WO2004100976A1 - Klotho protein, anti-klotho protein antibody and use thereof - Google Patents

Klotho protein, anti-klotho protein antibody and use thereof Download PDF

Info

Publication number
WO2004100976A1
WO2004100976A1 PCT/JP2004/006899 JP2004006899W WO2004100976A1 WO 2004100976 A1 WO2004100976 A1 WO 2004100976A1 JP 2004006899 W JP2004006899 W JP 2004006899W WO 2004100976 A1 WO2004100976 A1 WO 2004100976A1
Authority
WO
WIPO (PCT)
Prior art keywords
klotho
protein
antibody
fgf23
klotho protein
Prior art date
Application number
PCT/JP2004/006899
Other languages
French (fr)
Japanese (ja)
Inventor
Itaru Urakawa
Takashi Shimada
Yuji Yamazaki
Original Assignee
Kirin Beer Kabushiki Kaisha
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 Kirin Beer Kabushiki Kaisha filed Critical Kirin Beer Kabushiki Kaisha
Publication of WO2004100976A1 publication Critical patent/WO2004100976A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/02Nutrients, e.g. vitamins, minerals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones

Definitions

  • the present invention relates to a Klotho protein that alters phosphate metabolism, vitamin D metabolism, and FGF23 action, an antibody that recognizes the Klotho protein, a complex of the Klotho protein and an FGF23 polypeptide, and uses of these substances. About.
  • Phosphorus in living organisms is present in bone, intracellular and extracellular fluids, and plays an essential role in the source of mechanical strength of bone, energy metabolism in living organisms, and maintenance of cell functions. Because of this importance, the serum phosphorus concentration is maintained at a nearly constant level, suggesting the existence of a control mechanism for maintaining the blood phosphate homeostasis.
  • the balance of phosphorus in the body is regulated by excretion in the kidney and absorption in the intestinal tract, but the kidney is considered to be important as a phosphorus balance organ. Specifically, the regulation of phosphate transporter proteins in the proximal tubule of the kidney affects the regulation of blood phosphorus levels.
  • 1,25-dihydroxyvitamin D3 plays an important role in regulating calcium metabolism rather than phosphorus metabolism.
  • FGF23 is a substance found to be a potent factor in lowering serum phosphorus and serum 1 ⁇ , 25-dihydroxyvitamin D (W02 / 14504, and Shimada T, etal: Proc Natl
  • FGF23 is serum phosphorous
  • serum It has been shown to have a role in lowering droxyvitamin D, but the substances and pathways that transmit that signal have not been clarified.
  • klotho mice with a mutant phenotype similar to that of human aging have been shown to exhibit various phenotypes due to insertion mutations of other genes at the klotho locus.
  • the extracellular region of the klotho protein is composed of two regions, KL1 and KL2.By administering a protein in which the KL1 region and immunoglobulin are fused to a klotho mouse, the thymus, spleen, It has been shown to suppress testicular atrophy and ectopic calcification (WO00 / 27885).
  • klotho mutant mice have increased serum phosphate levels and increased active vitamin D (YoshidaT, et al- .Endocrinology. 2002 Feb; 143 (2): 683-9.), Phosphorus, vitamins))
  • the expression of klotho protein during renal insufficiency which is a condition that causes abnormal homeosis, is normal. It has also been reported that it is reduced as compared to the kidney (Koh H, et al: Biochem Biophys Res Comb.
  • Patent Document 1
  • Patent Document 2
  • the present invention relates to a klotho protein that decreases or increases blood phosphorus and Z or blood active vitamin D, an anti-klotho protein antibody that increases blood phosphorus and Z or blood active vitamin D, and the action of FGF23.
  • the present invention has been made in view of such circumstances, and an object thereof is to provide a substance capable of controlling blood phosphorus concentration, blood active vitamin D concentration, and FGF23 action, and a use thereof. .
  • the present inventors found that FGF23 transduced signals into cells (enhanced ERK phosphorylation and increased egrl expression) in the presence of klotho protein, regardless of the full-length klotho protein or solubilized klotho protein, in the kidney of animals. was found to be caused as well. In addition, it was revealed that FGF23 binds to klotho protein. In addition, the interaction between FGF23 and klotho protein regulates the metabolism of phosphorus and vitamin D. I found it important for the verse.
  • a pharmaceutical composition for lowering blood phosphorus concentration and blood or active vitamin D concentration comprising a klotho protein as an active ingredient
  • a pharmaceutical composition for increasing blood phosphorus concentration and blood or active vitamin D concentration comprising a klotho protein as an active ingredient
  • a pharmaceutical composition for enhancing the action of FGF23 comprising a klotho protein as an active ingredient
  • a pharmaceutical composition for inhibiting the action of FGF23 comprising a klotho protein as an active ingredient
  • a pharmaceutical composition for increasing blood phosphorus concentration and blood or active piminin D concentration which comprises an anti-klotho protein antibody as an active ingredient,
  • a pharmaceutical composition for inhibiting the action of FGF23 which comprises an anti-klotho protein antibody as an active ingredient;
  • Neoplastic osteomalacia ADHR, XLH, renal osteodystrophy, dialysis osteopathy, osteoporosis, hypophosphatemia, rickets, osteomalacia, tubular dysfunction, osteopenia, low calcium , Hyperphosphatemia, hyper1,25D, hyperparathyroidism, ectopic calcification, pruritus, bone sclerosis, Paget's disease, hypercalcemia, hypoparathyroidism, bone pain, Phosphorus and Z selected from the group consisting of muscle weakness, skeletal deformity, growth disorders and hypo1,25D
  • the present invention relates to klotho protein, an antibody that recognizes klotho protein, an antibody that recognizes klotho protein, a complex of FGF23 and klotho protein, and a use of those substances that alters the concentration of phosphorous in blood, active vitamin]) concentration, FGF23 action It is.
  • a pharmaceutical composition comprising at least one of a klotho protein, an antibody recognizing the klotho protein, and a complex of FGF23 and the klotho protein can be used to convert a pharmaceutical composition containing phosphorus and vitamin D into, for example, a tumorous condition.
  • Osteomalacia ADH, XLH, renal osteodystrophy, dialysis osteopathy, osteoporosis, hypophosphatemia, rickets, osteomalacia, tubular dysfunction, osteopenia, hypocalcemia, hyperphosphatemia Disease, hyper1, 25D, hyperparathyroidism, ectopic calcification, pruritus, osteosclerosis, Pajet's disease, hypercalcemia, hypoparathyroidism, bone pain, muscle weakness, skeletal deformity, It can be used to treat or prevent diseases such as growth disorders and hypo1,25Demia. Solubilized klotho
  • the solubilized klotho cDNA can be prepared by a known method based on the sequence of human-derived klotho cDNA described in SEQ ID NO: 1 (Genebank accession No. AB005142). For example, it can be prepared by preparing a cDNA library from cells or organs expressing the klotho protein and performing hybridization using a part of the klotho cDNA sequence (SEQ ID NO: 1) as a probe.
  • RNA was prepared from cells and organs expressing the klotho protein, cDNA was prepared using reverse transcriptase, and oligo DNA was synthesized based on the klotho cDNA sequence (SEQ ID NO: 1). It is also possible to amplify and prepare klotho cDNA by performing a PCR reaction using it as a primer.
  • the nucleotide sequence of the klotho cDNA obtained as described above is determined. Determination of nucleotide sequence The determination can be performed by a known method such as the Maxam-Gilbert chemical modification method or the dideoxynucleotide chain termination method using M13 phage. Usually, an automatic base sequencer (for example, 373A DNA Sequencer manufactured by PERKIN-ELMER) is used. , Etc.).
  • SEQ ID NO: 1 shows the nucleotide sequence of human kloto cDNA
  • SEQ ID NO: 2 shows the amino acid sequence of human full-length klotlio protein
  • SEQ ID NO: 3 shows the amino acid sequence of solubilized human klotho protein. Active and Z or low activity bimin
  • Mutations such as deletion, substitution, and addition may occur in one or several amino acids.
  • the homology at the amino acid sequence level between the human Klotho protein and the mouse Klotho protein is 85% (SEQ ID NO: 2 is the amino acid sequence of the human klotho protein, and SEQ ID NO: 23 is the amino acid sequence of the mouse klotho protein). It is predicted that mouse klotho protein has the same function as human klotho protein.
  • klotho protein includes not only klotho protein derived from human but also klotho protein of mouse klotho protein derived from mouse / human klotho protein having amino acid homology of 80% or more, preferably 85% or more. included.
  • a protein comprising an amino acid sequence of klotho protein derived from any of these animal species other than humans, wherein the protein has a hypophosphatemic-inducing activity and a Z- or low-activity bieminin D-inducing activity, and hyperphosphatemia. Inducing activity and
  • the protein has hypophosphatemia-inducing activity and / or low-activity biminmin D-induction activity or hyperphosphatemia-inducing activity and Z or hyperactive vitamin D-induction activity Examples described later
  • the 1,25-dihydroxyvitamin D concentration may be measured.
  • Whether or not the protein has an effect of regulating the action of FGF23 can be determined by administering a solubilized klotho protein and purified FGF23 to mammalian cells, for example, by referring to the method described in Example 28 below. What is necessary is just to determine whether the stimulating effect of FGF23 on cells is affected. Further, FGF23 is involved in the phosphorylation of ERK, and it can also be measured whether or not the effect of FGF23 is regulated by using as an index that the ERK phosphorylation of FGF23 is enhanced or suppressed.
  • one or several, preferably 1 to 10, more preferably 1 to 5 amino acids may be deleted from the amino acid sequence shown in SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 23.
  • One or several amino acids, preferably 1 to 10, more preferably 1 to 5 amino acids may be added to the amino acid sequence shown in SEQ ID NO: 2, SEQ ID NO: 3 or SEQ ID NO: 23; Even if one or several amino acids, preferably 1 to 10, and more preferably 1 to 5 amino acids of the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 3 or SEQ ID NO: 23 are substituted with other amino acids, Good.
  • Families which are generally classified based on the characteristics of the side chains of amino acid side chains, include the following.
  • Acidic amino acid family 1 aspartic acid, glutamic acid
  • Non-polar amino acid family alanine, norin, leucine, isoleucine, proline, fenylalanine, methionine, triptophan
  • Uncharged polar amino acid family Glycine, Asparagine, Glutamine, Cystine, Serine, Threonine, Tyrosine
  • Aromatic amino acid family phenylalanine, tributofan, tyrosine
  • a mutation is required in the nucleotide sequence of DNA encoding the amino acid.
  • the method of introduction is adopted.
  • Mutation can be introduced into DNA by a known method such as the Kimkel method or the Gapped duplex method, or a method analogous thereto.
  • a mutation is introduced based on a site-directed mutagenesis method using a mutant oligonucleotide as a primer.
  • Mutagenesis kits for example, Mutan-K (TAKARA), Mutan-G (TAKARA) ), TAKARA's LA PCR in vitro Mutagenesis series kit, etc.).
  • the DNA of the present invention also includes a DNA encoding a protein having a hyperphosphatemia-inducing activity and an activity of inducing Z or highly active vitamin D bloodemia, or an effect of regulating the action of FGF23.
  • a DNA sequence has a homology of 70% or more, preferably 80% or more, more preferably 90% or more, and particularly preferably 95% or more with the sequence of the DNA shown in SEQ ID NO: 1.
  • these DM sequences also include klotho proteins derived from non-human animal species such as mouse klotho protein.
  • the probe refers to a probe having a complementary sequence to the full length of the sequence shown in SEQ ID NO: 1 or a probe having a complementary sequence to a continuous sequence (partial sequence) having a length of 17 bases or more.
  • stringent conditions refer to those having a sodium concentration of 750 mM or more, preferably 900 mM or more, and a temperature of 40 ° C. or more, preferably 42. Specifically, it refers to the conditions of 6XSSC, 5XDenhardt, 0.5% SDS, 503 ⁇ 4 Formamide, 42.
  • 6XSSC means 900 mM NaCl and 90 mM sodium citrate.
  • Denhardt's solution is a solution containing BSA (polyserum albumin), polyvinylpyrrolidone and Ficoll400. 50xDenhardt is composed of 1% BSA, 1% polyvinylpyrrolidone and l% Ficoll400. Means one tenth of the concentration).
  • the protein encoded by the DNA of the present invention may have hypophosphatemia-inducing activity and Z or low-activity vitamin D-blood-inducing activity, hyperphosphatemia-inducing activity and / or a highly active vitamin]) Whether the protein has the disease-inducing activity or the effect of regulating the action of FGF23 is analyzed by referring to the methods described in Examples 13 and 29 described below.
  • the expressed cells can be transplanted into mice, or the protein can be administered to mice, etc., and the serum phosphorus concentration or 1,25-dihydroxyvitamin D concentration measured, and the protein regulates the action of FGF23. It is determined whether or not FGF23 is administered to mammalian cells by solubilizing klotho protein and purified FGF23, for example, by referring to the method described in Example 28 below. It is sufficient to determine whether the stimulus effect is affected.
  • the DNA of the present invention can be obtained by chemical synthesis or by PCR using a primer synthesized from the determined nucleotide sequence. Preparation of recombinant vector and transformant containing DNA of the present invention
  • the recombinant vector of the present invention can be obtained by ligating (inserting) the DNA of the present invention into an appropriate vector.
  • the vector into which the DNA of the present invention is inserted is not particularly limited as long as it can be replicated in a host, and examples thereof include plasmid DNA and phage DNA.
  • Plasmid DNAs include Escherichia coli-derived plasmids (eg, pBR322, pBR325, PUC118, pUC119, etc.), Bacillus subtilis-derived plasmids (eg, ⁇ , pTP5, etc.), and yeast-derived plasmids (eg, YEpl3, YEp24, YCp50) Phage DNA includes ⁇ phage. Also, animal viruses such as retrovirus, adenovirus or vaccinia virus, or insect virus vectors such as baculovirus can be used. Furthermore, a fusion plasmid in which GST, His-tag, etc. are linked can also be used.
  • purified DM is cut with an appropriate restriction enzyme, inserted into an appropriate vector DNA restriction enzyme site or a multicloning site, and ligated to the vector. Is adopted.
  • the vector of the present invention contains, in addition to the promoter and the DNA of the present invention, cis elements such as enhancers, splicing signals, poly-A addition signals, selection markers, and ribosome binding sequences (SD sequences), if desired.
  • cis elements such as enhancers, splicing signals, poly-A addition signals, selection markers, and ribosome binding sequences (SD sequences), if desired.
  • SD sequences ribosome binding sequences
  • the selection marker include a dihydrofolate reductase gene, an ampicillin resistance gene, a neomycin resistance gene, and the like.
  • the transformant of the present invention can be obtained by introducing the recombinant vector of the present invention into a host so that the target gene can be expressed.
  • the host is not particularly limited as long as it can express the DNA of the present invention.
  • bacteria belonging to the genus Escherichia such as Escherichia coli, Bacillus such as Bacillus subtilis iBacillus subtil is)
  • bacteria belonging to the genus Pseudomonas such as Pseudomonas [, or Saccharomyces cerevisiae [ Saccharomyces cerevisiae), mother of the like is 11
  • Nosakkaromisesu-Nbe Schotanidomonas
  • Animal cells such as COS cells, CH0 cells, HEK293 cells, and insect cells such as Sf9 and Si21 can also be used.
  • a bacterium such as Escherichia coli
  • the recombinant vector of the present invention is capable of autonomous replication in the bacterium, and comprises a promoter, a ribosome binding sequence, the DNA of the present invention, and a transcription termination sequence. Is preferred.
  • a gene that controls a promoter may be included.
  • Escherichia coli include Escherichia coli JM109 and HB101
  • Bacillus subtilis include Bacillus zubuchi (TABacillus ⁇ ⁇ / ⁇ ).
  • Any promoter can be used as long as it can be expressed in a host such as E. coli.
  • a T7 promoter derived from Escherichia coli phage such as a trp promoter, a lac promoter, a PL promoter, and a PR promoter, may be used.
  • An artificially designed and modified promoter such as the tac promoter may be used.
  • the method for introducing the recombinant vector into bacteria is not particularly limited as long as it is a method for introducing DNA into bacteria. For example, a method using calcium ions, an election port poration method and the like can be mentioned.
  • yeast When yeast is used as the host, for example, Saccharomyces cerevisiae (Schizosaccharomyces lord b) Pichia pastoris (Sodium A / a /?) Is used.
  • the promoter is not particularly limited as long as it can be expressed in yeast.
  • the method for introducing the recombinant vector into yeast is not particularly limited as long as it is a method for introducing DNA into yeast, and examples thereof include an elect-portation method, a spout-type plast method, and a lithium acetate method.
  • monkey cells COS-7, Vero, Chinese ham cells—ovary cells (CH0 cells), mouse L cells, rat GH3 cells, or human FL, HEK293, HeLa, or Jurkat cells are used.
  • the promoter an SRa promoter, SV40 promoter, LTR promoter, iS-actin promoter or the like may be used, or an early gene promoter of human cytomegalovirus may be used.
  • Methods for introducing the recombinant vector into animal cells include, for example, the electrophoresis method, the calcium phosphate method, and the lipofection method.
  • the klotho protein of the present invention refers to SEQ ID NO: 2, SEQ ID NO: 3 or SEQ ID NO: 2.
  • the klotho protein in the present invention includes a hypophosphatemic-inducing activity and / or a low-activity vitmin, a D-hyperphosphate-inducing activity, a hyperphosphatemia-inducing activity, and a Z- or highly-active vymin.
  • the klotho protein having activity, hyperphosphatemia-inducing activity and no- or highly-active biminmin D-hememia-inducing activity, or an effect of regulating the action of FGF23 has the nucleotide sequence shown in SEQ ID NO: 1 or SEQ ID NO: 1 A sequence containing the nucleotide numbers 1 to 293 4 of the nucleotide sequence shown in SEQ ID NO: 1, or a nucleotide sequence encoding the klotho protein of another animal species or nucleotide numbers 1 to 293 4 of the nucleotide sequence of human klotho DNA
  • the transformant can be produced by introducing a sequence containing the sequence corresponding to No.
  • the protein thus produced may be subject to modification of the polypeptide chain such as cleavage or glycosylation by the protein modification mechanism of the host.
  • the klotho protein of the present invention can be obtained by culturing the transformant and collecting from the culture.
  • culture means not only the culture supernatant but also cultured cells or cultured cells, or cells or disrupted cells.
  • the method for culturing the transformant of the present invention is performed according to a usual method used for culturing a host.
  • the culture medium for culturing transformants obtained using microorganisms such as Escherichia coli and yeast as a host contains carbon sources, nitrogen sources, inorganic salts, etc. that can be used by the microorganisms, and efficiently cultivates the transformants.
  • the medium can be used, either a natural medium or a synthetic medium may be used.
  • the carbon source include carbohydrates such as glucose, fructose, sucrose and starch, organic acids such as acetic acid and propionic acid, and alcohols such as ethanol and propanol.
  • Nitrogen sources include ammonia, ammonium chloride, ammonium sulfate, ammonium acetate, ammonium phosphate, and other inorganic or organic acid ammonium salts or other nitrogen-containing compounds, as well as peptone, meat extract, corn steep liquor, etc. Is mentioned.
  • the inorganic substance include potassium phosphate monobasic, potassium phosphate phosphate, magnesium phosphate, magnesium sulfate, sodium chloride, ferrous sulfate, manganese sulfate, copper sulfate, calcium carbonate, and the like.
  • the cultivation is usually carried out at 37 under aerobic conditions such as shaking culture or aeration / agitation culture at 4 to 48 hours.
  • the pH is maintained at 6.0 to 8.0.
  • the pH is adjusted using an inorganic or organic acid, an alkaline solution, or the like.
  • an antibiotic such as ampicillin-tetracycline may be added to the medium as needed.
  • an inducer may be added to the medium as needed.
  • an inducer may be added to the medium as needed.
  • IPTG isopropyl- / 3-D-thiogalactopyranoside
  • IPTG or the like can be added to the medium.
  • IAA indoleacrylic acid
  • Examples of a medium for culturing a transformant obtained using animal cells as a host include commonly used RPMH640 medium, DMEM medium, and a medium obtained by adding fetal calf serum or the like to such a medium. Culturing is usually, 5% C0 2 presence is performed 1-10 days at 37 ° C. During culture, antibiotics such as kanamycin and benicillin may be added to the medium as needed.
