいずれかに記載の蛋白質をコードする遺伝子の転写を抑制する方 法。  18. A method for suppressing transcription of a gene encoding the protein according to any one of claims 1 to 3, wherein the DNA according to any one of claims 4 to 7 is used.
19. 請求項 1 2記載のオリゴヌクレオチドを用いることを特徴とする、 請 求項 1〜 3のいずれかに記載の蛋白質をコ一ドする遺伝子の転写を抑制する方法。  19. A method for suppressing the transcription of a gene encoding a protein according to any one of claims 1 to 3, wherein the oligonucleotide according to claim 12 is used.
20. 請求項 4〜 7のいずれかに記載の D N Aを用いることを特徴とする、 請求項 1〜 3のいずれかに記載の蛋白質をコードする mR N Aの翻訳を抑制する 方法。  20. A method for suppressing translation of mRNA encoding the protein according to any one of claims 1 to 3, which comprises using the DNA according to any one of claims 4 to 7.
21. 請求項 1 2記載のオリゴヌクレオチドを用いることを特徴とする、 請 求項 1〜3のいずれかに記載の蛋白質をコードする mR N Aの翻訳を抑制する方 法。  21. A method for suppressing translation of mRNA encoding the protein according to any one of claims 1 to 3, wherein the oligonucleotide according to claim 12 is used.
22. 請求項 4〜 7のいずれかに記載の D N Aを用いることを特徴とする、 請求項 1〜 3のいずれかに記載の蛋白質をコ一ドする遺伝子のプロモーター領域 を取得する方法。  22. A method for obtaining a promoter region of a gene encoding the protein according to any one of claims 1 to 3, wherein the DNA according to any one of claims 4 to 7 is used.
23. 請求項 1 2記載のオリゴヌクレオチドを用いることを特徴とする、 請 求項 1〜 3のいずれかに記載の蛋白質をコードする遺伝子のプロモーター領域を 取得する方法。  23. A method for obtaining a promoter region of a gene encoding a protein according to any one of claims 1 to 3, wherein the oligonucleotide according to claim 12 is used.
24. 請求項 4〜 7のいずれかに記載の D N Aを含有する、 炎症、 癌、 骨粗 鬆症、 糖尿病または腎疾患の診断薬。 24. Inflammation, cancer, osteoporosis containing the DNA according to any one of claims 4 to 7 Diagnostics for porosis, diabetes or kidney disease.
25. 請求項 1 2記載のオリゴヌクレオチドを含有する、 炎症、 癌、 骨粗鬆 症、 糖尿病または腎疾患の診断薬。  25. A diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the oligonucleotide according to claim 12.
26. 請求項 4〜 7のいずれかに記載の D N Aを含有する医薬。  26. A medicament containing the DNA according to any one of claims 4 to 7.
27. 請求項 1 2記載のオリゴヌクレオチドを含有する医薬。  27. A medicament comprising the oligonucleotide according to claim 12.
28. 請求項 4〜 7のいずれかに記載の D N Aを含有する炎症、 癌、 骨粗鬆 症、 糖尿病または腎疾患の治療薬。  28. A therapeutic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the DNA according to any one of claims 4 to 7.
29. 請求項 1 2記載のオリゴヌクレオチドを含有する炎症、 癌、 骨粗鬆症、 糖尿病または腎疾患の治療薬。  29. A therapeutic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the oligonucleotide according to claim 12.