  • the objective polyclones are disrupted by sonication, repeated freeze-thawing, homogenizer treatment and the like to disrupt the cells or cells. Collect the peptide.
  • the culture solution is used as it is, or the cells or cells are removed by centrifugation or the like. Thereafter, by using a general biochemical method used for isolation and purification of the protein, for example, ammonium sulfate precipitation, gel chromatography, ion exchange chromatography, affinity mouth chromatography, etc., alone or in an appropriate combination,
  • the polypeptide of the present invention can be isolated and purified from the culture.
  • the in vivo activity of the klotho protein of the present invention is determined by the experiments of subcutaneously transplanting the above-mentioned recombinant cells expressing the klotho protein of the present invention into nude mice and the experiment of administering solubilized klotho protein intravenously to mice. Was evaluated.
  • FIG. 8 Example 1
  • mice transplanted with cells expressing the klotho protein of the present invention were transplanted with control CH0 cells not transfected with the DNA of the present invention to form individuals or tumors. They had apparent hypophosphatemia and hypoactive vitamin Demia compared to non-treated individuals. This indicates that the klotho protein of the present invention has hypophosphatemia-inducing activity and low-active vitamin Demia-inducing activity. Became clear.
  • the above proteins can be modified.
  • modification method any known method can be adopted, and is disclosed in detail, for example, in Japanese Patent Publication No. H10-510980.
  • Anti-klotho protein antibody any known method can be adopted, and is disclosed in detail, for example, in Japanese Patent Publication No. H10-510980.
  • the anti-klotho protein antibody in the present invention is an antibody or a part of an antibody reactive with the klotho protein or a part thereof as defined above.
  • the antibody of the present invention includes a heavy chain and a Z having an amino acid sequence in which one or several amino acids are deleted, substituted or added in the amino acid sequence of each of the heavy chain and Z or the light chain constituting the antibody.
  • a monoclonal antibody which consists of a light chain and can bind to the k101ho protein is also included.
  • the partial modification of amino acids (deletion, substitution, insertion, addition) as described above partially modifies the nucleotide sequence encoding the amino acid sequence. Can be introduced. These modification techniques are well known to those skilled in the art, and a commercially available mutagenesis kit or the like can be used.
  • the antibody of the present invention can be produced, for example, by the following production method. That is, for example, the klotho protein as defined above, or a part thereof, or a conjugate with an appropriate substance (for example, bovine serum albumin or the like) for enhancing the antigenicity of the antigen is optionally immunized. Immunize non-human mammals, including human antibody-producing transgenic mice, together with activators (such as Freimd's Adjuvant). Alternatively, immunization can be performed by administering an expression vector incorporating DNA encoding the klotho protein. Polyclonal antibodies can be obtained from serum obtained from immunized animals.
  • an appropriate substance for example, bovine serum albumin or the like
  • immunization can be performed by administering an expression vector incorporating DNA encoding the klotho protein.
  • Polyclonal antibodies can be obtained from serum obtained from immunized animals.
  • Monoclonal antibodies were prepared by preparing hybridomas from antibody-producing cells obtained from immunized animals and myeloma cells (myeloma cells) that lack the ability to produce autoantibodies, cloned the hybridomas, and used them as antigens for immunization. It is produced by selecting a clone that produces a monoclonal antibody showing specific affinity for it.
  • Hybridomas secreting monoclonal antibodies were prepared according to the method of Kohler and Milstein et al.
  • it is prepared by cell fusion with a myeloma cell derived from a mammal such as a human or the like which is not capable of producing autoantibodies.
  • Screening of hybridoma clones producing monoclonal antibodies is performed by culturing hybridomas, for example, in a microplate, and determining the reactivity of the culture supernatant in the wells in which proliferation has been observed with the immunogen, for example.
  • the measurement can be performed by enzyme immunoassay such as ELISA.
  • Production of monoclonal antibodies from hybridomas It can be performed by culturing in a toro and isolating from the culture supernatant. Alternatively, it can be isolated from ascites by culturing in an ascitic fluid in ascites of mice, rats, guinea pigs, hamsters, or egrets.
  • a gene encoding a human monoclonal antibody is cloned from an antibody-producing cell such as a hybridoma and inserted into an appropriate vector, which is then used as a host (eg, a mammalian cell line, Escherichia coli, yeast cells, insect cells, plant cells, etc.). Cells, etc.) to produce recombinant antibodies produced using genetic recombination technology.
  • a host eg, a mammalian cell line, Escherichia coli, yeast cells, insect cells, plant cells, etc.
  • hybridomas When hybridomas are cultured in vitro, the hybridomas are grown, maintained, and preserved in accordance with various conditions such as the characteristics of the cell type to be cultured, the purpose of the test and research, and the culture method. It can be carried out using any known nutrient medium such as that used to produce monoclonal antibodies or any nutrient medium derived and prepared from a known basal medium.
  • a polyclonal antibody of the present invention was prepared by binding a chemically synthesized partial peptide of kloto protein to cysiglobulin, which is a carrier protein, to immunize egrets.
  • Antibodies to each of the peptides induced by immunization were purified by an affinity column on which the peptide used for immunization was immobilized. The characteristics of the antibody thus obtained were examined by Western blotting, and the reactivity with the klotho protein was clarified.
  • a specific antibody against the klotho protein was prepared using the purified solubilized klotho protein as an antigen.
  • FGF23 has an important role in the metabolism of phosphorus 1,25D in vivo. For example, It has been suggested that missense mutations in the FGF-23 gene are involved in the induction of hypophosphatemic disease in ADHR patients (The ADHR Consortium. Autosomal dominant hypophosphatemic rickets is associated with mutations in FGF23. Nature Genet. 26 : 345-348, 2000). In addition, we have identified FGF-23 as a tumor-inducing disease-causing factor in neoplastic osteomalacia (Shimada, T., Mizutani, S., Muto, ⁇ ., Yoneya, T., Hino, R.
  • FGF23 may be involved in hypophosphatemic diseases and diseases associated with cull and osteomalacia.
  • XLH the most prevalent of the inherited hypophosphatemia, is said to be present in about 1 in 20,000 people.
  • PHEX the gene responsible for XLH
  • this gene consists of a single exon, and its gene region spans 220 kb, so that mutation analysis that causes a genetic disease is currently impossible for diagnostic purposes. is there. Sudden cases and adult-onset cases have also been suggested. So far, the association between PHEX and FGF-23 has not been clarified.
  • Hyp mice which are spontaneously mutated mice, are known as model animals for XLH. In this mouse, the deletion of the 3 'region of the PHEX gene has been confirmed, and it is known that the mouse exhibits hypophosphatemia, cull disease and osteomalacia.
  • FGF-23 has an activity to reduce blood phosphorus concentration and 1.25D concentration.
  • Experiments on neutralization of FGF-23 activity revealed that FGF-23 plays an important role in maintaining the metabolic balance of phosphorus and 1,25D even in normal health
  • FGF-23 is involved in phosphorus metabolism and vitamin D metabolism, and is associated with diseases such as neoplastic osteomalacia, ADHR, XLH, renal osteodystrophy, Dialysis osteopathy, osteoporosis, hypophosphatemia, rickets, osteomalacia, renal tubular dysfunction, osteopenia, hypocalcemia, hyperphosphatemia, hyper1,25D blood disease, hyperparathyroidism Disease, ectopic calcification, pruritus, bone sclerosis, Paget's disease, hypercalcemia, hypoparathyroidism, bone pain, muscle weakness, skeletal deformity, growth disorders, and hypo1,25D Diseases may be treated by controlling the action of FGF-23.
  • diseases such as neoplastic osteomalacia, ADHR, XLH, renal osteodystrophy, Dialysis osteopathy, osteoporosis, hypophosphatemia, rickets, osteomalacia, renal tubular dysfunction, osteopenia, hypocalcemia, hyperphosphatemia, hyper1,25
  • Example 28 the addition of a complex of FGF23 and solubilized klotho protein stimulates osteoblast-derived cells, which may affect cell morphology and intracellular signal transduction. It became clear. These results suggest that FGF23 and klotho protein directly act on cells related to bone metabolism such as osteoblasts, and that the effects of FGF23 on bone metabolism-related diseases can be measured using klotho protein-anti-klotho antibody. It is thought that treatment can be made possible by controlling the temperature.
  • the complex of FGF23 and klotho protein in the present invention can be produced by mixing an FGF23 molecule (described in W002 / 14504) with a klotho protein as defined above.
  • the FGF23 molecule in the present invention can be produced by appropriately using a known method known in the technical field such as a chemical synthesis method, a cell culture method, or a modification method thereof, in addition to a genetic recombination technique.
  • FGF23 can be produced by the method of Shimada et al ⁇ Endocrinology 143, 3179 (2002) ⁇ .
  • the klotho protein of the present invention is also produced by appropriately using a known method or a modification method known in the technical field such as a chemical synthesis method, a cell culture method, etc., in addition to the genetic recombination technique. be able to.
  • the composite of the present invention can also be produced, for example, by the following production method. That is, it can be prepared by mixing the above-mentioned FGF23 and the klotho protein and leaving them to stand at 4 ° C. to room temperature for several minutes to several hours. In order to further strengthen the binding of the complex, a cross-linking agent such as di succ inimidyl suberate and bis (sul fosucc inimidyl) suberate can be used. Also, in addition to the gene recombination technique, a known method known in the technical field such as a chemical synthesis method, a cell culture method, or the like, may be used for the gene linked to the FGF23 and klotho protein.
  • a cross-linking agent such as di succ inimidyl suberate and bis (sul fosucc inimidyl) suberate
  • a known method known in the technical field such as a chemical synthesis method, a cell culture method, or the like, may be used for the
  • composition comprising the klotho protein of the present invention
  • the klotho protein of the present invention is useful for increasing or decreasing blood phosphate concentration and increasing or decreasing Z or blood activated biminmin D concentration, or for diseases in which increasing or decreasing the action of FGF23 is undesirable.
  • a pharmaceutical composition containing the present klo tho protein or the present klo tho protein changes blood phosphorus, blood active vitamin D concentration or FGF23 action.
  • a disease that causes changes in blood phosphate levels, blood active vitamin D levels, and FGF23 activity such as hyperphosphatemia, hyper1,25D blood disease, renal osteodystrophy, Dialysis osteopathy, osteoporosis, renal tubular dysfunction, osteopenia, hypotensive lucidemia, hyperparathyroidism, ectopic calcification, pruritus, osteosclerosis, Paget's disease, hypercalcemia, vice It can be used to treat hypothyroidism, bone pain, muscle weakness, skeletal deformity, and growth disorders.
  • a pharmaceutical composition comprising the anti-klotho protein antibody of the present invention.
  • the antibody of the present invention has the effect of increasing the blood phosphate concentration and the concentration of Z or active vitamin D in the blood and the effect of inhibiting the action of FGF23 in the body. Therefore, it can be used as a pharmaceutical composition for diseases considered to be affected by changes in blood phosphorus and / or active vitamin D concentration, such as neoplastic osteomalacia, ADHR, and XLH. It can be expected to improve hypophosphatemia, bone calcification insufficiency, bone pain, muscle weakness, skeletal deformity, growth disorders, and low 1,25Demia, which are commonly found in these diseases.
  • FGF-23 plays an important role under physiological conditions, and the antibody of the present invention controls the action of FGF-23 to regulate phosphorus metabolism and the action of FGF-23 to regulate calcium metabolism via vitamin D metabolism
  • minerals such as osteoporosis, rickets, hypercalcemia, hypocalcemia, ectopic calcification, sclerosis, Paget's disease, hyperparathyroidism, hypoparathyroidism, and pruritus Teens It can be used therapeutically and prophylactically as a pharmaceutical composition for diseases caused by abnormalities in vitamin X metabolism and vitamin D metabolism.
  • Pharmaceutical composition comprising the complex of FGF23 and klotho protein of the present invention
  • FGF23 binds to solubilized klotho protein (Example 12), and addition of FGF23 induces intracellular signals (enhanced ERK phosphorylation and increased EGR1 expression) only in the presence of klotho protein (Examples 6, 7, 8) or decreased (Example 13) or increased (Example 13) blood phosphorus and Z or active vitamin D levels in blood in transplantation experiments with solubilized klotho protein-expressing cells. This suggests that FGF23 and klotho protein may alter the concentration of blood phosphorus and / or active vitamin D in the blood by some interaction.
  • the complex of the Klotho protein and FGF23 of the present invention can be used as a pharmaceutical composition for diseases in which an increase or decrease in blood phosphate concentration and an increase or decrease in Z or blood activated biminin D concentration are undesirable. It can be used as a product.
  • the pharmaceutical composition of the present invention containing the klotho protein, the anti-klotho protein antibody, and the complex of the klotho protein and FGF23 as an active ingredient may contain a pharmaceutically acceptable carrier or additive.
  • a pharmaceutically acceptable carrier or additive examples include water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinylpyrrolidone, lipoxyvinyl polymer, nadridium alginate, water-soluble dextran, sodium carboxymethyl starch, Pectin, xanthan gum, gum arabic, casein, gelatin, agar, glycerin, propylene glycol, polyethylene glycol, petrolatum, paraffin, stearyl alcohol, stearic acid, human serum albumin, mannitol, sorbitol, la Kutose, a surfactant acceptable as a pharmaceutical additive, and the like.
  • the additives to be used are appropriately or in combination selected from the above depending on the dosage form of the present invention
  • the prophylactic or therapeutic agent containing the klotho protein, anti-klotho protein antibody, and complex of klotho protein and FGF23 of the present invention can be administered orally or parenterally.
  • solid preparations such as tablets, granules, powders, and pills to be applied thereto, or liquid preparations such as liquids and syrups may be used.
  • liquid preparations such as liquids and syrups
  • granules and powders can be in unit dosage form as capsules, or in the case of liquid preparations, as dry products to be re-dissolved when used.
  • Oral solid preparations of these dosage forms usually contain additives such as binders, excipients, lubricants, disintegrants, wetting agents and the like which are generally used in pharmaceutical preparations in their compositions.
  • Oral liquid preparations usually contain additives such as stabilizers, buffering agents, flavoring agents, preservatives, fragrances and coloring agents which are generally used in preparations in their compositions.
  • injections For injections, they will usually be presented in unit dose ampoules or in multi-dose containers, which may be reconstituted in a suitable carrier, eg, sterile pyrogen-free water, for use. These dosage forms usually contain additives, such as emulsifiers and suspending agents, which are generally used in pharmaceutical compositions.
  • Injection techniques include, for example, intravenous infusion, intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, and intradermal injection. The dose varies depending on the age of the subject, the route of administration, and the number of administrations, and can vary widely.
  • an effective amount of a klotho protein, an anti-klotho protein antibody, a complex of klotho protein and FGF23 of the present invention, an appropriate diluent, and an effective amount administered as a combination with a pharmacologically usable carrier is 0.01 to 100 g, preferably 0.5 to 20 _g / kg body weight at a time for klotho protein or complex.
  • the dose is 0.1 lg to 2 mg, preferably 1 to 500 / zg per kg of body weight at a time.
  • Abnormalities in blood phosphorus and Z or active vitamin D levels can be caused by a variety of disorders, such as hyperphosphatemia, high 1.25D, renal osteodystrophy, dialysis osteopathy, osteoporosis, low Linemia, keratosis, osteomalacia, tubular dysfunction, osteopenia, hypocalcemia, hyperparathyroidism, ectopic calcification, pruritus, osteosclerosis, Paget's disease, hypercalcemia Blood serum and hypoparathyroidism, bone pain, muscle weakness, skeletal deformity, growth disorders and low 1,25D bloodemia, etc., and blood phosphorus and Z or blood active vitamin D levels It is believed that substances that regulate the disease are useful for the treatment of such diseases.
  • the system using the interaction between FGF23 and klotho protein according to the present invention is a substance that treats the above-mentioned diseases (eg, protein, peptide , Carbohydrates, lipids, non-peptidic compounds, Forming compounds, fermentation products, available to explore the biological material).
  • Indicators for searching for such substances include systems that detect intracellular signals due to FGF23-klotho protein interaction (eg, phosphorylation of intracellular proteins, activation of early response genes, intracellular calcium, cAMP, changes in intracellular signaling molecules such as cGMP, changes in cell membrane potential, changes in intracellular pH, release of extracellular signaling molecules, changes in cell morphology, etc.).
  • FGF23 acts on a cell line expressing the full-length klotho protein and inhibits or enhances the interaction between FGF23 and klotho protein by using a system that confirms that intracellular ERK phosphorylation is enhanced. It is possible to search for such substances (small molecules, antibodies, etc.). In addition, by using a system that confirms that intracellular E phosphorylation is enhanced by adding solubilized klotho protein and FGF23 to various cell lines, the FGF23-klotho interaction (eg, It is possible to search for a substance that inhibits or enhances concentration changes or changes in blood active vitamin D concentration).
  • FGF23 is allowed to act on a cell line expressing full-length klotho protein
  • FGF23-klo tho protein interaction by using a system that confirms that EGR1 gene expression increases or a system that uses the EGR1 promoter luciferase reporter gene to detect activation of the EGR1 promoter. Search for substances (small molecules, antibodies, etc.) It is.
  • a system that confirms that the expression of the EGR1 gene is increased by adding solubilized klotiio protein and FGF23 to various cell lines, or that the EGR1 promoter-luciferase reporter is activated every time the EGR1 promoter is activated.
  • FGF23-klo tho protein interaction by using a detection system that uses the overnight gene
  • vitamins that are thought to cause gene expression changes by FGF23-klotho protein interaction]) 1 ⁇ -hydroxylase gene, vitamin D2-4 hydroxylase gene, and other gene changes by FGF23-klotho protein interaction FGF23-klo tho protein interaction eg, changes in blood phosphorus levels or active vitamin D levels
  • FGF23-klo tho protein interaction eg, changes in blood phosphorus levels or active vitamin D levels
  • the present invention relates to a method for decreasing or increasing blood phosphorus concentration and Z or blood active vitamin D concentration by administering klo tho protein into the body, and a method for administering anti-klotho protein antibody to the body to reduce blood phosphorus concentration.
  • a method to increase the concentration of active vitamin D in the blood or blood, a method to enhance or inhibit the action of FGF23 by administering klo tho protein to the body, and a method to increase FGF23 by administering anti-k101 ho protein antibody to the body Methods that inhibit the action of 23 are also included.
  • the present invention relates to klotho for the production of a pharmaceutical composition for lowering or increasing blood phosphorus concentration and / or active vitamin D concentration or for enhancing or inhibiting the action of FGF23 in the body.
  • Anti-klotho protein antibodies for protein use and for the production of pharmaceutical compositions for increasing blood phosphorus and / or blood active vitamin]) levels or for inhibiting the action of FGF23 in the body
  • This description includes part or all of the contents as disclosed in the description and / or drawings of Japanese Patent Application No. 2003-137950, which is a priority document of the present application.
  • Figure 1 is a photograph showing the results of RT-PCR analysis of EGR1 gene expression changes in mouse kidney 1 hour and 8 hours after a single administration of human FGF23 recombinant. is there.
  • Figure 2 shows the results of Western blotting of the changes in ERK protein phosphorylation in mouse kidney at 5 and 10 minutes after a single dose of human FGF23 recombinant. It is.
  • FIG. 3 is a photograph showing the results of Western blotting analysis of changes in ERK protein phosphorylation 10 minutes after the addition of human FGF23 recombinant to PEAK cells expressing full-length klotho protein.
  • FIG. 4 shows changes in luciferase activity during stimulation of human FGF23 in PEAK cells transfected with a reporter gene linked to luciferase downstream of the EGR1 promoter together with a klotho expression plasmid.
  • FIG. 5 is a photograph showing the enhancement of ERK phosphorylation in PEAK cells when human FGF23 was added to the culture supernatant of CHOras clone l cells stably expressing soluble mouse klotho protein in PEAK cells.
  • Figure 6 shows the results of the mklotho-97 antibody, mklotho-118 antibody, and mklot o-299, which were obtained by using a solubilized mouse klotho protein transiently expressing PEAK rapid cell supernatant sample and a PEAK raid cell culture supernatant sample as a control.
  • 4 is a photograph showing the results of analysis by Western blotting using an antibody and the mklotho-941 antibody.
  • Fig. 7B shows a sample of the supernatant of the solubilized mouse klotho protein transiently expressing PEAK rapid cells, a sample of the culture supernatant of PEAK rapid cells as a control, and the FGF-23-immobilized resin.