30. 請求項 1〜3のいずれかに記載の蛋白質を用いることを特徴とする、 該蛋白質と特異的に結合するリガンドのスクリーニング方法。  30. A method for screening for a ligand that specifically binds to the protein, comprising using the protein according to any one of claims 1 to 3.
31. 請求項 3 0の方法により取得されるリガンド。  31. A ligand obtained by the method of claim 30.
32. 請求項 1 1記載の抗体を用いることを特徴とする、 請求項 1〜3のい ずれかに記載の蛋白質の免疫学的検出法。  32. The method for immunologically detecting a protein according to any one of claims 1 to 3, wherein the antibody according to claim 11 is used.
33. 請求項 1 1記載の抗体を用い、 請求項 1〜 3のいずれかに記載の蛋白 質を検出することを特徴とする免疫組織染色法。  33. An immunohistochemical staining method using the antibody according to claim 11 to detect the protein according to any one of claims 1 to 3.
34. 請求項 1 1記載の抗体を含有する炎症、 癌、 骨粗鬆症、 糖尿病または 腎疾患の診断薬。  34. A diagnostic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the antibody according to claim 11.
35. 請求項 1 1記載の抗体を含有する医薬。  35. A drug containing the antibody according to claim 11.
36. 請求項 1 1記載の抗体を含有する炎症、 癌、 骨粗鬆症、 糖尿病または 腎疾患の治療薬。  36. A therapeutic agent for inflammation, cancer, osteoporosis, diabetes or kidney disease, comprising the antibody according to claim 11.
37. 請求項 9記載の形質転換体を用いることを特徴とする、 請求項 1〜3 のいずれかに記載の蛋白質をコードする遺伝子の転写を制御する物質のスクリー 二ング方法。  37. A method for screening a substance that controls transcription of a gene encoding a protein according to any one of claims 1 to 3, wherein the transformant according to claim 9 is used.
38. 請求項 9記載の形質転換体を用いることを特徴とする、 請求項 1〜3 のいずれかに記載の蛋白質による転写制御機能に関与する物質のスクリーニング 方法。  38. A method for screening a substance involved in a transcriptional control function of a protein according to any one of claims 1 to 3, wherein the transformant according to claim 9 is used.
39. 請求項 9記載の形質転換体を用いることを特徴とする、 請求項 1〜3 のいずれかに記載の蛋白質により転写制御を受ける遺伝子のスクリーニング方法。  39. A method for screening a gene that is transcriptionally regulated by the protein according to any one of claims 1 to 3, wherein the transformant according to claim 9 is used.
40. 請求項 1〜 3のいずれかに記載の蛋白質をコードする遺伝子の発現が 一部または完全に抑制されたノックアウト非ヒト動物。 40. Expression of the gene encoding the protein according to any one of claims 1 to 3 is Knockout non-human animals that have been partially or completely suppressed.
41. 請求項 1〜3のいずれかに記載の蛋白質の有する活性が一部または完 全に抑制されたノックァゥト非ヒト動物。  41. A knockout non-human animal in which the activity of the protein according to any one of claims 1 to 3 is partially or completely suppressed.
PCT/JP2000/000106 1999-01-14 2000-01-12 Novel protein WO2000042180A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU20025/00A AU2002500A (en) 1999-01-14 2000-01-12 Novel protein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP11/7226 1999-01-14
JP722699 1999-01-14