  • 4 is a photograph showing the result of analyzing a sample of an FGF-23-conjugated substance subjected to the above by Western blotting using the mklotho-118 antibody or the mklotho-941 antibody.
  • Figure 7A is a photograph showing the result of a similar sample stained by the silver staining method.
  • 3 is a graph showing serum calcium concentrations. Results are mean soil standard error The significance test with the control group was performed by Student's t.
  • FIG. 9A is a graph showing the serum phosphorus concentration of the anti-klotho protein antibody (P118) group and the vehicle administration group 24 hours after administration.
  • results are expressed as mean soil standard errors, and a significant difference test with the control group was performed by Student's st.
  • FIG. 9B is a diagram showing serum active vitamin D concentrations 9 hours after administration in the anti-klotho protein antibody (pi 18) group and the vehicle administration group.
  • results are expressed as the standard error of the mean, and a significant difference test with the control group was performed by Student's st.
  • Figure 10A shows CHO-FGF23H cell culture supernatant separated by SDS-polyacrylamide gel electrophoresis and detection of recombinant FGF-23 protein and its metabolite by Western blotting with anti-FGF-23 polyclonal antibody It is a photograph taken.
  • the culture supernatant there are a full-length FGF-23H protein and an N-terminal fragment and a C-terminal fragment generated by cleavage between SEQ ID NO: 179 and SEQ ID NO: 180 of the amino acid sequence of SEQ ID NO: 35.
  • the hFGF23-48 antibody and the hFGF23-148 antibody recognized the full-length FGF-23H protein and the N-terminal fragment peptide. The presence of small fragmented peptides is observed in the N-terminal fragment.
  • the anti-His6_tag antibody full-length protein and C-terminal fragment were detected.
  • Figure 10B shows that FGF-23 full-length protein purified from CHO-FGF23 cell culture supernatant, N-terminal fragment and C-terminal fragment were separated by SDS-polyacrylamide gel electrophoresis and detected by CBB staining. It is a photograph.
  • FIG. 11 shows the results obtained when the anti-FGF-23 antibody (2C3B) was added during stimulation of human FGF-23 in PEAK cells transfected with a klotho expression plasmid and a reporter gene linked to luciferase downstream of the EGR1 promoter.
  • FIG. 3 is a diagram showing a change in luciferase activity of the present invention. It was shown that luciferase activity increased by FGF-23 stimulation was suppressed by simultaneous addition of 2C3B.
  • FIG. 12 shows whether anti-FGF23 antibody (2C3B) suppresses the increase in ERK phosphorylation caused by the addition of FGF23 and the culture supernatant of CHOras clonel cells, which express soluble klotho protein in CHOras clonel cells.
  • 4 is a photograph showing the results of the measurement. It was shown that the enhancement of ERK phosphorylation when FGF23 and solubilized klotho protein-expressing CHOras clonel cell culture supernatant were added simultaneously was suppressed in a 2C3B antibody concentration-dependent manner.
  • FIG. 13 is a diagram showing the concentrations of phosphoric acid in serum and 1,25-dihydroxyvitamin D in serum 24 hours after administration of the 2C3B antibody, anti-TPO antibody, and vehicle.
  • FIG. 14 is a photograph showing the purified solubilized klotho after SDS-PAGE and silver staining and detection with an anti-klotho antibody (mklotho-118 antibody).
  • FIG. 15 is a photograph in which solubilized klotho protein was detected by mastl blotting using mskl antibody-1 and mskl antibody-2.
  • FIG. 16 is a photograph showing immunoprecipitation of purified solubilized dolot using an anti-mouse klotho monoclonal antibody, and detection of the precipitated solubilized klotho by Western blotting using the fflklotho-118 antibody.
  • FIG. 17A is a graph showing the effect of mskl antibody-1 on the FGF23 action in the EGR1 Atsushi strain on IS activity.
  • FIG. 17B is a diagram illustrating the inhibitory effect of the mskl antibody-2 on the FGF23 action in the EGR1 Atsushi system.
  • FIG. 18A shows the effect of administration of mskl antibody-1 and mskl antibody_2 to mice on serum dihydroxyvitamin D.
  • FIG. 18B shows the effect of administration of mskl antibody-1 and mskl antibody-2 to mice on serum phosphorus concentration.
  • FIG. 19 is a graph showing the effect of the anti-klotho monoclonal antibody on FGF23 action in the EGR1 Atsushi strain.
  • FIG. 2 OA shows the effect of administration of an anti-klotho monoclonal antibody to mice on serum dihydroxyvitamin D concentration.
  • FIG. 20B shows the effect of administration of an anti-klotho monoclonal antibody to mice on serum phosphorus concentration.
  • FIG. 21 is a photograph showing the effect of adding purified solubilized klotho and FGF23 on the enhancement of ERK phosphorylation in HEK293 cells. '
  • FIG. 22 is a photograph showing the effect of the addition of purified solubilized klotho and FGF23 on the cell morphology of MC3T3 cells.
  • FIG. 23 shows the effect of administering purified solubilized klotho to mice on serum dihydroxybiminin D concentration.
  • FIG. 24A is a diagram in which binding between klotho expressed in HEK293 cells and FGF23 on cells was detected by FACS.
  • FIG. 24B shows the results of a control in which cells do not express klotho.
  • Example 1 Confirmation of the effect of increasing the expression of egrl in the kidney by administration of FGF23 to mice
  • Human membrane array Atlas mouse 1.2k to search for genes whose expression fluctuates in the kidney when FGF23 is administered to mice , Atlas mouse 1.2kII (Clontech, USA).
  • the membrane array was prepared according to the attached protocol.
  • the samples used were human kidney, human kidney 1 hour after administration of FGF23 (5 / g / head), human
  • EGR1 was found as a gene whose expression increased one hour after administration of human FGF23.
  • First strand cDNA was prepared using the above-mentioned three kinds of mouse kidney total RNA, and RT-PCR was performed. First strand cDNA was synthesized using First strand cDNA synthes is kit (Invitrogen, USA) according to the attached protocol. First strand cDNA was prepared using total RNA extracted from mouse kidney 1 hour after administration of human FGF23, mouse kidney 8 hours after administration of human FGF23, and mouse kidney after administration of vehicle. Synthesized.
  • FW primer for mouse EGR1 RT-PCR CTTAATACCACCTACCAATCCCAGC (SEQ ID NO: 4)
  • RV primer for mouse EGR1 RT-PCR GTTGAGGTGCTGAAGGAGCTGCTGA (SEQ ID NO: 5)
  • FIG. 1 FW primer for mouse EGR1 RT-PCR: CTTAATACCACCTACCAATCCCAGC (SEQ ID NO: 4)
  • RV primer for mouse EGR1 RT-PCR GTTGAGGTGCTGAAGGAGCTGCTGA (SEQ ID NO: 5)
  • FIG. 1 As shown in FIG. 1, these PCR products were electrophoresed on a 2% agarose gel. However, an increase in EGR1 expression was confirmed only in the mouse kidney one hour after administration of human FGF23. On the other hand, when the target G3PDH primer was used, the same level of expression was observed in all samples. From these results, it was confirmed that 1 hour after the administration of FGF23, EGR1 expression was increased in mouse kidney.
  • the frozen kidney was extracted with extraction buffer (20 mM Hepes (pH 7.5), 150 ⁇ NaCl, 1 NP-40, lmM EDTA, 0.1% SDS, 50niM sodium fluoride, 2mM sodium Vanadate, 1 tab./7ml COMPLETE (US (Roche Co., Ltd.)) and homogenized with a Polytron homogenizer. After standing on ice for 2 hours, the mixture was centrifuged at 1000 G for 10 minutes, and the supernatant fraction was used for subsequent experiments. The protein concentration of the supernatant fraction was measured with a BCA kit (PIERCE, USA), and all samples were prepared with an extraction buffer to a protein concentration of 22.7 mg / ml.
  • extraction buffer 20 mM Hepes (pH 7.5), 150 ⁇ NaCl, 1 NP-40, lmM EDTA, 0.1% SDS, 50niM sodium fluoride, 2mM sodium Vanadate, 1 tab./7ml COMPLETE (US (Roche Co
  • the full-length cDNA cloning of the mouse kl 01 ho gene was performed by PCR.
  • the following primers were synthesized based on GeneBank data (Accession No. AB005141).
  • the primer of SEQ ID NO: 6 contains a primer i? On the 5 'side of the mouse klotho gene, an initiation codon and a Kozak sequence portion of 5 bases upstream thereof. In addition, an EcoRI site has been added.
  • the primer of SEQ ID NO: 7 is a primer on the 3 'side of the mouse klotho gene, which contains a termination codon and a Not I site.
  • mouse klotho cDNA cloned using these primers As a result of the analysis, it completely matched the mouse klotho gene, and the cloning of the mouse klotho gene was completed.
  • this plasmid is referred to as mklotho / pEAK8.
  • mouse klotho cDNA obtained by digesting the mklotho / pEAK8 'vector with EcoRI and Notl is ligated to plasmid pEAK8 by linking an intramolecular ribosome entry site (IRES) and enhanced green fluorescent protein (EGFP).
  • IRES-EGFP-pEAK8 was obtained by inserting it into EcoRI and Notl sites of the prepared vector IRES-EGFP-PEAK8.
  • Example 4 Cloning of gene encoding mouse-solubilized klotho protein
  • the mouse kl01ho gene prepared in Example 3 was solubilized into type II to produce a klotho protein.
  • the following primers were synthesized for production.
  • RV3 primer for cloning mouse-solubilized klotho protein-encoding gene
  • the primer of SEQ ID NO: 20 contains up to 2940 bases of the mouse klotho gene, adds a TAA termination codon, and further adds a Notl site.
  • the PCR reaction was carried out using the primers of SEQ ID NO: 8 and SEQ ID NO: 20 and Pyrobest Polymerase (Yukara, Japan). That is, TaKaRa Pyrobest polymerase 0.5 10X Pyrobest Buffer
  • mouse solubilized klotho cDNA was digested with EcoRI and Notl, and plasmid pEAK8 (US Edge) BioSystems Inc.) into the EcoRI.NotI site of the vector IES-EGFP-PEAK8, which was created by ligating an intramolecular ribosome entry site (IRES) and enhanced green fluorescent protein (EGFP).
  • IRES intramolecular ribosome entry site
  • EGFP enhanced green fluorescent protein
  • the plasmid-mklotho / IRES-EGFP-pEAK8 plasmid was introduced into CHOras clone cells according to the package insert.
  • G Select drug-resistant cells in ⁇ medium containing 5 g / ml pure bitemycin and 10% FBS, and use FACS vantage (Becton Dickinson, USA) to select cells with strong EGFP (Green Fluorescent Protein) emission. Cloned by sorting. When the cloned cells became confluent, the medium was replaced with serum-free DF (DMEM / F-12) medium, and the supernatant was recovered 2 days later. Collect the supernatant using anti-klotho protein antibody
  • the mouse full-length klotho protein was transiently expressed in PEAKrapid cells (Edge Biosystems, USA) using the mouse full-length klotho protein expression plasmid prepared in Example 3, and the intracellular ERK phosphorus was stimulated with FGF23. The enhanced oxidation was verified.
  • the mklotho / pEAK8 plasmid prepared in Example 3 was transiently transfected into a 12-well plate according to the protocol attached to the PEAKrapid cells. Trans Hue transfected 2 4 hours later the medium was replaced with DMEM medium without serum, additional 2 4 h (;. 0 2 Inkyube Isseki was performed Inkyubeto within one heparin (U.S.
  • mice solubilized klotho protein-expressing CHOras clonel cells prepared in Example 5 were seeded in a tissue culture flask (225 cm 2 , Corning, USA). After culturing for 24 hours, the medium was replaced with a serum-free ⁇ medium, and the cells were cultured at 37 for 5 days, and the culture supernatant was collected.
  • CHOras clone cells and HEK293 cells were seeded on 24-well plates (Coming, USA). After culturing for 24 hours, the culture medium was replaced with the mouse-soluble solubilized klotho protein-expressing CHOras clone cell culture supernatant prepared in 8- (1), and culturing was further performed for 4 hours. Subsequently, 10 g / ml of heparin was added, and then human FGF23 (100 ng / ml) was added. After performing incubation at 10 minutes 37, the supernatant was aspirated, washed with PBS, and lysed with SDS-PAGE sample buffer (Daiichi Kagaku, Japan).
  • Example 9 EGR1 Promoter ⁇ klotho protein expression using luciferase Acetase in PEAK cells
  • mouse EGR1 promoter-luciferase fusion gene was prepared. Cloning of the mouse EGR1 promoter was performed by PCR. Primers were prepared as follows based on the literature (Barbara, A. et al., Proc. Natl. Acad. Sci. USA Vol. 85, pp. 7857-7861).
  • the primer of SEQ ID NO: 21 is a primer starting from the ⁇ 1023 base portion of the mouse EGR1 promoter, and a Hindlll site is further added.
  • the primer of SEQ ID NO: 22 is a primer including the mouse EGR1 mRNA start site (+1) and has a Kpnl site added thereto.
  • mouse type ⁇ ⁇ ⁇ mouse Genomic DNA (Clontech, USA) was used. The PCR reaction was performed using LA Taq GC Buffer (Takara Shuzo, Japan) according to the attached manual.
  • the PCR reaction conditions were as follows: primary denaturation at 94 ° C for 5 minutes, followed by 30 cycles of 94 ° C for 20 seconds, 60 ° C for 30 seconds, and 72 ° C for 1 minute, and finally elongation. The reaction was performed at 72 ° C for 7 minutes.
  • the amplified product by PCR was confirmed by electrophoresis on a 2% agarose gel. As a result, a single band was detected at about lkbp. The band was cut out from the gel, and the GeneClean kit (US Bio 1) was used.
  • the transfection was performed in a 10 cm-diameter culture dish (Becton Dickinson, USA). Four hours after transfection, the cells were detached with PBS containing 0.53 mM EDTA, and seeded on a white 96-well plate (Corning, USA). After culturing for 24 hours, human FGF23 was added at 0 to 10 Ong / ml, and heparin (Sigma, USA) was also added so as to be 10 g / ml. After standing still at 37 ° C for 24 hours, luciferase activity was measured using a Steady-Glo Luciferase assay system (Promega, USA) according to the protocol. The measurement was performed using a TopCount (US-based Pacikido). The results are shown in FIG.
  • peptide mklotlio-97 (SEQ ID NO: 24) with a cysteine residue added to the C-terminal of a peptide consisting of 16 amino acids starting from histidine at residue number 97 in SEQ ID NO: 23, starting from proline at position 118 21
  • Peptide mklotho-118 (SEQ ID NO: 25) with a cysteine residue added to the N-terminus of the peptide consisting of amino acid residues
  • peptide mklotho-299 (SEQ ID NO: 26) consisting of 20 amino acid residues starting from alanine at position 299
  • a peptide mklotho-941 (SEQ ID NO: 27) consisting of 25 amino acid residues starting from the 941th proline was selected as an antigen and chemically synthesized.
  • mklotho- 118 CPPLSSTGDVASDSYNNVYRDT (SEQ ID NO: 25)
  • mkl ot o-299 ALSS-INPR TDYNIREC (SEQ ID NO: 26)
  • peptides were bound to carrier protein ⁇ -thyroglobulin via their cysteine residues and subjected to immunization. Immunization was performed using two egrets for one antigen peptide. In the first immunization, a peptide conjugated to lOO g of a carrier protein per bird was emulsified with Freud's complete adjuvant and administered intradermally or subcutaneously to a egret. One week after the first immunization, a peptide conjugated to lOO g of the carrier protein was emulsified with incomplete Freund's adjuvant and administered similarly. The same administration was performed six times at two-week intervals, and one week after the final administration, whole blood was collected to prepare an antiserum.
  • each peptide used for immunization was gel-prepared using SulfoLink Kit (PIERCE, USA). Was immobilized. Antiserum was added to this column using PBS (-) as an adsorption buffer, and the antibody that binds to the peptide used for immunization was retained on the column. Next, the antibody bound to the column was eluted and recovered using a 0.1 M glycine buffer (pH 2.8) as an elution buffer. The eluted fraction was adjusted to around pH 7.2 by adding 1M Tris-HCl (H 9.0).
  • the prepared antibody solution was applied to a gel filtration column NAP25 (Amersham Pharmacia Biotech, USA) to replace the buffer with PBS (-), and then a membrane filter with a pore size of 0.MILLEX-GV (Mill, USA) The solution was sterilized by filtration using ipore) to obtain antibodies against each peptide.
  • the concentration of the purified antibody was calculated by measuring the absorbance of 280 dishes and setting lmg / ml to 1.350D.
  • Each of the purified antibodies thus obtained will be indicated using the name of the peptide used for immunization.
  • an antibody obtained by immunizing the mklotho-97 peptide of SEQ ID NO: 24 will be referred to as a mklotho-97 antibody.
  • mice solubilized klot o protein transiently expressed PEAK rapid cell supernatant prepared in Example 8
  • Mouse solubilization in mouse The protein was analyzed by Western plotting. As shown in FIG. 6, mouse-solubilized k-lotho protein near 130 kDa was detected for all antibodies. Therefore, these four types of antibodies were found to be antibodies that can recognize mouse klotho.
  • Piotin labeling was performed using polyclonal antibodies against the above-mentioned purified four partial peptides of the mouse klotho protein.
  • FGF-23-immobilized resin 50 ⁇ 1 was added, and the mixture was inverted at 4 ° C for 1 hour to react the solubilized mouse klotho protein with FGF-23. Thereafter, the resin was washed four times with PBS to remove unreacted substances. 300 1 sample buffer (50 mM
  • FIG. 7 shows the results.
  • Fig. 7 also shows the results of silver staining of a similar sample using 2D-silver staining reagent II II (Daiichi Chemical Co., Ltd., Japan). 'These results indicated that the solubilized mouse klotho protein was bound to the FGF-23RQH-immobilized column and concentrated. Therefore, it was clarified that FGF-23 binds to klotho protein.
  • solubilized klotho protein recombinant stably expressed CH0 cells obtained in Example 5 were subcutaneously implanted into nude mice to obtain the solubilized klotho protein recombinant.
  • Seven-week-old male BALB / c nude mice were used as experimental animals, and cell transplantation was performed as follows. Mice were kept in plastic cages throughout the period from their arrival to dissection and were allowed free access to commercially available rodent chow and tap water.
  • mice were intraperitoneally administered once with the anti-klotho protein antibody P118 obtained in Example 10 at 200 g / O. 2 mL / mouse.
  • 0.2 mL of the solvent (PBS) was intraperitoneally administered.
  • blood was collected from the orbit using a glass capillary, serum was separated using Microtina (Becton Dickson, USA), and 24 hours later, blood was collected from the abdominal vena cava, using Microtina again. To separate the serum.
  • the concentration of 1,25-dihydroxyvitamin D was measured using a 1,25 (0H) 2D RIA kit “TFB” (TFB, Japan).
  • the serum phosphate concentration 24 hours later was determined using Phosphor-Test II Co. (Wako Pure Chemical Industries, Japan) according to the package insert.
  • the anti-klotho protein antibody and the control group consisted of 6 mice each, and were supplied with tap water and a solid diet containing 1.03% inorganic phosphate and 1.18% calcium CE2 (CLEA Japan, Japan). They were allowed free intake.
  • the anti-klotho protein antibody can increase the serum phosphorus concentration and the serum activated vitamin D concentration.
  • the cDNA coding for FGF-23 was derived from a cDNA library of the tumor responsible for neoplastic osteomalacia, type III, using the FlEcoRI primer (SEQ ID NO: 28), the LHisNot primer (SEQ ID NO: 29), and LA- After incubating at 96 ° C for 1 minute using Tad DNA polymerase,
  • Amplification was performed by performing 35 cycles of PCR, in which a cycle consisting of 30 seconds at 55, 30 seconds at 55, and 30 seconds at 72 ° C was one cycle.
  • the FlEcoRI primer anneals to a sequence located further 5 ′ upstream of the nucleotide sequence encoding FGF-23, and Add an EcoRI restriction enzyme site 5 'to the region encoding FGF-23.
  • the LHisNot primer is a sequence that anneals to the sequence 5 'of the stop codon of the sequence encoding FGF-23 and the sequence following the sequence that encodes the His6_tag sequence (His-His-His-His-His-His-His-His-His). Contains Don and Notl restriction enzyme sequences.
  • the amplified fragment encodes the FGF-23 protein with a His6-tag sequence added to the C-terminus, and has a Notl restriction enzyme site downstream.
  • This amplified fragment was digested with EcoRI and Notl, and ligated to pcDNA3.lZeo (Invitrogen, USA), which was an animal cell expression vector similarly digested with EcoRI and Notl.
  • the expression vector thus prepared was cloned, and its nucleotide sequence was determined. It was confirmed that the FGF-23 protein protein to which the desired His6-1-ag sequence had been added was encoded. This vector is called pcDNAFGF-23H.
  • FlEcoRI CCGGAATTCAGCCACTCAGAGCAGGGCACG (SEQ ID NO: 28)
  • Expression vector pCAGGS (Niwa H, et al., Gene. 1991, 108: 193-199) was digested with EcoRI, blunt-ended with Klenow fragment (Roche, Switzerland), and the small intestine alkaline phosphatase ( Dephosphorylation was performed using Takara Shuzo, Japan. The thus prepared cDNA fragment encoding FGF-23 was ligated to the pCAGGS vector. The expression vector thus prepared was cloned and its nucleotide sequence was determined to confirm that the sequence encoding the desired FGF-23 protein was correctly inserted. This vector is called pCAGGS / FGF-23.
  • the FGF-23-encoding fragment amplified with the FlEcoRI and LNot primers was digested with EcoRI and Notl and purified. This is the expression vector
  • Intramolecular ribosome entry sequence in PEAK8 (Edge Biosystem, USA)
  • EGFP enhanced green fluorescent protein
  • the nucleotide sequence of the obtained plasmid was determined, and it was confirmed that it encodes the FGF-23 protein.
  • This vector is called PEAK8 / IRES / EGFP / FGF-23.
  • the FGF-23 protein was found to be susceptible to protein cleavage between the Arg residue at position 179 and the Ser residue at position 180.
  • the N-terminal amino acid sequence at this cleavage site is Argl76-Hisl77-Trl78-Argl79 (SEQ ID NO: 31) corresponding to the Arg-XX-Arg sequence, which is a recognition sequence of the protein converting enzyme. Is known to be a substitution mutation at the 176th or 179th Arg residue. Therefore, we developed a model for the mutant FGF-23 found in ADHR that has resistance to cleavage by protein converting enzyme, has a His6-tag at the C-terminus, and removes the 176th and 179th Arg residues from the Gin residue.
  • a vector was constructed to experimentally produce a mutant FGF-23 protein (hereinafter referred to as FGF-23RQH) substituted with a group.
  • RQF SEQ ID NO: 32
  • RQR SEQ ID NO: 33
  • ME1 (SEQ ID NO: 34) and HNt (SEQ ID NO: 35), which are primers for amplifying the FGF-23 sequence, were prepared.
  • ME1 contains the start codon encoded by the FGF-23 cDNA
  • HNt can insert a His6-tag sequence-encoding codon sequence before the stop codon encoded by FGF-23 cDNA, and add a Notl restriction enzyme sequence.
  • the reverse primer can be.
  • RQF ATACCACGGCAGCACACCCAGAGCGCCGAG (SEQ ID NO: 32)
  • RQR CTCGGCGCTCTGGGTGTGCTGCCGTGGTAT (SEQ ID NO: 33)
  • ME1 ATGAATTCCACCATGTTGGGGGCCCGCCTCAGG (SEQ ID NO: 34)
  • HNt ATGCGGCCGCCTAATGATGATGATGATGATGGATGAACTTGGCGAAGGG (SEQ ID NO: 35)
  • PCR reaction was performed with primers (200 nM each) of the combination of RQF and HNt and the combination of ME1 and RQR.
  • the reaction was carried out using pfu DNA polymerase (Promega, USA) at 94 ° C for 1 minute, followed by one cycle consisting of 94 ° C for 30 seconds, 55 ° C for 30 seconds, and 72 ° C for 1 minute. 25 cycles were performed.
  • the two kinds of reaction solutions thus obtained were diluted 10-fold, and 1/21 of each was mixed to form a ⁇ , and ME1 and HNt were added to a final concentration of 200 nM.
  • PCR reaction solution incubate at 94 ° C for 1 minute, and perform 25 cycles of PCR reaction consisting of a process consisting of 30 seconds at 94 ° C, 30 seconds at 55 ° C, and 1 minute at 72 ° C. Carried out.
  • LA Ta ⁇ A polymerase (Takara Shuzo, Japan) was used.
  • the obtained amplification product of about 800 bp was digested with EcoRI and Notl, and then purified to obtain an insert DNA.
  • PEAK8 / IRES / EGFP vector in which an intramolecular liposome entry sequence (IRES) and enhanced green fluorescent protein (EGFf) are linked to the expression vector PEAKS (Edge Biosystem, USA). It was inserted into the enzyme site and cloned. The nucleotide sequence of the obtained plasmid, whose sequence is determined and encodes the mutant FGF-23 protein with the desired Arg at positions 176 and 179 converted to Gin and a His6-tag sequence added to the C-terminus I was sure that .
  • This vector is called PEAK8 / IRES / EGFP / FGF-23RQH.
  • One cell contains 107 ( ⁇ 01 ⁇ 5 (10116-1 cells (Shirahata, S., et al. Biosci Biotech Biochem, 59: 345-347, 1995), and the gene was introduced into cells by electroporation using Gene Pulser II (Bio Rad, USA). After culturing the cells for 24 hours in a MEM ⁇ ; culture medium (Gibco BRL, USA) containing 10% FCS, add Zeocin (Invitrogen, USA) to a final concentration of 0.5 mg / ml for 1 week Cultured.
  • the cells that adhered and proliferated were released with trypsin and cloned by the limiting dilution method in the presence of Zeocin at a final concentration of 0.3 mg / ml to obtain 35 cloned cells.
  • Zeocin at a final concentration of 0.3 mg / ml to obtain 35 cloned cells.
  • FGF-23H protein Cells that best expressed FGF-23H protein were identified by Western blotting as described below. After collecting the culture supernatant of each cloned cell and performing SDS-polyacrylamide electrophoresis, the protein was transferred to a PVDF membrane (Millipore, USA), and an anti-His-tag (C-terminal) antibody (US, Invitrogen) and an ECL luminescence system (Amersham Pharmacia Biotech, USA) were used to detect a signal derived from the FGF-23H protein of about 32 kDa.
  • CH0-0ST311H is referred to as CHO-FGF23H.
  • the PEAK8 / IES / EGFP / FGF-23 and EAK8 / IRES / EGFP / FGF-23RQH vectors were introduced into CH0 Ras clone-1 cells by the gene transfer method using membrane fusion lipid.
  • the band with a small molecular weight confirmed the amino acid sequence from the 180th position of SEQ ID NO: 36, and was found to be a fragment generated by cleavage between the 179th position and the 180th position.
  • the presence of a peptide thought to have a sequence up to the 179th position was also confirmed.
  • the N-terminal sequence from the 179th position also had an affinity for the metal column and was effective for purification. Furthermore, purification was performed using an SP-Sepharose column, and it was possible to obtain full-length FGF-23H as a single band by CBB staining. The result is shown in FIG. 10B.
  • the FGF-23 protein can be purified in a similar manner.
  • the culture supernatant of CH0-FGF23 was purified using SuperCap (registered trademark), a membrane having a pore size of 0.2 m (Pall, USA).
  • fraction containing the target protein was adsorbed and eluted on a SP Separose FF column and purified.
  • full-length FGF-23RQ protein was purified from the CHO-FGF23RQ supernatant.
  • the preparation of the monoclonal antibody in this example was prepared according to a general method as described in "Introduction to Monoclonal Antibody Experimental Procedures" (written by Tamoe Ando et al., Published by Kodansha 1991).
  • BALB / c mice were used as immunized animals, and human FGF-23 was immunized by the following two methods depending on the difference in immunogen.
  • the FGF-23RQH protein (20 to 30 g / animal) prepared in Example 15- (4) was suspended in RIBI adjuvant (Corixa, USA) containing squalene, Tween80, Monophosporyl lipid A and Trehalose dimycolate, and emulsion was added.
  • RIBI adjuvant Corixa, USA
  • Example 15- (4) For BALB / c mice? 6? -23 (22 / animal) were suspended in the above-mentioned 81 adjuvant and primed by intraperitoneal injection. In addition, the same protein was boosted once a week by intraperitoneal injection for 4 weeks, and FGF-23 (10 zg / animal) was immunized by tail vein injection 3 days before the acquisition of spleen cells described below. . 16- (3) Preparation and selection of high prioma
  • the spleen was excised from the mouse immunized as described above, and the spleen cells collected from the spleen were mixed with mouse myeloma SP2 / 0 (ATCC: CRL 1581) at a ratio of 5: 1, and polyethylene glycol 1500 (Roche, Japan) was used as a fusion agent.
  • the cells were fused using Diagnostics, Inc. to produce hybridomas.
  • the selection of hybridomas was carried out using a HAT medium containing HAT containing 10% fetal calf serum (Fetal Calf SermiK FCS) and hypoxanthine (H), aminopterin (A) and thymidine (T) (US , Gibco BRL).
  • cloning was performed by a limiting dilution method using HT-containing DMEM medium. A cloned hybridoma derived from a single cell was obtained. 16- (4) Selection of cloned hybridoma producing anti-FGF-23 antibody
  • hybridoma producing an antibody that specifically recognizes the FGF-23 protein was selected.
  • the selection of hybridomas obtained by immunization according to the first method described above was carried out as follows. Diluted to a concentration of 1 zg / ml in a solution of 50 mM NaHCO 3 FGF the 23H protein solution, a 96-well microplate for ELISA (Maxisorp (TM), USA, Nunc, Inc.) was added in 50 1 in each Ueru of 37 Incubate for 30 minutes at ° C or 12 hours at 4 ° C,
  • the FGF-23H protein was adsorbed on the microplate. The solution was then removed and each well was filled with a blocking reagent (SuperBIock® Blocking Buf fer, USA,
  • the plate was washed twice with Tris-buf fered saline containing Tween20 (TRIZMA pre-setcrys tals (registered trademark, Sigma, USA) containing 500 mM NaCl) (T-TBS).
  • Tween20 Tris-buf fered saline containing Tween20
  • 50 1 of the culture supernatant of each hybridoma was added and reacted for 30 minutes.
  • each well was washed twice with T-TBS.
  • a peroxidase-labeled goat anti-mouse IgG antibody Zymed Laboratories, USA
  • diluted 3000-fold to 50: 1 was added to each well, and incubated at room temperature for 30 minutes.
  • hybridomas obtained by immunization according to the second method described above was carried out as follows. Diluted in a solution of 50 mM NaHCO 3 to a concentration of l / ig / ml FGF- 23 protein solution,
  • each well of the microplate coated with FGF-23 protein in this manner add 50 1 of the culture supernatant of each hybridoma and react for 30 minutes.After that, each well contains 0.1% Tween20. Washed twice with Tris-bui iered saline (T-TBS). Next, peroxidase-labeled goat anti-mouse IgG antibody (Zymed Laboratories, USA) diluted 3000-fold was added to each well, and incubated at room temperature for 30 minutes. After washing the plate three times with T-TBS, 50 ml of a substrate buffer containing tetramethylbenzidine (DAK0, Denmark) was added to each well, and the mixture was incubated at room temperature for 15 minutes.
  • DAK0 tetramethylbenzidine
  • the anti-FGF-23 antibody-producing hybridoma was ligated with 10 g / ml of periinsulin (Sigma, USA), 5.5 g / ml of human transferrin (Sigma, USA), and 0.1 lmM of ethanolamine ( Adapted to eRDF medium (Farto Pharmaceuticals, Japan) containing 5 ng / ml sodium selenite (Sigma, USA).
  • Culture of the eight hybridomas to prepare antibodies was performed in spinner flasks. The culture solution was passed through a filter having a pore size of 0.2 Xm (Pall Gelman Laboratory, USA) to remove foreign substances such as hybridomas, and the antibody-containing culture supernatant was recovered.
  • the culture supernatant containing the anti-FGF-23 antibody was applied to a protein G Sepharose 4 FF column (US,
  • the antibody was adsorbed onto the column through Amersani Pharmacia Biotech) and eluted with 0.1 M glycine buffer (PH2.8). The eluted fraction is added with 1 M Tris_HCl. The pH was adjusted to 7.2.
  • the antibody solution thus prepared was subjected to dialysis substitution with PBS (-) using a membrane with a cut-off molecular weight of 10,000 cuts (Spectrum Laboratories, U.S.A.), and a 0.22 m membrane filter MILLEX- The solution was sterilized by filtration with GV (Millipore, USA) to obtain a purified anti-FGF-23 antibody. The concentration of the purified antibody was calculated by measuring absorbance at 280 nm and setting lmg / ml to 1.350D.
  • anti-FGF-23 antibody Using a protein A carrier column (Immune Biological Laboratories, Japan), glycine buffer (pH 8.9) as the adsorption buffer, and citrate buffer (pH 4.0) as the elution buffer, anti-FGF- The antibody was affinity purified from the culture supernatant containing the 23 antibody. 1M Tris-HCl was added to the eluted fraction containing this antibody to adjust the pH to around 7.2. Next, the solution containing the antibody was replaced with PBS (-) using a dialysis membrane, and further sterilized by filtration with a membrane filter having a pore size of 0.22 m to obtain a purified anti-FGF-23 antibody. The concentration of the purified antibody was calculated by measuring the absorbance at 280 mn and setting lmg / ml to 1.350D.
  • each of the purified monoclonal antibodies thus obtained will be indicated using the name of the produced hybridoma.
  • an antibody produced by hybridoma 1D6A will be referred to as 1D6A antibody.
  • the anti-FGF-23 antibody (2C3B) prepared in Example 15 was examined.
  • the method was performed according to Example 9, and the 2C3B antibody was added to a final concentration of 2.5 Ig / ml immediately before the addition of FGF-23.
  • the 2C3B antibody significantly suppressed the increase in luciferase caused by FGF-23 (FIG. 11). This indicates that the Atssey system in klotho protein-expressing cells using EGR-1 Promoter-Luciferase is effective in searching for substances that suppress signal transduction of FGF-23 and klotho protein.
  • EGR-1 Promoter-Luciferase is effective in searching for substances that suppress signal transduction of FGF-23 and klotho protein.
  • Example 20 Effect of administration of anti-FGF-23 antibody "2C3B antibody” on mice
  • Normal mice BALBA; male, 12 weeks old
  • PBS was used as a vehicle in group 1 and 0 in group 1
  • a single dose of 0.15 ml each of 67 mg / ml anti-human FGF-23 monoclonal antibody (2C3B) and 0.67 mg / ml anti-TP0 monoclonal antibody as a control was injected into the tail vein. .
  • the mouse solubilized klo tho protein stably expressing cells prepared in Example 5 were cultured in a roller bottle (Falcon, USA) in SFMI I medium (Invitrogen, USA) for 5 days. After removing the suspended cells from the culture supernatant with a Millistack DE60 filter (Millipore, USA), the supernatant was concentrated approximately 10-fold with a SART0C0N Cassete Hydrosart Cu toff 30000 (Sartorius, Germany). . Concentrated culture supernatant was adjusted to pH 7.95 with NaOH, and then adsorbed to Q Sepharose TM FF (Amersham Biosciences, USA).
  • FIG. 14 shows the results of silver staining of the purified product and the results of Western blotting using the anti-klotho antibody prepared in Example 10.
  • the mouse solubilized klotho protein purified by the method described in Example 21 was used for immunization as an antigen. 'Immunization went to two birds.
  • 50 tig of an antigen per rabbit was emulsified with Freund's complete adjuvant and administered intradermally or subcutaneously to the rabbit.
  • an antigen prepared by emulsifying with Freund's incomplete adjuvant was administered in the same manner. The same administration was performed six more times at two-week intervals, and one week after the final administration, whole blood was collected to prepare antisera.
  • Antiserum containing anti-mouse-solubilized KL 0 tho antibody was transferred to Protein G Sepharose 4 FF column. (Amersham Pharmacia Biotech, USA) to adsorb the antibody to the column and elute it with 0.1 M glycine buffer (PH2.8). The eluted fraction was adjusted to pH 7.2 by adding 1 M Tris-HCl (pH 9.0). After replacing the antibody solution thus prepared with PBS (-) using a NAP10 column (Amersham Pharmacia Biotech, USA), a 0.22 m pore size membrane filter MILLEX-GV (Millipore, USA) ) To obtain a purified anti-klotho antibody.
  • the concentration of the purified antibody was calculated by measuring absorbance at 280 nm and setting lmg / ml to 1.350D. Since two puppies were immunized, two types of antibodies were obtained. For the purpose of distinguishing between them, these purified antibodies are hereinafter referred to as msKl antibody-1 and msKl antibody-2.
  • the mouse solubilized klotho protein prepared in Example 21 was used as an antigen.
  • the antigen per rat was emulsified with Freund's complete adjuvant and administered intradermally or subcutaneously to the rats.
  • One week after the first immunization once weekly administration was performed a total of three times, and a 50 S antigen prepared by emulsification with Freund's incomplete adjuvant was similarly administered.
  • 50 ig was administered into the tail vein for final immunization.
  • hybrid I made one.
  • the spleen was excised from the immunized rat as described above, and the spleen cells recovered from the spleen were mixed with myeloma X-63 Ag 8.6.5.3 (ATCC CRL 1580) and used as a fusion agent in polyethylene glycol solution (Japan). , Immunological Biology Laboratories) to produce hybridomas.
  • the selection of Hypridoma was performed using TIL Med ial containing HAT containing 10% fetal calf serum (Fetal Calf Serum, FCS) and hypoxanthine (H), aminopterin (A), and thymidine (T). (Immune Biological Laboratory, Japan). Cloning was performed by the limiting dilution method to obtain a cloned hybridoma derived from a single cell.
  • hybridomas producing an antibody that specifically recognizes the klotho protein were selected. 1 ⁇ 10 5 cells of the klotho-expressing HEK293 cells described in Example 31 described later were added to each well of a 96-well ELISA microplate (Grainer Inc., USA) and allowed to bind to the plate. Next, the medium was removed, and each well was washed with PBS (-) containing 2% FCS, and then the supernatant of the hybridoma was added, followed by incubation at 4 ° C for 30 minutes, followed by washing.
  • Peroxidase-labeled anti-rat IgG goat IgG Fab '(Southern Biotech, USA) diluted 2000-fold was added and incubated at 4 ° C for 30 minutes. After washing, a substrate buffer containing tetramethylbenzidine (DAK0, Denmark) was added to each well and incubated at room temperature for 8 minutes. Then, 1 M sulfuric acid was added to each well to stop the reaction. The absorbance at a wavelength of 450 nm was measured with a microplate reader (Emax, Molecular Devices, USA).
  • hybridomas showing a clear increase in absorbance were selected, and six clones (3GK 4A1, 9A1, 12BK 67G4 and 70E2) were obtained as hybridomas producing antibodies recognizing the klotho protein.
  • the subclass of the antibody specifically recognizing the klotho protein thus obtained was identified using Rat MonoAB ID / SP KIT (ZYMED LABORATORIES, USA). The results are shown in Table 2.
  • Table 2 Table 2
  • ski 9A1 FERM BP- 08679
  • Anti-klotho antibody-producing hybridoma was prepared by adding 10 g / ml of retainnulin (Sigma, USA), 5.51 g / ml of human transferrin (Sigma, USA), 0. OlmM ethanolamine (Sigma, USA), 5 ng / ml ERDF medium (Farto Seiyaku, Japan) containing 1% sodium selenite (Sigma, USA) and 1% low IgGFCS (Hyclone, USA).
  • the hybridoma is cultured to prepare the antibody, and the culture solution is passed through a filter with a pore size of 0 (Pall Gelman Laboratory, USA) to remove foreign substances such as the hybridoma, and the culture containing the antibody is removed. Qing was recovered.
  • the culture supernatant containing the anti-klotho antibody was applied to a protein G Sepharose4FF column (US,
  • the antibody solution thus prepared is applied to a gel filtration column NAP25 (US,
  • Each of the purified monoclonal antibodies thus obtained will be indicated using the name of the produced hybridoma.
  • an antibody produced by hybridoma 4A1 will be referred to as a 4A1 antibody.
  • Example 24 Measurement of Neutralizing Activity of Anti-klotho Polyclonal Antibody Using Purified Solubilized klotho Protein as Antigen in EGR 1 Promoter Lucifera Zeatosis System Anti-klotho Polyclonal Prepared in Example 22 The inhibitory effect of the antibody on the FGF23 stimulation in the EGR1 promoter-Lucifera azetus system was examined.
  • the method was performed according to Example 9, and an anti-klotho polyclonal antibody was added to the cell culture solution before stimulation with FGF23.
  • FGF23 stimulation was performed at a final concentration of 10 ng / ml.
  • FIG. 17 all of the anti-klotho polyclonal antibodies suppressed the stimulation by FGF23 in a concentration-dependent manner. This indicated that the anti-klotho antibody prepared in Example 22 suppressed FGF23-induced stimulation of cells.
  • Example 25 Effect of administration of anti-clotho polyclonal antibody to mice using purified solubilized kliotho protein as an antigen
  • mice The effect of administering the anti-klotho polyclonal antibody prepared in Example 22 to mice was examined.
  • BALB / c male, ⁇ week-old normal mouse Anti-mouse klotho protein obtained in Example 22 ⁇ Heron polyclonal antibody mskl antibody-1, 2
  • a single dose of 0.2 mL was administered.
  • 0.2 mL of the persimmon serum before klotho immunization purified under the same conditions as in Example 22 was injected into the tail vein in an amount of 0.2 mL.
  • the group treated with anti-klotho polyclonal antibody and the control group each consisted of 5 mice, and had a solid diet containing tap water and 1.03% inorganic phosphate and 1.18% calcium CE2 (Japan, (Clear Japan, Inc.)
  • Example 26 Measurement of neutralizing activity of anti-klotho monoclonal antibody using purified purified solubilized klotho protein as antigen as an antigen in EGR 1 promoter / luciferase Atsushi system
  • Example 2 of anti-klotho monoclonal antibody prepared in Example 23 The inhibitory effect of the EGR1 promoter-luciferase atsey system on FGF23 stimulation was examined. The method was the same as in Example 24. FGF23 stimulation was performed at a final concentration of 10 ng / ml.
  • Anti-klotho monoclonal antibodies were skl4Al, skl9Al, and ski12B1 antibodies, and IgGl and IgG2b subtype control antibodies were used as controls.
  • the concentration was shaken in tenths from lOO ⁇ g / ml to 0.1 g / ml.
  • the skl9Al anti-klotho monoclonal antibody suppressed the stimulation by FGF23 in a dose-dependent manner. No inhibition was confirmed with the skl4Al antibody or the skl12Bl antibody. This indicated that the anti-klotho antibody skl'9Al prepared in Example 23 suppressed the stimulation of cells by FGF23.
  • Example 27 Effect of mouse administration of anti-klotho monoclonal antibody using purified solubilized klotho protein as antigen
  • Example 27 BALB / c, male, 6-week-old normal mouse, 6 kinds of anti-mouse-solubilized klotho rat monoclonal antibodies obtained in Example 27 (3G1, 4A1, 9A1, 12B1, 67G4, and 70E2 antibodies) Each animal was administered once from the tail vein at a dose of 100 g / 0.2 mL per animal. To the control group, a rat IgG1 antibody that did not recognize klotho was administered as a control antibody by 0.2 mL in a tail vein.
  • Anti-mouse-solubilized klotho rat Monoclonal antibody-administered group and control group each consisted of 5 mice, each consisting of tap water and a solid diet containing 1.03% inorganic phosphate and 1.18% calcium CE2 (Clea Japan, Japan).
  • Fig. 20 after a single dose of the anti-mouse solubilized klotho rat monoclonal antibody, 9 hours after treatment with 4 types of 3G1, 9A1, 67G4, and 70E2 antibodies compared to the control antibody-administered group Significantly increased in serum dihydroxyvitamin D (t test * 0.05, 0.01) and serum phosphate 24 hours later (t test * 0.05, ** p ⁇ 0.01) .
  • Example 28 Effect of simultaneous addition of purified solubilized klotho protein and purified FGF23 to human and mouse cultured cells to stimulate cells
  • Example 21 In order to confirm the activity of the purified solubilized klotho obtained in Example 21, the effect of stimulating cells with the solubilized klotho and FGF23 was examined using HEK293 cells in the same manner as in Example 8.
  • the solubilized klotho used for the stimulation was stimulated at a concentration of 4 g / m FGF23 at a concentration of 100 ng / ml.
  • FIG. 21 only when stimulation with solubilized klotho and FGF23 was performed, the phosphorylation of ERK in cells was enhanced.
  • mouse osteoblast-derived cells were solubilized in MC3T3-E1 cells.
  • Klotho was stimulated at a concentration of 4 g./mK FGF23 at a concentration of 100 ng / ml, and cultured for 48 hours.
  • the cell morphology of the cells stimulated with solubilized klotho and FGF23 changed to fibroblast-like cells as in the case of bFGF stimulation.
  • mice BALB / c, male, 7-week-old normal mice were administered a single dose of the mouse-solubilized klotho protein obtained in Example 21 via the tail vein at 10 g / 0.2 mL.
  • 0.2 mL of PBS was administered into the tail vein in a dose of 0.2 mL.
  • blood was collected from the orbit using a glass capillary, and the serum was separated using Microtina (Becton Dickson, USA).
  • mice The measurement was performed using a 1,25 (0H) 2D RIA kit "TFB" (Tifby, Japan).
  • Mouse-solubilized klotho protein-treated and control groups consisted of 5 mice each, and consisted of a solid diet containing tap water and 1.03% inorganic phosphate and 1.18% calcium CE2 (CLEA Japan, Japan) ).
  • an anti-FGF23 full-length polyclonal antibody was obtained in the same manner as in Example 22 using the purified human FGF23 obtained in Example 15.
  • mouse klotho full-length stable expression HEK293 cells were obtained in the same manner as in Example 5, using the mklot o / IRES-EGFP-pEAK8 vector obtained in Example 3.
  • HEK293 cells were detached from the culture vessel using a cell detachment buffer (0.52 mM EDTA, PBS), and suspended in a suspension buffer (2% calf serum, lmM EDTA, 0.05 NaN 3 ).
  • the human FGF23 obtained in Example 15 was added at a concentration of l ⁇ g / ml and left on ice for 30 minutes.
  • the anti-FGF23 polyclonal antibody obtained in Example 30 was added at a concentration of 20/2 g / ml, and the mixture was allowed to stand on ice for 30 minutes.
  • administration of klotho protein reduces or increases the blood 1,5 dihydroxyvitamin D concentration and serum phosphorus concentration.
  • administration of anti-kolotho protein antibody increases blood 1,25-dihydroxyvitamin D concentration and serum phosphorus concentration.
  • the administration of klotho protein enhances or inhibits the action of FGF23 in the body, and the administration of the antibody klotho protein antibody inhibits the action of FGF23 in the body.

Abstract

It is intended to provide medicinal compositions usable in treating or preventing diseases in which vitamin D participates, that comprise a klotho protein capable of lowering or elevating blood phosphorus and/or activated vitamin D levels and an anti-klotho protein antibody capable of elevating blood phosphorus and/or activated vitamin D levels. A medicinal composition for lowering or elevating phosphorus concentration in blood and/or activated vitamin D concentration in blood which contains a klotho protein as the active ingredient; and a medicinal composition for elevating phosphorus concentration in blood and/or activated vitamin D concentration in blood which contains an anti-klotho protein antibody as the active ingredient.

Description

明 細 書 klo thoタンパク質および抗 kl o thoタンパク質抗体ならびにそれらの用途  Description klo tho protein and anti-klo tho protein antibodies and their uses
技術分野 Technical field
本発明はリン酸代謝、 ビタミン D代謝、 FGF23作用を変化させるような Kl o tho タンパク質、同 Klothoタンパク質を認識する抗体、同 Kl o thoタンパク質と FGF23 ポリペプチドとの複合体、 ならびにこれら物質の用途に関する。  The present invention relates to a Klotho protein that alters phosphate metabolism, vitamin D metabolism, and FGF23 action, an antibody that recognizes the Klotho protein, a complex of the Klotho protein and an FGF23 polypeptide, and uses of these substances. About.
背景技術 Background art
生体におけるリンは骨、 細胞内、 細胞外液中に存在し、 骨の力学的強さの元や 生体のエネルギー代謝、 細胞機能維持等に不可欠な重要な役割を担っている。 こ のような重要性から血清中のリン濃度はほぼ一定の濃度に保たれており、 血中リ ン酸濃度の恒常性を維持する為の制御機構の存在が示唆されている。 生体内のリ ンの出納は腎での排泄と腸管での吸収によって調節されているが、 リン出納調節 臓器としては腎臓が重要であると考えられている。 具体的には腎臓の近位尿細管 におけるリン酸輸送体タンパク質の調節が血中リン濃度調節に影響を与える。 血 清中のリン酸濃度の変化と調節に影響を与える物質としては副甲状腺ホルモン、 1 a , 25 -ジヒドロキシビタミン D3、 FGF 3などの関与が現在のところ考えられてい るが、 副甲状腺ホルモンや 1ひ, 25-ジヒドロキシビタミン D3は、 リン代謝よりも むしろカルシウム代謝調節において重要な役割を果たしている。  Phosphorus in living organisms is present in bone, intracellular and extracellular fluids, and plays an essential role in the source of mechanical strength of bone, energy metabolism in living organisms, and maintenance of cell functions. Because of this importance, the serum phosphorus concentration is maintained at a nearly constant level, suggesting the existence of a control mechanism for maintaining the blood phosphate homeostasis. The balance of phosphorus in the body is regulated by excretion in the kidney and absorption in the intestinal tract, but the kidney is considered to be important as a phosphorus balance organ. Specifically, the regulation of phosphate transporter proteins in the proximal tubule of the kidney affects the regulation of blood phosphorus levels. At present, the involvement of parathyroid hormone, 1a, 25-dihydroxyvitamin D3 and FGF3 as substances affecting the change and regulation of serum phosphate concentration is considered. 1,25-dihydroxyvitamin D3 plays an important role in regulating calcium metabolism rather than phosphorus metabolism.
FGF23は血清リン、血清 1 α, 25-ジヒドロキシビタミン Dを強力に低下させる因 子として発見された物質である(W0 02/14504、および Shimada T , e t al : Proc Na t l FGF23 is a substance found to be a potent factor in lowering serum phosphorus and serum 1α, 25-dihydroxyvitamin D (W02 / 14504, and Shimada T, etal: Proc Natl
Acad Sc i USA, 2001 May 22 : 98 (1 1): 6500-p6505) 0 血清リンの低下を伴う腫瘍性 骨軟化症の腫瘍特異的に高発現している遺伝子を探索した結果発見された物質で、 腎臓近位尿細管のリン酸輸送体を減少させることにより血清リン酸の低下を引き 起こす。 また、 腎臓での 1 α, 25-ジヒドロキシビタミン D3 (以下活性型ビタミンAcad Sc i USA, 2001 May 22: 98 (11): 6500-p6505) 0 A substance discovered as a result of searching for a tumor-specific gene highly expressed in tumoral osteomalacia accompanied by a decrease in serum phosphorus It causes a decrease in serum phosphate by reducing phosphate transporters in the renal proximal tubule. Also, 1α, 25-dihydroxyvitamin D3 (hereinafter called active vitamin)
D3) の合成酵素である 1 水酸化酵素の発現を抑制し、 逆に活性型ビタミン D3の 分解酵素である 2 4水酸化酵素の発現を増大させることにより、 血中の活性型ビ 夕ミン D3濃度を低下させる。 このように、 FGF23は血清リン、 血清 1 ひ, 25-ジヒ ドロキシビタミン Dを共に低下させる作用を持つことは明らかとなっているが、 そのシグナルを伝達する物質や経路に関しては明らかにはなっていない。 Suppresses the expression of 1-hydroxylase, which is a synthase of D3), and conversely increases the expression of 24 hydroxylase, which is an enzyme that decomposes active vitamin D3, to increase the activity of activated vitamin D3 in blood. Decrease concentration. Thus, FGF23 is serum phosphorous, serum It has been shown to have a role in lowering droxyvitamin D, but the substances and pathways that transmit that signal have not been clarified.
一方、 ヒトの老化症状に似た変異表現型をもつ klothoマウスは klotho遺伝子 座へのほかの遺伝子の挿入変異により様々な表現型を示すことが明らかになって おり、 その表現型は短寿命、 成長遅延、 老年性皮膚萎縮、 肺気腫、 難聴、 心臓機 能障害、 メンケベルク型動脈硬化症、 低回転性の骨密度の減少、 異所性石灰化、 不妊症、血糖制御異常、胸腺萎縮、 B細胞分化障害などである(W098/29544, Kuro-o M , et al : Nature 1997 Vol 390 p45- 51)。 また、 これらの表現型はウィルスべ クタ一を用いた全長 klotho遺伝子の klothoマウスへの導入により回復すること が示されている (W098/29544, Kuro-o M , et al : Nature 1997 Vol 390 p45-51)0 さらに、 klothoタンパク質の細胞外領域は KL1と KL2の二つの領域から構成され ているが、 KL1領域と免疫グロブリンを融合させたタンパク質を klothoマウスへ 投与することにより、 胸腺、 脾臓、 精巣の萎縮や異所性石灰化を抑制することが 明らかとなっている (WO00/27885)。 上記に挙げた klothoマウスの様々な症状の 他に、 klotho変異マウスは血清中のリン酸濃度の亢進、 活性型ビタミン Dの亢進 が引き起こされていることが報告されており(YoshidaT, et al -.Endocrinology. 2002 Feb; 143 (2) :683-9. ), また、 リン、 ビタミン])ホメォス夕シスが異常をきた すような状態である腎不全時での klotho タンパクの発現は正常時の腎臓と比較 して減少していることも報告されている (Koh H, et al : Bioc em Biophys Res Com讓. 2001 Feb 2;280 (4) :1015-20. )0 klotho 変異マウスでは 1 α水酸化酵素 の発現亢進が引き起こされていることが明らかとなっており、 klotho遺伝子異常 によるビタミン D代謝異常が考えられているが、 klothoタンパク質自身の機能が 未解明であることもあり、 klotho遺伝子異常におけるリン酸代謝、 ビタミン D代 謝に関わる分子メカニズムは明らかとなっていない。 On the other hand, klotho mice with a mutant phenotype similar to that of human aging have been shown to exhibit various phenotypes due to insertion mutations of other genes at the klotho locus. Growth retardation, senile skin atrophy, emphysema, deafness, cardiac dysfunction, Monkeberg-type atherosclerosis, low rotational bone loss, ectopic calcification, infertility, abnormal glycemic control, thymic atrophy, B cells Differentiation disorders and the like (W098 / 29544, Kuro-o M, et al: Nature 1997 Vol 390 p45-51). In addition, it has been shown that these phenotypes can be restored by introducing the full-length klotho gene into klotho mice using a virus vector (W098 / 29544, Kuro-o M, et al: Nature 1997 Vol 390 p45). -51) 0 Furthermore, the extracellular region of the klotho protein is composed of two regions, KL1 and KL2.By administering a protein in which the KL1 region and immunoglobulin are fused to a klotho mouse, the thymus, spleen, It has been shown to suppress testicular atrophy and ectopic calcification (WO00 / 27885). In addition to the various symptoms of klotho mice listed above, it has been reported that klotho mutant mice have increased serum phosphate levels and increased active vitamin D (YoshidaT, et al- .Endocrinology. 2002 Feb; 143 (2): 683-9.), Phosphorus, vitamins)) The expression of klotho protein during renal insufficiency, which is a condition that causes abnormal homeosis, is normal. It has also been reported that it is reduced as compared to the kidney (Koh H, et al: Biochem Biophys Res Comb. 2001 Feb 2; 280 (4): 1015-20.) 1α in 0 klotho mutant mice It has been clarified that increased expression of hydroxylase is caused, and it is considered that abnormalities of vitamin D metabolism due to abnormal klotho gene are considered.However, the function of klotho protein itself may not be elucidated. Molecular mechanisms related to phosphate metabolism and vitamin D metabolism in abnormalities Parkinsonism is not clear.
特許文献 1  Patent Document 1
W002/14504  W002 / 14504
特許文献 2  Patent Document 2
W098/29544  W098 / 29544
特許文献 3  Patent Document 3
WO00/27885 非特許文献 1 WO00 / 27885 Non-patent document 1
S imada T , et al : Proc Natl Acad Sci USA, 2001 May 22 ;98 (11): 6500-ρ65Ό5  Simada T, et al: Proc Natl Acad Sci USA, 2001 May 22; 98 (11): 6500-ρ65Ό5
非特許文献 2  Non-patent document 2
Kuro-o ·Μ , et al : Nature 1997 Vol 390 p45-51  Kuro-o · Μ, et al: Nature 1997 Vol 390 p45-51
非特許文献 3  Non-patent document 3
Endocrinology. 2002 Feb; 143 (2) :683-9  Endocrinology. 2002 Feb; 143 (2): 683-9
非特許文献 4  Non-patent document 4
Koh H , et al : Biochem Biophys Res Com匪. 2001 Feb 2;280 (4) : 1015-20  Koh H, et al: Biochem Biophys Res Com Marauder. 2001 Feb 2; 280 (4): 1015-20
発明の開示 Disclosure of the invention
本発明は、 血中リンおよび Zまたは血中活性型ビタミン Dを低下または上昇さ せる klothoタンパク質、血中リンおよび Zまたは血中活性型ビタミン Dを上昇さ せる抗 klothoタンパク質抗体ならびに FGF23の作用を調節し得る klothoタンパ ク質または抗 klothoタンパク質抗体を含む医薬組成物の提供を目的とし、 リン、 ビタミン])が関与する腫瘍性骨軟化症、 ADHR、 XLH、 腎性骨異栄養症、 透析骨症、 骨粗鬆症、 低リン血症、 クル病、 骨軟化症、 尿細管機能障害、 骨減少症、 低カル シゥム血症、 高リン血症、 高 1, 25D血症、 副甲状腺機能亢進症、 異 性石灰化、 搔痒、 骨硬化症、パジエツト病、高カルシウム血症、副甲状腺機能低下症、 骨痛、 筋力低下、 骨格変形、 成長障害および低 1, 25D血症などの疾患の治療または予防 のための医薬組成物の提供を目的とする。  The present invention relates to a klotho protein that decreases or increases blood phosphorus and Z or blood active vitamin D, an anti-klotho protein antibody that increases blood phosphorus and Z or blood active vitamin D, and the action of FGF23. For the purpose of providing a pharmaceutical composition containing a klotho protein or an anti-klotho protein antibody that can be regulated, phosphorus, vitamins) -related neoplastic osteomalacia, ADHR, XLH, renal osteodystrophy, dialysis bone Disease, osteoporosis, hypophosphatemia, rickets, osteomalacia, renal tubular dysfunction, osteopenia, hypocalcemia, hyperphosphatemia, hyper1,25D blood disease, hyperparathyroidism, abnormal Treatment or prevention of diseases such as calcified calcification, pruritus, bone sclerosis, Paget's disease, hypercalcemia, hypoparathyroidism, bone pain, muscle weakness, skeletal deformity, growth disorders and hypo1,25Demia Pharmaceutical composition for And an object thereof is to provide a.
本発明はこのような状況に鑑みてなされたものであり、 その目的は血中リン濃 度、 血中活性型ビタミン D濃度、 FGF23作用をコントロールしうる物質とその用 途を提供することにある。  The present invention has been made in view of such circumstances, and an object thereof is to provide a substance capable of controlling blood phosphorus concentration, blood active vitamin D concentration, and FGF23 action, and a use thereof. .
本発明者らは全長 klothoタンパク質、 可溶化 klothoタンパク質に関わらず、 klothoタンパク質が存在する条件下において FGF23による細胞内へのシグナル伝 達 (ERKリン酸化亢進や egrl発現上昇) が動物での腎臓と同様に引き起こされる ことを見出した。 また、 FGF23と klothoタンパク質とは結合することを明らかと した。 また、 FGF23と klothoタンパクとの相互作用がリン、 ビタミン Dの代謝調 節に重要であることを見出した。 また、 klothoタンパク質を認識する抗体のマウ スへの投与により血中のリン、 活性型ビタミン Dの濃度を上昇させることに成功 し、 また可溶化 klotho夕ンパク質発現細胞のヌードマウスへの移植により、血中 のリン濃度、 活性型ビタミン D濃度を低下させることにも成功した。 さらに、 精 製した可溶化 klothoタンパク質をマウスへ投与することにより血中のリン濃度、 活性型ビタミン濃度を上昇させることにも成功した。 さらに、 klothoタンパク質 または抗 klotho夕ンパク質抗体が FGF23の作用を調節し得ることも見出した。こ れらの結果から本発明を完結するに至り、 本発明により血中リン、 血中ビタミン D を制御することにより関係する疾患を治療することのできる薬剤となりうる、 もしくは関連する疾患の診断を可能にしうるものと考える。 従って、 本発明は以 下の通りである。 The present inventors found that FGF23 transduced signals into cells (enhanced ERK phosphorylation and increased egrl expression) in the presence of klotho protein, regardless of the full-length klotho protein or solubilized klotho protein, in the kidney of animals. Was found to be caused as well. In addition, it was revealed that FGF23 binds to klotho protein. In addition, the interaction between FGF23 and klotho protein regulates the metabolism of phosphorus and vitamin D. I found it important for the verse. In addition, administration of an antibody that recognizes klotho protein to the mouse successfully increased the concentration of phosphorus and active vitamin D in the blood, and transplanted solubilized klotho protein-expressing cells into nude mice. It also succeeded in lowering blood phosphorus levels and active vitamin D levels. Furthermore, by administering purified solubilized klotho protein to mice, we succeeded in increasing blood phosphorus concentration and active vitamin concentration. Furthermore, it was found that klotho protein or anti-klotho protein antibody could regulate the action of FGF23. From these results, the present invention has been completed, and the present invention can be used as a drug capable of treating related diseases by controlling blood phosphorus and blood vitamin D, or diagnoses of related diseases. I think it could be possible. Therefore, the present invention is as follows.
[ 1 ] klothoタンパク質を有効成分として含む、血中リン濃度およびノまたは血 中活性型ビタミン D濃度を低下させるための医薬組成物、  [1] a pharmaceutical composition for lowering blood phosphorus concentration and blood or active vitamin D concentration, comprising a klotho protein as an active ingredient;
[ 2 ] klothoタンパク質を有効成分として含む、血中リン濃度およびノまたは血 中活性型ビタミン D濃度を上昇させるための医薬組成物、  [2] A pharmaceutical composition for increasing blood phosphorus concentration and blood or active vitamin D concentration, comprising a klotho protein as an active ingredient,
[ 3 ] klothoタンパク質を有効成分として含む、 FGF23の作用を増強するための 医薬組成物、  [3] a pharmaceutical composition for enhancing the action of FGF23, comprising a klotho protein as an active ingredient;
[ 4 ] klothoタンパク質を有効成分として含む、 FGF23の作用を阻害するための 医薬組成物、  [4] a pharmaceutical composition for inhibiting the action of FGF23, comprising a klotho protein as an active ingredient;
[ 5 ] klothoタンパク質が可溶化 klothoタンパク質である、 [ 1 ]から [ 4 ]のい ずれかの医薬組成物、  [5] The pharmaceutical composition according to any one of [1] to [4], wherein the klotho protein is a solubilized klotho protein.
[ 6 ] 抗 klothoタンパク質抗体を有効成分として含む、血中リン濃度およびノま たは血中活性型ピ夕ミン D濃度を上昇させるための医薬組成物、  [6] A pharmaceutical composition for increasing blood phosphorus concentration and blood or active piminin D concentration, which comprises an anti-klotho protein antibody as an active ingredient,
[ 7 ] 抗 klothoタンパク質抗体を有効成分として含む、 FGF23の作用を阻害する ための医薬組成物、 [7] a pharmaceutical composition for inhibiting the action of FGF23, which comprises an anti-klotho protein antibody as an active ingredient;
[ 8 ] 腫瘍性骨軟化症、 ADHR、 XLH、 腎性骨異栄養症、 透析骨症、 骨粗鬆症、 低リ ン血症、 クル病、 骨軟化症、 尿細管機能障害、 骨減少症、 低カルシウム血症、 高 リン血症、 高 1, 25D血症、副甲状腺機能亢進症、 異所性石灰化、搔痒、 骨硬化症、 パジェット病、 高カルシウム血症、 副甲状腺機能低下症、 骨痛、 筋力低下、 骨格 変形、 成長障害および低 1, 25D血症からなる群から選択されるリンおよび Zまた はビタミン D が関与する疾患の治療および/または予防に使用される [ 1 ]から [ 7 ]のいずれかの医薬組成物、 および [8] Neoplastic osteomalacia, ADHR, XLH, renal osteodystrophy, dialysis osteopathy, osteoporosis, hypophosphatemia, rickets, osteomalacia, tubular dysfunction, osteopenia, low calcium , Hyperphosphatemia, hyper1,25D, hyperparathyroidism, ectopic calcification, pruritus, bone sclerosis, Paget's disease, hypercalcemia, hypoparathyroidism, bone pain, Phosphorus and Z selected from the group consisting of muscle weakness, skeletal deformity, growth disorders and hypo1,25D Is a pharmaceutical composition according to any one of [1] to [7], which is used for treating and / or preventing a disease associated with vitamin D, and
[ 9 ] . 血中リン濃度お'よび または血中活性型ビタミン D濃度を変動させる物質 をスクリーニングする方法であって、 FGF23 と klotho タンパク質を接触させ、 FGF23と klothoタンパク質との相互作用による細胞内シグナルを検出することに より、 スクリーニングする方法。  [9]. A method for screening a substance that alters the blood phosphorus level and the active vitamin D level by contacting FGF23 with the klotho protein, and intracellularly interacting with the FGF23 and the klotho protein. Screening method by detecting signal.
以下、 本発明を詳細に説明する。  Hereinafter, the present invention will be described in detail.
本発明は血中のリン濃度、 活性型ビタミン ])濃度、 FGF23作用を変化させるよ うな klothoタンパク質、 klothoタンパク質を認識する抗体、 FGF23と klotho夕 ンパク質との複合体とそれら物質の用途に関するものである。  The present invention relates to klotho protein, an antibody that recognizes klotho protein, an antibody that recognizes klotho protein, a complex of FGF23 and klotho protein, and a use of those substances that alters the concentration of phosphorous in blood, active vitamin]) concentration, FGF23 action It is.
本発明を用いることにより、 klothoタンパク質、 klothoタンパク質を認識する 抗体、 FGF23と klothoタンパク質との複合体の少なくともいずれかを含む医薬組 成物を、 リン、 ビタミン Dが関与する病態、例えば、腫瘍性骨軟化症、 ADH , XLH、 腎性骨異栄養症、 透析骨症、 骨粗鬆症、 低リン血症、 クル病、 骨軟化症、 尿細管 機能障害、 骨減 症、 低カルシウム血症、 高リン血症、 高 1, 25D血症、 副甲状腺 機能亢進症、異所性石灰化、搔痒、 骨硬化症、パジエツト病、高カルシウム血症、 副甲状腺機能低下症、 骨痛、 筋力低下、 骨格変形、 成長障害および低 1, 25D血症 などの疾患の治療または予防に用いることができる。 可溶化 klotho  By using the present invention, a pharmaceutical composition comprising at least one of a klotho protein, an antibody recognizing the klotho protein, and a complex of FGF23 and the klotho protein can be used to convert a pharmaceutical composition containing phosphorus and vitamin D into, for example, a tumorous condition. Osteomalacia, ADH, XLH, renal osteodystrophy, dialysis osteopathy, osteoporosis, hypophosphatemia, rickets, osteomalacia, tubular dysfunction, osteopenia, hypocalcemia, hyperphosphatemia Disease, hyper1, 25D, hyperparathyroidism, ectopic calcification, pruritus, osteosclerosis, Pajet's disease, hypercalcemia, hypoparathyroidism, bone pain, muscle weakness, skeletal deformity, It can be used to treat or prevent diseases such as growth disorders and hypo1,25Demia. Solubilized klotho
可溶化 klotho cDNA としては配列番号 1 (Genebank access ion No. AB005142) に記載のヒト由来の klotho cDNAの配列を元にして公知の方法により調製するこ とができる。 例えば、 klothoタンパク質を発現している細胞、 臓器より cDNAラ イブラリーを作製し、 klotho cDNA の配列 (配列番号 1 ) の一部をプローブにし てハイブリダィゼーシヨンを行なうことにより調製できる。 また、 klothoタンパ ク質を発現している細胞、臓器より RNAを調製し、逆転写酵素により cDNAを作製 した後 kl otho cDNAの配列 (配列番号 1 ) に基づいてオリゴ DNAを合成し、 これ をプライマーとして用いて PCR反応を行い、 klotho cDNAを増幅させ調製するこ とも可能である。  The solubilized klotho cDNA can be prepared by a known method based on the sequence of human-derived klotho cDNA described in SEQ ID NO: 1 (Genebank accession No. AB005142). For example, it can be prepared by preparing a cDNA library from cells or organs expressing the klotho protein and performing hybridization using a part of the klotho cDNA sequence (SEQ ID NO: 1) as a probe. In addition, RNA was prepared from cells and organs expressing the klotho protein, cDNA was prepared using reverse transcriptase, and oligo DNA was synthesized based on the klotho cDNA sequence (SEQ ID NO: 1). It is also possible to amplify and prepare klotho cDNA by performing a PCR reaction using it as a primer.
上記通り得られた klotho cDNAについて塩基配列の決定を行う。 塩基配列の決 定はマキサム-ギルバートの化学修飾法、又は M13ファージを用いるジデォキシヌ クレオチド鎖終結法等の公知手法により行うことができるが、 通常は自動塩基配 列決定機 (例えば PERKIN- ELMER社製 373A DNAシークェンサ一等) を用いて配列 決定が行われる。 The nucleotide sequence of the klotho cDNA obtained as described above is determined. Determination of nucleotide sequence The determination can be performed by a known method such as the Maxam-Gilbert chemical modification method or the dideoxynucleotide chain termination method using M13 phage. Usually, an automatic base sequencer (for example, 373A DNA Sequencer manufactured by PERKIN-ELMER) is used. , Etc.).
配列番号 1にヒト' klot o cDNA の塩基配列を、 配列番号 2にヒト全長 klotlio タンパク質のアミノ酸配列、配列番号 3に可溶化ヒト klothoタンパク質のァミノ 酸配列を例示するが、 低リン酸血症誘導活性および Zもしくは低活性型ビ夕ミン SEQ ID NO: 1 shows the nucleotide sequence of human kloto cDNA, SEQ ID NO: 2 shows the amino acid sequence of human full-length klotlio protein, and SEQ ID NO: 3 shows the amino acid sequence of solubilized human klotho protein. Active and Z or low activity bimin
D血症誘導活性、 高リン酸血症誘導活性およびノもしくは高活性型ビ夕ミン D血 症誘導活性、 または FGF23の作用を調節 (増強または阻害) する作用を有する限 り、 当該アミノ酸配列において 1個若しくは数個のアミノ酸に欠失、 置換、 付加 等の変異が生じてもよい。 また、 ヒト Klothoタンパク質とマウス Klothoタンパ ク質のアミノ酸配列レベルでの相同性は 8 5 %であり (配列番号 2がヒ卜 klotho タンパク質のアミノ酸配列を、配列番号 2 3がマウス klothoタンパク質のァミノ 酸配列を示す)、マウス klothoタンパク質はヒト klothoタンパク質と同等の機能 を有していると予測される。従って、本発明において klothoタンパク質はヒト由 来の klotho タンパク質だけではなく、 マウス由来 klotho タンパク質ゃヒト klothoタンパク質とアミノ酸の相同性が、 80%以上、好ましくは 85%以上の他動 物種の klothoタンパク質も含まれる。 これらヒト以外の動物種由来の klotho夕 ンパク質のアミノ酸配列からなるタンパク質であって、 低リン酸血症誘導活性お よび Zもしくは低活性型ビ夕ミン D血症誘導活性、 高リン酸血症誘導活性およびAs long as it has D-disease-inducing activity, hyperphosphatemia-inducing activity and no- or highly-active biminin D-disease-inducing activity, or an effect of regulating (enhancing or inhibiting) the action of FGF23, Mutations such as deletion, substitution, and addition may occur in one or several amino acids. The homology at the amino acid sequence level between the human Klotho protein and the mouse Klotho protein is 85% (SEQ ID NO: 2 is the amino acid sequence of the human klotho protein, and SEQ ID NO: 23 is the amino acid sequence of the mouse klotho protein). It is predicted that mouse klotho protein has the same function as human klotho protein. Therefore, in the present invention, klotho protein includes not only klotho protein derived from human but also klotho protein of mouse klotho protein derived from mouse / human klotho protein having amino acid homology of 80% or more, preferably 85% or more. included. A protein comprising an amino acid sequence of klotho protein derived from any of these animal species other than humans, wherein the protein has a hypophosphatemic-inducing activity and a Z- or low-activity bieminin D-inducing activity, and hyperphosphatemia. Inducing activity and
Zもしくは高活性型ビタミン!)血症誘導活性、 または FGF23の作用を調節する作 用を有する限り、当該アミノ酸配列において 1個もしくは数個のアミノ酸に欠失、 置換、 付加等の変異が生じていてもよい。 タンパク質が低リン酸血症誘導活性お よび もしくは低活性型ビ夕ミン D血症誘導活性または高リン酸血症誘導活性お よび Zもしくは高活性型ビタミン D血症誘導活性を有しているかは後述の実施例Z or highly active vitamin!) As long as it has an activity to induce bloodemia or regulate the action of FGF23, one or several amino acids in the amino acid sequence have mutations such as deletion, substitution, or addition. May be. Whether the protein has hypophosphatemia-inducing activity and / or low-activity biminmin D-induction activity or hyperphosphatemia-inducing activity and Z or hyperactive vitamin D-induction activity Examples described later
1 3や実施例 2 9に記載の方法を参考にタンパク質を発現している細胞 マウス に移植、 もしくは同タンパク質をマウス等に投与し、 血清中のリン濃度またはCells expressed in cells expressing the protein were transplanted into mice, or the protein was administered to mice, etc., according to the method described in 13 or Example 29, and the serum phosphorus concentration or
1, 25ジヒドロキシビタミン D濃度を測定すればよい。 また、 タンパク質が FGF23 の作用を調節する作用を有しているか否かは、 例えば後述の実施例 2 8に記載の 方法を参考に可溶化 klothoタンパク質と精製 FGF23を哺乳類動物細胞に投与して、 FGF23 の細胞への刺激効果が影響を受けるかどうかを測定すればよい。 また、 FGF23は、 ERKのリン酸化に関与しており、 FGF23の ERKリン酸化作用が亢進また は抑制されることを指標にして、 FGF23 の作用を調節するか否かを測定すること もできる。 The 1,25-dihydroxyvitamin D concentration may be measured. Whether or not the protein has an effect of regulating the action of FGF23 can be determined by administering a solubilized klotho protein and purified FGF23 to mammalian cells, for example, by referring to the method described in Example 28 below. What is necessary is just to determine whether the stimulating effect of FGF23 on cells is affected. Further, FGF23 is involved in the phosphorylation of ERK, and it can also be measured whether or not the effect of FGF23 is regulated by using as an index that the ERK phosphorylation of FGF23 is enhanced or suppressed.
例えば、 配列番号 2、 配列番号 3、 もしくは配列番号 2 3に示されるアミノ酸 配列から 1又は数個、 好ましくは 1〜10個、 さらに好ましくは 1〜5個のァミノ 酸が欠失してもよく、 配列番号 2、 配列番号 3もしくは配列番号 2 3に示される アミノ酸配列に 1又は数個、 好ましくは 1〜10個、 さらに好ましくは 1〜 5個の アミノ酸が付加してもよく、 あるいは、 配列番号 2、 配列番号 3もしくは配列番 号 2 3で表わされるアミノ酸配列の 1又は数個、 好ましくは 1〜10個、 さらに好 ましくは 1〜5個のアミノ酸が他のアミノ酸に置換してもよい。  For example, one or several, preferably 1 to 10, more preferably 1 to 5 amino acids may be deleted from the amino acid sequence shown in SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 23. One or several amino acids, preferably 1 to 10, more preferably 1 to 5 amino acids may be added to the amino acid sequence shown in SEQ ID NO: 2, SEQ ID NO: 3 or SEQ ID NO: 23; Even if one or several amino acids, preferably 1 to 10, and more preferably 1 to 5 amino acids of the amino acid sequence represented by SEQ ID NO: 2, SEQ ID NO: 3 or SEQ ID NO: 23 are substituted with other amino acids, Good.
また、 置換の方法としては、 アミノ酸の特性をある程度保持したファミリー内 での保存的置換を行っても良い。 アミノ酸側鎖の側鎖の特性から一般的に分類さ れるフアミリ一は以下のものが挙げられる。  As a substitution method, conservative substitution within a family that retains the properties of amino acids to some extent may be performed. Families, which are generally classified based on the characteristics of the side chains of amino acid side chains, include the following.
( 1 ) 酸性アミノ酸ファミリ一: ァスパラギン酸、 グルタミン酸  (1) Acidic amino acid family 1: aspartic acid, glutamic acid
( 2 ) 塩基性アミノ酸ファミリー: リジン、 アルギニン、 ヒスチジン  (2) Basic amino acid family: lysine, arginine, histidine
( 3 )非極性アミノ酸ファミリー:ァラニン、ノ リン、 ロイシン、イソロイシン、 プロリン、 フエ二ルァラニン、 メチォニン、 トリプ卜ファン  (3) Non-polar amino acid family: alanine, norin, leucine, isoleucine, proline, fenylalanine, methionine, triptophan
( 4 ) 非帯電極性アミノ酸ファミリ一: グリシン、 ァスパラギン、 グルタミン、 システィン、 セリン、 スレオニン、 チロシン  (4) Uncharged polar amino acid family: Glycine, Asparagine, Glutamine, Cystine, Serine, Threonine, Tyrosine
( 5 ) 脂肪族ヒドロキシアミノ酸ファミリー:セリン、 スレオニン  (5) Aliphatic hydroxyamino acid family: serine, threonine
( 6 ) アミド含有アミノ酸ファミリー:ァスパラギン、 グルタミン  (6) Amide-containing amino acid family: asparagine, glutamine
( 7 ) 脂肪族アミノ酸:ァラニン、 ノ リン、 ロイシン、 イソロイシン  (7) Aliphatic amino acids: alanine, norin, leucine, isoleucine
( 8 ) 芳香族アミノ酸ファミリー:フエ二ルァラニン、 トリブトファン、 チロシ ン  (8) Aromatic amino acid family: phenylalanine, tributofan, tyrosine
( 9 ) 疎水性アミノ酸ファミリ一: ロイシン、 イソロイシン、 ノ リン '  (9) Hydrophobic amino acid family: leucine, isoleucine, norin '
( 1 0 )小型アミノ酸ファミリー:ァラニン、セリン、 スレオニン、 メチォニン、 グリシン  (10) Small amino acid family: alanine, serine, threonine, methionine, glycine
本発明において、本発明の可溶化 klothoタンパク質のアミノ酸配列の少なくと も一部に変異を導入するには、 該アミノ酸をコードする DNAの塩基配列に変異を 導入する手法が採用される。 In the present invention, in order to introduce a mutation into at least a part of the amino acid sequence of the solubilized klotho protein of the present invention, a mutation is required in the nucleotide sequence of DNA encoding the amino acid. The method of introduction is adopted.
DNAに変異を導入するには、 Kimkel法若しくは Gapped duplex法等の公知手法 又はこれに準ずる方法により行うことができる。 例えば、 変異オリゴヌクレオチ ドをプライマーとして用いた部位特異的突然変異誘発法に基づいて変異を導入す るが、変異導入用キット (例えば Mutan-K(TAKARA社製)、 Mutan- G (TAKARA社製)、 TAKARA社の LA PCR in vitro Mutagenesis シリーズキットなど)を用いて変異を 導入することもできる。  Mutation can be introduced into DNA by a known method such as the Kimkel method or the Gapped duplex method, or a method analogous thereto. For example, a mutation is introduced based on a site-directed mutagenesis method using a mutant oligonucleotide as a primer. Mutagenesis kits (for example, Mutan-K (TAKARA), Mutan-G (TAKARA) ), TAKARA's LA PCR in vitro Mutagenesis series kit, etc.).
さらに、 上記本発明の DNA (配列番号 1) から調製されるプローブとストリン ジェン卜な条件下でハイブリダィズし、 かつ、 低リン血症誘導活性および もし くは低活性型ビタミン D血症誘導活性、 高リン酸血症誘導活性および Zもしくは 高活性型ビタミン D血症誘導活性、 または FGF23の作用を調節する作用を有する タンパク質をコードする DNAも、 本発明の DNAに含まれる。 このような DNAの配 列は、 配列番号 1に示される DNAの有する配列と相同性が 70%以上、 好ましくは 80%以上、 さらに好ましくは 90%以上、 特に好ましくは 95%以上である。 上記の ようにこれらの DM配列にはマウス klothoタンパク質等ヒト以外の動物種由来の klothoタンパク質も含まれる。 プローブとは、 配列番号 1に示す配列の全長に対 し相補配列を有するもの、あるいは長さが 17塩基以上の連続する配列(部分配列) に対し相補配列を有するものをいう。  Further, it hybridizes with the probe prepared from the DNA of the present invention (SEQ ID NO: 1) under stringent conditions, and has a hypophosphatemia-inducing activity and / or a low-activity-type vitamin Demia-inducing activity. The DNA of the present invention also includes a DNA encoding a protein having a hyperphosphatemia-inducing activity and an activity of inducing Z or highly active vitamin D bloodemia, or an effect of regulating the action of FGF23. Such a DNA sequence has a homology of 70% or more, preferably 80% or more, more preferably 90% or more, and particularly preferably 95% or more with the sequence of the DNA shown in SEQ ID NO: 1. As described above, these DM sequences also include klotho proteins derived from non-human animal species such as mouse klotho protein. The probe refers to a probe having a complementary sequence to the full length of the sequence shown in SEQ ID NO: 1 or a probe having a complementary sequence to a continuous sequence (partial sequence) having a length of 17 bases or more.
ここで、 ストリンジェン卜な条件とは、 ナトリウム濃度が 750mM以上、 好まし くは 900mM以上であり、温度が 40°C以上、好ましくは 42 の条件を満たすものを いう。 具体的には、 6XSSC、 5XDenhardt、 0.5% SDS、 50¾ Formamide、 42 の条 件をいう。 なお、 6XSSC とは、 900mM NaCl、 90mM クェン酸ナトリウムを意味す る。 デンハルト溶液 (Denhardt) とは、 BSA (ゥシ血清アルブミン)、 ポリビニル ピロリドン及び Ficoll400を含む溶液であり、 50xDenhardtは 1%BSA、 1% ポリ ビニルピロリドン、 l%Ficoll 400の組成からなる (5XDenhardtは 50xDenhardt の 10分の 1の濃度を意味する)。  Here, stringent conditions refer to those having a sodium concentration of 750 mM or more, preferably 900 mM or more, and a temperature of 40 ° C. or more, preferably 42. Specifically, it refers to the conditions of 6XSSC, 5XDenhardt, 0.5% SDS, 50¾ Formamide, 42. 6XSSC means 900 mM NaCl and 90 mM sodium citrate. Denhardt's solution (Denhardt) is a solution containing BSA (polyserum albumin), polyvinylpyrrolidone and Ficoll400. 50xDenhardt is composed of 1% BSA, 1% polyvinylpyrrolidone and l% Ficoll400. Means one tenth of the concentration).
また、 本発明の DNAがコードするタンパク質が低リン酸血症誘導活性および Z もしくは低活性型ビタミン D血症誘導活性、 高リン酸血症誘導活性および/もし くは高活性型ビタミン])血症誘導活性、 または FGF23の作用を調節する作用を有 しているかは後述の実施例 13や実施例 29に記載の方法を参考にタンパク質を 発現している細胞をマウスに移植、 もしくは同タンパク質をマウス等に投与し、 血清中のリン濃度または 1, 25ジヒドロキシビタミン D濃度を測定すればよレ^ま た、 タンパク質が FGF23の作用を調節する作用を有しているか否かは、 例えば後 述の実施例 2 8に記載の方法を参考に可溶化 kl othoタンパク質と精製 FGF23を哺 乳類動物細胞に投与して、 FGF23 の細胞への刺激効果が影響を受けるかどうかを 測定すればよい。 In addition, the protein encoded by the DNA of the present invention may have hypophosphatemia-inducing activity and Z or low-activity vitamin D-blood-inducing activity, hyperphosphatemia-inducing activity and / or a highly active vitamin]) Whether the protein has the disease-inducing activity or the effect of regulating the action of FGF23 is analyzed by referring to the methods described in Examples 13 and 29 described below. The expressed cells can be transplanted into mice, or the protein can be administered to mice, etc., and the serum phosphorus concentration or 1,25-dihydroxyvitamin D concentration measured, and the protein regulates the action of FGF23. It is determined whether or not FGF23 is administered to mammalian cells by solubilizing klotho protein and purified FGF23, for example, by referring to the method described in Example 28 below. It is sufficient to determine whether the stimulus effect is affected.
一旦本発明の DNAの塩基配列が決定されると、 その後は、 化学合成によって、 又は決定された当該塩基配列から合成したプライマ一を用いた PCRによって、 本 発明の DNAを得ることができる。 本発明の DNAを含む組換えベクター及び形質転換体の作製  Once the nucleotide sequence of the DNA of the present invention is determined, the DNA of the present invention can be obtained by chemical synthesis or by PCR using a primer synthesized from the determined nucleotide sequence. Preparation of recombinant vector and transformant containing DNA of the present invention
(1) 組換えベクターの作製 .  (1) Preparation of recombinant vector.
本発明の組換えベクターは、 適当なベクターに本発明の DNAを連結(挿入)する ことにより得ることができる。 本発明の DNAを挿入するためのベクターは、 宿主 中で複製可能なものであれば特に限定されず、 例えば、 プラスミド DNA、 ファー ジ DNA等が挙げられる。  The recombinant vector of the present invention can be obtained by ligating (inserting) the DNA of the present invention into an appropriate vector. The vector into which the DNA of the present invention is inserted is not particularly limited as long as it can be replicated in a host, and examples thereof include plasmid DNA and phage DNA.
プラスミド DNAとしては、 大腸菌由来のプラスミド (例えば pBR322, pBR325, PUC 118, pUC119等)、 枯草菌由来のプラスミド (例えば ρϋΒΙ ΙΟ, pTP5等)、 酵母 由来のプラスミド (例えば YEp l3, YEp24, YCp50 等)などが挙げられ、 ファージ DNA としては λファージ等が挙げられる。 また、 レトロウイルス、 アデノウィル ス又はワクシニアウィルスなどの動物ウィルス、 あるいはバキュロウィルスなど の昆虫ウィルスベクタ一を用いることもできる。 さらに、 GST、 His-tagなどが連 結された融合プラスミドを用いることもできる。  Plasmid DNAs include Escherichia coli-derived plasmids (eg, pBR322, pBR325, PUC118, pUC119, etc.), Bacillus subtilis-derived plasmids (eg, ρϋΒΙ, pTP5, etc.), and yeast-derived plasmids (eg, YEpl3, YEp24, YCp50) Phage DNA includes λ phage. Also, animal viruses such as retrovirus, adenovirus or vaccinia virus, or insect virus vectors such as baculovirus can be used. Furthermore, a fusion plasmid in which GST, His-tag, etc. are linked can also be used.
ベクターに本発明の DNAを挿入するには、 まず、 精製された DMを適当な制限 酵素で切断し、適当なベクター DNAの制限酵素部位又はマルチクローニングサイ 卜に挿入してベクターに連結する方法などが採用される。  In order to insert the DNA of the present invention into a vector, first, purified DM is cut with an appropriate restriction enzyme, inserted into an appropriate vector DNA restriction enzyme site or a multicloning site, and ligated to the vector. Is adopted.
本発明の DNAは、 その DNAの機能が発揮されるようにベクターに組み込まれる ことが必要である。そこで、本発明のベクターには、 プロモーター、 本発明の DNA のほか、 所望によりェンハンサーなどのシスエレメント、 スプライシングシグナ ル、 ポリ A付加シグナル、 選択マーカー、 リボソーム結合配列 (SD配列) などを 連結することができる。 なお、 選択マーカーとしては、 例えばジヒドロ葉酸還元 酵素遺伝子、アンピシリン耐性遺伝子、ネオマイシン耐性遺伝子等が挙げられる。The DNA of the present invention needs to be incorporated into a vector so that the function of the DNA is exhibited. Therefore, the vector of the present invention contains, in addition to the promoter and the DNA of the present invention, cis elements such as enhancers, splicing signals, poly-A addition signals, selection markers, and ribosome binding sequences (SD sequences), if desired. Can be linked. Examples of the selection marker include a dihydrofolate reductase gene, an ampicillin resistance gene, a neomycin resistance gene, and the like.
(2) 形質転換体の作製 (2) Preparation of transformant
本発明の形質転換体は、 本発明の組換えベクターを、 目的遺伝子が発現し得る ように宿主中に導入することにより得ることができる。 ここで、 宿主としては、 本発明の DNAを発現できるものであれば特に限定されるものではない。 例えば、 エッシェリヒァ 'コリ {Escherichia coli) 等のエツシェリヒァ属、 バチルス ·ズ プチリス iBacillus subtil is)等のバチルス属、 シユードモナス · プチダ [Pseudomonas 〃 )等のシユードモナス属に属する細菌、 あるいはサッカロミ セス ·セレじシェ [Saccharomyces cerevisiae)、 シ 11ノサッカロミセス · ンべ (Schizosaccharomyces pombe)等の 母が尊^られる。 また、 COS細胞、 CH0細胞、 HEK293細胞等の動物細胞や、 Sf 9、 Si2 1等の昆虫細胞を用いることもできる。 大腸菌等の細菌を宿主とする場合は、 本発明の組換えベクターが該細菌中で自 律複製可能であると同時に、 プロモーター、 リボゾーム結合配列、本発明の DNA、 転写終結配列により構成されていることが好ましい。 また、 プロモーターを制御 する遺伝子が含まれていてもよい。 大腸菌としては、 例えばエッシェリヒァ -コ リ Escherichia coli) JM109, HB 101 などが挙げられ、 枯草菌としては、 例えば バチルス ·ズプチ O TABacillus ^Μ/ ^)などが挙げられる。 プロモーターは、 大腸菌等の宿主中で発現できるものであればいずれを用いてもよい。 例えば t rp プロモーター、 l acプロモー夕一、 PLプロモーター、 PRプロモーターなどの、 大 腸菌ゃファージに由来する T7プロモーターなどが用いられる。 t acプロモーター などのように、 人為的に設計改変されたプロモーターを用いてもよい。 細菌への 組換えベクターの導入方法は、 細菌に DNAを導入する方法であれば特に限定され るものではない。 例えばカルシウムイオンを用いる方法、 エレクト口ポレーショ ン法等が挙げられる。 The transformant of the present invention can be obtained by introducing the recombinant vector of the present invention into a host so that the target gene can be expressed. Here, the host is not particularly limited as long as it can express the DNA of the present invention. For example, bacteria belonging to the genus Escherichia such as Escherichia coli, Bacillus such as Bacillus subtilis iBacillus subtil is), bacteria belonging to the genus Pseudomonas such as Pseudomonas [, or Saccharomyces cerevisiae [ Saccharomyces cerevisiae), mother of the like is 11 Nosakkaromisesu-Nbe (Schizosaccharomyces pombe) is respected ^ be. Animal cells such as COS cells, CH0 cells, HEK293 cells, and insect cells such as Sf9 and Si21 can also be used. When a bacterium such as Escherichia coli is used as a host, the recombinant vector of the present invention is capable of autonomous replication in the bacterium, and comprises a promoter, a ribosome binding sequence, the DNA of the present invention, and a transcription termination sequence. Is preferred. In addition, a gene that controls a promoter may be included. Examples of Escherichia coli include Escherichia coli JM109 and HB101, and examples of Bacillus subtilis include Bacillus zubuchi (TABacillus ^ Μ / ^). Any promoter can be used as long as it can be expressed in a host such as E. coli. For example, a T7 promoter derived from Escherichia coli phage, such as a trp promoter, a lac promoter, a PL promoter, and a PR promoter, may be used. An artificially designed and modified promoter such as the tac promoter may be used. The method for introducing the recombinant vector into bacteria is not particularly limited as long as it is a method for introducing DNA into bacteria. For example, a method using calcium ions, an election port poration method and the like can be mentioned.
酵母を宿主とする場合は、 例えばサッカロミセス ·セレビシェ(Sacc nmyces cerevisiae) シヽゾサッカ iミセス - ンべ {Schizosaccharomyces卿 b ) ピヒ ァ ·パストリス(尸 A/a /? などが用いられる。 この場合、 プロモーターと しては酵母中で発現できるものであれば特に限定されず、例えば gal lプロモー夕 一、 gal lOプロモーター、 ヒートショックタンパク質プロモータ一、 MF o! 1プロモ 一夕一、 PH05プロモーター、 PGKプロモーター、 GAPプロモーター、 ADHプロモー ター、 A0X1プロモータ一等が挙げられる。 酵母への組換えベクターの導入方法と しては、 酵母に DNAを導入する方法であれば特に限定されず、 例えばエレクト口 ポレーシヨン法、 スフヱ口プラスト法、 酢酸リチウム法等が挙げられる。 When yeast is used as the host, for example, Saccharomyces cerevisiae (Schizosaccharomyces lord b) Pichia pastoris (Sodium A / a /?) Is used. The promoter is not particularly limited as long as it can be expressed in yeast. For example, gal promoter, gal l promoter, heat shock protein promoter, MF o! 1 promoter One night, PH05 promoter, PGK promoter, GAP promoter, ADH promoter, A0X1 promoter and the like. The method for introducing the recombinant vector into yeast is not particularly limited as long as it is a method for introducing DNA into yeast, and examples thereof include an elect-portation method, a spout-type plast method, and a lithium acetate method.
動物細胞を宿主とする場合は、 サル細胞 COS- 7、 Vero、 チャイニーズハムス夕 —卵巣細胞 (CH0細胞)、 マウス L細胞、 ラット GH3細胞、 又はヒト FL、 HEK293, HeLa若しくは Jurkat細胞などが用いられる。 プロモーターとして SR aプロモー ター、 SV40プロモーター、 LTRプロモーター、 iS -ァクチンプロモーター等が用い られ、 また、 ヒトサイトメガロウィルスの初期遺伝子プロモーター等を用いても よい。 動物細胞への組換えベクターの導入方法としては、 例えばエレクトロボレ —シヨン法、 リン酸カルシウム法、 リポフエクシヨン法等が挙げられる。  When animal cells are used as the host, monkey cells COS-7, Vero, Chinese ham cells—ovary cells (CH0 cells), mouse L cells, rat GH3 cells, or human FL, HEK293, HeLa, or Jurkat cells are used. . As the promoter, an SRa promoter, SV40 promoter, LTR promoter, iS-actin promoter or the like may be used, or an early gene promoter of human cytomegalovirus may be used. Methods for introducing the recombinant vector into animal cells include, for example, the electrophoresis method, the calcium phosphate method, and the lipofection method.
昆虫細胞を宿主とする場合は、 Si9細胞、 Si21細胞などが用いられる。 昆虫細 胞への組換えベクターの導入方法としては、 例えばリン酸カルシウム法、 リボフ ェクシヨン法、 エレク卜口ポレーション法などが用いられる。 本発明の klotho  When insect cells are used as a host, Si9 cells, Si21 cells and the like are used. As a method for introducing a recombinant vector into an insect cell, for example, a calcium phosphate method, a riboaction method, an electoral poration method, or the like is used. Klotho of the present invention
本発明の klothoタンパク質とは、配列番号 2、配列番号 3もしくは配列番号 2 The klotho protein of the present invention refers to SEQ ID NO: 2, SEQ ID NO: 3 or SEQ ID NO: 2.
3に示されるアミノ酸配列を有するタンパク質、 またはこれらのタンパク質とA protein having the amino acid sequence shown in 3, or
80%以上、好ましくは 85 %以上のアミノ酸配列の相同性を有するヒトおよびマウ ス以外の他動物種由来の klothoタンパク質のことであり、また前述の低リン酸血 症誘導活性およびノもしくは低活性型ビタミン D血症誘導活性、 高リン酸血症誘 導活性およびノもしくは高活性型ビ夕ミン D血症誘導活性、 または FGF23の作用 を調節 (増強または阻害) する作用を有するものである。 さらに、 本発明におけ る klothoタンパク質には、低リン酸血症誘導活性および もしくは低活性型ビ夕 ミン D血症誘導活性、 高リン酸血症誘導活性および Zもしくは高活性型ビ夕ミンA klotho protein derived from other animal species other than human and mouse having 80% or more, preferably 85% or more amino acid sequence homology, and the above-mentioned hypophosphatemia-inducing activity and no or low activity It has an activity of inducing type V vitamin D, an activity of inducing hyperphosphatemia and an activity of inducing or hyperactive biminin D, or an effect of regulating (enhancing or inhibiting) the action of FGF23. Further, the klotho protein in the present invention includes a hypophosphatemic-inducing activity and / or a low-activity vitmin, a D-hyperphosphate-inducing activity, a hyperphosphatemia-inducing activity, and a Z- or highly-active vymin.
D血症誘導活性、 または FGF23の作用を調節する作用を有する限り.、 前述のアミ ノ酸配列中の 1個又は数個のアミノ酸が置換、 欠失及び、 または修飾されている アミノ酸配列を有するタンパク質も包含される。さらに、そのような置換、欠失、 修飾及び付加が複数の組み合わせの場合であつてもよい。 As long as it has D-disease-inducing activity or an effect of regulating the action of FGF23, it has an amino acid sequence in which one or several amino acids in the above-mentioned amino acid sequence are substituted, deleted and / or modified Proteins are also included. Furthermore, such substitutions, deletions, modifications and additions may be in the case of a plurality of combinations.
本発明の低リン酸血症誘導活性および もしくは低活性型ビタミン D血症誘導 活性、高リン酸血症誘導活性およびノもしくは高活性型ビ夕ミン D血症誘導活性、 または FGF23の作用を調節する作用を有する kl othoタンパク質は、配列番号 1に 示す塩基配列もしくは配列番号 1に示す塩基配列の塩基番号 1番〜 2 9 3 4番を 含む配列、または他動物種の kl othoタンパク質をコードする DNAの配列もしくは ヒト klotho DNAの塩基配列の塩基番号 1番〜 2 9 3 4番に相当する配列を含む配 列を、 発現できる形態で適当な宿主細胞に導入して形質転換細胞を調製し、 該形 質転換細胞に導入した DNAを発現させることによって製造することができる。 ま た、 このように産生されたタンパク質は、 宿主が持つタンパク質修飾機構により 切断や糖鎖付加などのポリぺプチド鎖の改変を受けることがある。 Hypophosphatemia-inducing activity and / or low-activity type vitamin D-induction of the present invention The klotho protein having activity, hyperphosphatemia-inducing activity and no- or highly-active biminmin D-hememia-inducing activity, or an effect of regulating the action of FGF23, has the nucleotide sequence shown in SEQ ID NO: 1 or SEQ ID NO: 1 A sequence containing the nucleotide numbers 1 to 293 4 of the nucleotide sequence shown in SEQ ID NO: 1, or a nucleotide sequence encoding the klotho protein of another animal species or nucleotide numbers 1 to 293 4 of the nucleotide sequence of human klotho DNA The transformant can be produced by introducing a sequence containing the sequence corresponding to No. 1 into a suitable host cell in an expressible form, preparing a transformed cell, and expressing the introduced DNA in the transformed cell. In addition, the protein thus produced may be subject to modification of the polypeptide chain such as cleavage or glycosylation by the protein modification mechanism of the host.
本発明の kl othoタンパク質は、前記形質転換体を培養し、その培養物から採取 することにより得ることができる。 「培養物」 とは、 培養上清のほか、 培養細胞若 しくは培養菌体又は細胞若しくは菌体の破砕物のいずれをも意味するものである。 本発明の形質転換体を培養する方法は、 宿主の培養に用いられる通常の方法に 従って行われる。  The klotho protein of the present invention can be obtained by culturing the transformant and collecting from the culture. The term “culture” means not only the culture supernatant but also cultured cells or cultured cells, or cells or disrupted cells. The method for culturing the transformant of the present invention is performed according to a usual method used for culturing a host.
大腸菌や酵母菌等の微生物を宿主として得られた形質転換体を培養する培地は、 微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 効率的に行うことができる培地であれば、 天然培地、 合成培地のいずれを用いて もよい。 炭素源としては、 グルコース、 フラク ] ^一ス、 スクロース、 デンプン等 の炭水化物、 酢酸、 プロピオン酸等の有機酸、 エタノール、 プロパノール等のァ ルコール類が挙げられる。 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、 酢酸アンモニゥム、 リン酸アンモニゥム等の無機酸若しくは有 機酸のアンモニゥム塩又はその他の含窒素化合物のほか、 ペプトン、 肉エキス、 コーンスティープリカ一等が挙げられる。無機物としては、リン酸第一カリウム、 リン酸第二力リゥム、リン酸マグネシウム、硫酸マグネシウム、塩化ナ卜リゥム、 硫酸第一鉄、 硫酸マンガン、 硫酸銅、 炭酸カルシウム等が挙げられる。  The culture medium for culturing transformants obtained using microorganisms such as Escherichia coli and yeast as a host contains carbon sources, nitrogen sources, inorganic salts, etc. that can be used by the microorganisms, and efficiently cultivates the transformants. As long as the medium can be used, either a natural medium or a synthetic medium may be used. Examples of the carbon source include carbohydrates such as glucose, fructose, sucrose and starch, organic acids such as acetic acid and propionic acid, and alcohols such as ethanol and propanol. Nitrogen sources include ammonia, ammonium chloride, ammonium sulfate, ammonium acetate, ammonium phosphate, and other inorganic or organic acid ammonium salts or other nitrogen-containing compounds, as well as peptone, meat extract, corn steep liquor, etc. Is mentioned. Examples of the inorganic substance include potassium phosphate monobasic, potassium phosphate phosphate, magnesium phosphate, magnesium sulfate, sodium chloride, ferrous sulfate, manganese sulfate, copper sulfate, calcium carbonate, and the like.
培養は、 通常、 振盪培養又は通気攪拌培養などの好気的条件下、 37でで 4〜48 時間行う。 培養期間中、 pHは 6. 0〜8. 0に保持する。 pHの調整は、 無機又は有機 酸、 アルカリ溶液等を用いて行う。 培養中は必要に応じてアンピシリンゃテトラ サイクリン等の抗生物質を培地に添加してもよい。  The cultivation is usually carried out at 37 under aerobic conditions such as shaking culture or aeration / agitation culture at 4 to 48 hours. During the cultivation period, the pH is maintained at 6.0 to 8.0. The pH is adjusted using an inorganic or organic acid, an alkaline solution, or the like. During the culture, an antibiotic such as ampicillin-tetracycline may be added to the medium as needed.
プロモータ一として誘導性のプロモータ一を用いた発現ベクターで形質転換し た微生物を培養する場合は、 必要に応じてインデューサーを培地に添加してもよ い。 例えば、 イソプロピル- /3 - D-チォガラクトピラノシド(IPTG)で誘導可能な Γ7 プロモー夕一を有する発現ベクターで形質転換した微生物を培養するときには、Transformation with an expression vector using an inducible promoter When culturing a microorganism, an inducer may be added to the medium as needed. For example, when culturing a microorganism transformed with an expression vector having a を 7 promoter which is inducible with isopropyl- / 3-D-thiogalactopyranoside (IPTG),
IPTG 等を培地に添加することができる。 また、 インドールアクリル酸(IAA)で誘 導可能な trpプロ乇一夕一を用いた発現べクタ一で形質転換した微生物を培養す るときには、 IAA等を培地に添加することができる。 IPTG or the like can be added to the medium. When culturing a microorganism transformed with an expression vector using a trp protein that can be induced with indoleacrylic acid (IAA), IAA or the like can 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 RPMH640培地、 DMEM培地又はこれらの培地に牛胎児血清等を添加 した培地等が挙げられる。培養は、 通常、 5 %C02存在下、 37°Cで 1〜10日行う。 培養中は必要に応じてカナマイシン、 ベニシリン等の抗生物質を培地に添加して もよい。培養後、本発明の klothoタンパク質が菌体内又は細胞内に生産される場 合には、 超音波処理、 凍結融解の繰り返し、 ホモジナイザー処理などを施して菌 体又は細胞を破碎することにより目的のポリペプチドを採取する。 また、 本発明 の klothoタンパク質が菌体外又は細胞外に生産される場合には、培養液をそのま ま使用するか、 遠心分離等により菌体又は細胞を除去する。 その後、 タンパク質 の単離精製に用いられる一般的な生化学的方法、 例えば硫酸アンモニゥム沈殿、 ゲルクロマトグラフィー、 イオン交換クロマトグラフィー、 ァフィ二ティーク口 マトグラフィ一等を単独で又は適宜組み合わせて用いることにより、 前記培養物 中から本発明のポリペプチドを単離精製することができる。 Examples of a medium for culturing a transformant obtained using animal cells as a host include commonly used RPMH640 medium, DMEM medium, and a medium obtained by adding fetal calf serum or the like to such a medium. Culturing is usually, 5% C0 2 presence is performed 1-10 days at 37 ° C. During culture, antibiotics such as kanamycin and benicillin may be added to the medium as needed. When the klotho protein of the present invention is produced intracellularly or intracellularly after culturing, the objective polyclones are disrupted by sonication, repeated freeze-thawing, homogenizer treatment and the like to disrupt the cells or cells. Collect the peptide. When the klotho protein of the present invention is produced outside the cells or cells, the culture solution is used as it is, or the cells or cells are removed by centrifugation or the like. Thereafter, by using a general biochemical method used for isolation and purification of the protein, for example, ammonium sulfate precipitation, gel chromatography, ion exchange chromatography, affinity mouth chromatography, etc., alone or in an appropriate combination, The polypeptide of the present invention can be isolated and purified from the culture.
本発明の klothoタンパク質の in vivoでの活性は、 本発明の klothoタンパク 質を発現する上述の組み換え体細胞をヌードマウスの皮下に移植する実験および 可溶化 klothoタンパク質をマウスの静脈内に投与する実験により評価した。  The in vivo activity of the klotho protein of the present invention is determined by the experiments of subcutaneously transplanting the above-mentioned recombinant cells expressing the klotho protein of the present invention into nude mice and the experiment of administering solubilized klotho protein intravenously to mice. Was evaluated.
本試験において移植した細胞は、 ヌードマウスの皮下において増殖して腫瘍を 形成する。 これに伴い細胞が生産し、 分泌する本発明のポリペプチドはマウスの 体液中に放出されることを特徴としている。 この実験において、 図 8 (実施例 1 The cells implanted in this study grow subcutaneously in nude mice to form tumors. Accordingly, the polypeptide of the present invention, which is produced and secreted by cells, is released into the body fluid of mice. In this experiment, FIG. 8 (Example 1)
3 ) に示すように、本発明の klothoタンパク質を発現する細胞を移植したマウス は、 本発明の DNAを導入していない対照の CH0細胞を移植して腫瘍を形成させた 個体、 又は腫瘍を形成していない個体と比較して、 明らかな低リン酸血症および 低活性型ビタミン D血症を呈した。 このことから、本発明の klothoタンパク質は 低リン酸血症誘導活性および低活性型ビタミン D血症誘導活性を有していること が明らかとなった。 As shown in 3), mice transplanted with cells expressing the klotho protein of the present invention were transplanted with control CH0 cells not transfected with the DNA of the present invention to form individuals or tumors. They had apparent hypophosphatemia and hypoactive vitamin Demia compared to non-treated individuals. This indicates that the klotho protein of the present invention has hypophosphatemia-inducing activity and low-active vitamin Demia-inducing activity. Became clear.
また、マウスに可溶化 klothoタンパク質を投与した場合、血中活性型ビタミン D濃度の上昇が認められ、本発明の klothoタンパク質は高リン酸血症誘導活性お よび高活性型ビタミン D血症誘導活性を有していることも明らかとなった。 副甲 状腺ホルモンは持続投与と間歇投与で骨に対する作用が逆になることも明らかと なっており、本発明の klothoタンパク質も投与方法の違いにより異なる活性を示 すことが示唆された。  In addition, when solubilized klotho protein was administered to mice, an increase in blood active vitamin D concentration was observed, and the klotho protein of the present invention showed hyperphosphatemia-inducing activity and highly active vitamin D-inducing activity. It became clear that it had. It has also been clarified that parathyroid hormone reverses the effect on bone between continuous administration and intermittent administration, suggesting that the klotho protein of the present invention also shows different activities depending on the administration method.
本発明においては、 上記タンパク質を修飾することもできる。 例えば、 ポリエ チレングリコール、 デキストラン、 ポリ (N-ビエル—ピロリドン)、 ポリプロピレ ングリコールホモポリマー、ポリプロピレンォキシド /エチレンォキシドのコポリ マ一、 ポリオキシェチル化ポリオール、 ポリピニルアルコールなどを適宜選択し て使用する。 修飾方法は、 公知の任意の手法を採用することができ、 例えば特表 平 10-510980号公報に詳細に開示されている。 抗 klothoタンパク質抗体  In the present invention, the above proteins can be modified. For example, appropriately select and use polyethylene glycol, dextran, poly (N-vier-pyrrolidone), polypropylene glycol homopolymer, polypropylene oxide / ethylene oxide copolymer, polyoxyethylated polyol, polypinyl alcohol, etc. I do. As the modification method, any known method can be adopted, and is disclosed in detail, for example, in Japanese Patent Publication No. H10-510980. Anti-klotho protein antibody
本発明における抗 klothoタンパク質抗体とは、 前記に定義したような klotho タンパク質またはその一部に反応性を有する抗体あるいは抗体の一部である。 本 発明の抗体には、 抗体を構成する重鎖及び Zまたは軽鎖の各々のアミノ酸配列に おいて、 1または数個のアミノ酸が欠失、 置換若しくは付加されたアミノ酸配列 を有する重鎖及び Zまたは軽鎖からなり、 k 101 hoタンパク質と結合し得るモノク 口一ナル抗体も包含される。本発明の抗 klothoタンパク質抗体のアミノ酸配列中 に、 前記のようなアミノ酸の部分的改変 (欠失、 置換、 挿入、 付加) は、 そのァ ミノ酸配列をコードする塩基配列を部分的に改変することにより導入することが できる。 これらの改変技術は当業者に周知であり、 市販の突然変異導入キット等 を使用することができる。  The anti-klotho protein antibody in the present invention is an antibody or a part of an antibody reactive with the klotho protein or a part thereof as defined above. The antibody of the present invention includes a heavy chain and a Z having an amino acid sequence in which one or several amino acids are deleted, substituted or added in the amino acid sequence of each of the heavy chain and Z or the light chain constituting the antibody. Alternatively, a monoclonal antibody which consists of a light chain and can bind to the k101ho protein is also included. In the amino acid sequence of the anti-klotho protein antibody of the present invention, the partial modification of amino acids (deletion, substitution, insertion, addition) as described above partially modifies the nucleotide sequence encoding the amino acid sequence. Can be introduced. These modification techniques are well known to those skilled in the art, and a commercially available mutagenesis kit or the like can be used.
(1) klothoタンパク質の部分的構造を特異的に認識して結合する抗体 '  (1) Antibodies that specifically recognize and bind to the partial structure of klotho protein ''
klotho 夕ンパク質の検出や生物活性の調節に有用な抗体を取得するには、 klothoタンパク質の構造的特徴を認識する抗体、 高親和性を有する抗体、 生物活 性を中和できる抗体などの取得が有効である。 Klothoタンパク質の構造や抗原性 については不明であるので、 klothoタンパク質の部分配列に相当する複数のぺプ チドを合成して、 各ペプチドに対する抗体の取得を行った。 これらを用いて発現 細胞が分泌した klotho タンパク質の検出をウエスタンブロッテイングにより行 ない、発現細胞中および培養上清中には成熟型 klothoタンパク質が発現している ことを明らかにした。また、得られた抗体は成熟型 klothoタンパク質とその切断 により生じるペプチドに対して多様な結合特異性を示した。 To obtain antibodies that are useful for detecting klotho proteins and regulating biological activities, obtain antibodies that recognize structural characteristics of klotho proteins, antibodies with high affinity, and antibodies that can neutralize biological activities. Is valid. Since the structure and antigenicity of the Klotho protein are unknown, multiple An antibody against each peptide was obtained by synthesizing the tide. Using these, the klotho protein secreted by the expressing cells was detected by Western blotting, and it was revealed that the mature klotho protein was expressed in the expressing cells and in the culture supernatant. In addition, the obtained antibodies showed various binding specificities for the mature klotho protein and the peptide generated by its cleavage.
(2) 抗体の製造  (2) Antibody production
本発明の抗体は、 例えば、 下記のような製造方法によって製造することができ る。 即ち、 例えば、 前記で定義したような klothoタンパク質又はその一部、 ある いは抗原の抗原性を高めるための適当な物質 (例えば、 bovine serum albumin等) との結合物を、 必要に応じて免疫賦活剤 (Freimd' s Adjuvant 等) とともに、 ヒ ト抗体産生トランスジエニックマウス等を含む非ヒト哺乳動物に免疫する。 ある いは、 klothoタンパク質をコードする DNAを組み込んだ発現べクタ一を投与する ことにより免疫感作を行うことができる。 ポリクローナル抗体は、 免疫感作動物 から得た血清から取得することができる。 またモノクローナル抗体は、 免疫感作 動物から得た抗体産生細胞と自己抗体産生能のない骨髄腫系細胞 (ミエローマ細 胞) からハイプリドーマを調製し、 ハイプリ ドーマをクローン化し、 免疫に用い た抗原に対して特異的親和性を示すモノクローナル抗体を産生するクローンを選 択することによって製造される。  The antibody of the present invention can be produced, for example, by the following production method. That is, for example, the klotho protein as defined above, or a part thereof, or a conjugate with an appropriate substance (for example, bovine serum albumin or the like) for enhancing the antigenicity of the antigen is optionally immunized. Immunize non-human mammals, including human antibody-producing transgenic mice, together with activators (such as Freimd's Adjuvant). Alternatively, immunization can be performed by administering an expression vector incorporating DNA encoding the klotho protein. Polyclonal antibodies can be obtained from serum obtained from immunized animals. Monoclonal antibodies were prepared by preparing hybridomas from antibody-producing cells obtained from immunized animals and myeloma cells (myeloma cells) that lack the ability to produce autoantibodies, cloned the hybridomas, and used them as antigens for immunization. It is produced by selecting a clone that produces a monoclonal antibody showing specific affinity for it.
さらに具体的には下記のようにして製造することができる。 モノクローナル抗 体を分泌するハイプリドーマの調製は、 ケーラー及びミルシュタインらの方法 More specifically, it can be produced as follows. Hybridomas secreting monoclonal antibodies were prepared according to the method of Kohler and Milstein et al.
(Nature, 1975 Vol. 256 : 495-497) 及びそれに準じて行うことができる。 即ち、 前述の如く免疫感作された動物から取得される脾臓、リンパ節、骨髄又は扁桃等、 好ましくはリンパ節または脾臓に含まれる抗体産生細胞と、 好ましくはマウス、 ラット、 モルモット、 ハムスター、 ゥサギまたはヒト等の哺乳動物に由来する自 己抗体産生能のないミエローマ細胞と細胞融合させることにより調製される。 モ ノクローナル抗体を産生するハイプリドーマクローンのスクリーニングは、 ハイ ブリ ドーマを、 例えばマイクロ夕イタ一プレート中で培養し、 増殖の見られたゥ エル中の培養上清の免疫抗原に対する反応性を、 例えば ELISA等の酵素免疫測定 法によつて測定することにより行なうことができる。 (Nature, 1975 Vol. 256: 495-497) and the method according to it. That is, antibody producing cells contained in the spleen, lymph node, bone marrow, tonsil, etc., preferably in the lymph node or spleen, obtained from the animal immunized as described above, and preferably in mice, rats, guinea pigs, hamsters, and egrets. Alternatively, it is prepared by cell fusion with a myeloma cell derived from a mammal such as a human or the like which is not capable of producing autoantibodies. Screening of hybridoma clones producing monoclonal antibodies is performed by culturing hybridomas, for example, in a microplate, and determining the reactivity of the culture supernatant in the wells in which proliferation has been observed with the immunogen, for example. The measurement can be performed by enzyme immunoassay such as ELISA.
ハイプリドーマからのモノクローナル抗体の製造は、 ハイブリドーマをインビ トロで培養して培養上清から単離することにより行える。また、マウス、ラット、 モルモット、 ハムスターまたはゥサギ等の腹水中等でのインピポで培養し、 腹水 から単離することもできる。 Production of monoclonal antibodies from hybridomas It can be performed by culturing in a toro and isolating from the culture supernatant. Alternatively, it can be isolated from ascites by culturing in an ascitic fluid in ascites of mice, rats, guinea pigs, hamsters, or egrets.
また、 ハイプリドーマ等の抗体産生細胞からヒトモノクローナル抗体をコード する遺伝子をクロ一ニングし、 適当なベクターに組み込んで、 これを宿主 (例え ば哺乳類細胞細胞株、 大腸菌、 酵母細胞、 昆虫細胞、 植物細胞など) に導入し、 遺伝子組換え技術を用いて産生させた組換型抗体を調製することができる In addition, a gene encoding a human monoclonal antibody is cloned from an antibody-producing cell such as a hybridoma and inserted into an appropriate vector, which is then used as a host (eg, a mammalian cell line, Escherichia coli, yeast cells, insect cells, plant cells, etc.). Cells, etc.) to produce recombinant antibodies produced using genetic recombination technology.
( P. J. Delves. , ANTIBODY PRODUCTION ESSENTIAL TECHNIQUES. , 1997 WILEY、 P. Shepherd and C. Dean. , Monoc lonal Ant ibodies., 2000 OXFORD UNIVERSITY PRESS, J. W. God ing., Monoc l onal Ant ibod ies : princ iples and pract ice. , 1993 ACADEMI C PRESS) 0 さらに、 トランスジエニック動物作製技術を用いて目的抗体の遺伝子が 内在性遺伝子に組み込まれたトランスジエニックなゥシ、 ャギ、 ヒッジまたはブ 夕を作製し、 そのトランスジエニック動物のミルク中からその抗体遺伝子に由来 するモノクローナル抗体を大量に取得することも可能である。 ハイブリドーマを インビトロで培養'する場合には、 培養する細胞種の特性、 試験研究の目的及び培 養方法等の種々条件に合わせて、 ハイプリドーマを増殖、 維持及び保存させ、 培 養上清中にモノクローナル抗体を産生させるために用いられるような既知栄養培 地あるいは既知の基本培地から誘導調製されるあらゆる栄養培地を用いて実施す ることが可能である。 (PJ Delves., ANTIBODY PRODUCTION ESSENTIAL TECHNIQUES., 1997 WILEY, P. Shepherd and C. Dean., Monoclonal Ant ibodies., 2000 OXFORD UNIVERSITY PRESS, JW Goding., Monoclonal Ant ibod ies: princ iples and pract ice., 1993 ACADEMI C PRESS) 0 Further, using a transgenic animal production technology, a transgenic transgenic goat, goat, sheep, or horse in which the gene of the antibody of interest has been incorporated into the endogenous gene is produced. It is also possible to obtain a large amount of a monoclonal antibody derived from the antibody gene from the milk of the transgenic animal. When hybridomas are cultured in vitro, the hybridomas are grown, maintained, and preserved in accordance with various conditions such as the characteristics of the cell type to be cultured, the purpose of the test and research, and the culture method. It can be carried out using any known nutrient medium such as that used to produce monoclonal antibodies or any nutrient medium derived and prepared from a known basal medium.
本発明のポリクローナル抗体は、 実施例 1 0に示すように、 化学合成した klot o タンパク質の部分べプチドとキヤリァタンパク質であるゥシサイログロ ブリンと結合させたものを作製し、 ゥサギを免疫した。 免疫により誘導された各 ぺプチドに対する抗体は、 免疫に用いたペプチドを固相化したァフィ二ティー力 ラムで精製した。 このようにして得た抗体の特性については、 ウエスタンブロッ ティングにて検討し klothoタンパクに対する反応性を明らかにした。 また、実施 例 2 2および 2 3に示すように精製可溶化