Publications (1)

Publication Number Publication Date
WO2000042180A1 true WO2000042180A1 (en) 2000-07-20

Family

ID=11660092

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2000/000106 WO2000042180A1 (en) 1999-01-14 2000-01-12 Novel protein

Country Status (2)

Country Link
AU (1) AU2002500A (en)
WO (1) WO2000042180A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003002741A2 (en) * 2001-06-29 2003-01-09 Decode Genetics Ehf. Nucleic acids encoding nuclear receptors
EP1572866A2 (en) * 2001-03-23 2005-09-14 Syngenta Participations AG Insect nuclear receptor genes and uses thereof
US7358350B2 (en) 2003-01-20 2008-04-15 Nec Corporation Oncogene, recombinant protein derived therefrom, and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999010367A1 (en) * 1997-08-27 1999-03-04 Merck & Co., Inc. Dna molecules encoding human nuclear receptor proteins
EP0939123A2 (en) * 1998-01-15 1999-09-01 Smithkline Beecham Plc Estrogen receptor-related polypetide SBPERR4

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999010367A1 (en) * 1997-08-27 1999-03-04 Merck & Co., Inc. Dna molecules encoding human nuclear receptor proteins
EP0939123A2 (en) * 1998-01-15 1999-09-01 Smithkline Beecham Plc Estrogen receptor-related polypetide SBPERR4

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KATARINA PETTERSSON ET AL.: "Expression of a novel member of estrogen response element-binding nuclear receptor is restricted to the early stages of chorion formation during mouse embryogenesis", MECHANISMS OF DEVELOPMENT, vol. 54, no. 2, 1996, pages 211 - 223, XP002926034 *
VINCENT GIGUERE ET AL.: "Identification of a new class of steroid hormone receptors", NATURE, vol. 331, no. 6151, 1998, pages 91 - 94, XP002926033 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1572866A2 (en) * 2001-03-23 2005-09-14 Syngenta Participations AG Insect nuclear receptor genes and uses thereof
EP1572866A4 (en) * 2001-03-23 2008-01-16 Syngenta Participations Ag Insect nuclear receptor genes and uses thereof
WO2003002741A2 (en) * 2001-06-29 2003-01-09 Decode Genetics Ehf. Nucleic acids encoding nuclear receptors
WO2003002741A3 (en) * 2001-06-29 2003-03-13 Decode Genetics Ehf Nucleic acids encoding nuclear receptors
US7358350B2 (en) 2003-01-20 2008-04-15 Nec Corporation Oncogene, recombinant protein derived therefrom, and uses thereof
US7432358B2 (en) 2003-01-20 2008-10-07 Nec Corporation Oncogene, recombinant protein derived therefrom, and uses thereof
US7893246B2 (en) 2003-01-20 2011-02-22 Nec Corporation Sirna capable of inhibiting the expression of an oncogene involved in cervical cancer

Also Published As

Publication number Publication date
AU2002500A (en) 2000-08-01

Similar Documents

Publication Publication Date Title
JP4112627B2 (en) Novel polypeptide, novel DNA and novel antibody
JPWO2005035577A1 (en) Antibody composition that specifically binds to ganglioside GD3
JPH1175863A (en) 25-hydroxyvitamin d3-1-alpha-hydroxylase and dna encoding the same
WO2000027885A1 (en) Novel chimeric polypeptide
EP1225224A1 (en) Shear stress-response dna
JP4112374B2 (en) Polypeptides and DNAs for angiogenesis markers
WO2001098493A1 (en) Insulin-like growth factor-binding protein
JP2002171978A (en) Gene of fulminant hepatitis c virus strain
JP2001231578A (en) Protein belonging to il-1 family
WO2000042180A1 (en) Novel protein
JPWO2005035578A1 (en) Antibody composition that specifically binds to ganglioside GM2
JPWO2005035583A1 (en) Antibody composition that specifically binds to IL-5 receptor
WO2007037245A1 (en) Polypeptide having anti-angiogenic activity
JP3910536B2 (en) Novel polypeptide and its DNA
JP4651893B2 (en) Novel polypeptide, novel DNA, novel antibody, and novel genetically modified animal
JP2000041681A (en) New protein
EP1180525B1 (en) Transcriptional activation inhibitory protein
JP3929744B2 (en) Novel polypeptide, novel DNA, novel antibody and novel genetically modified animal
WO2001066716A1 (en) Polypeptide having phosphodiesterase activity
JP4220033B2 (en) Novel polypeptide
JPWO2002024923A1 (en) Polypeptide having phospholipase A2 activity
WO2001083705A1 (en) Myocardial cell proliferation-associated genes
JPWO2002081515A1 (en) Insulin-like growth factor binding protein
JPWO2004005510A1 (en) Novel Nogo receptor-like polypeptide and its DNA
WO2000063392A1 (en) Novel tyrosine phosphatase

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 593737

Kind code of ref document: A

Format of ref document f/p: F

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase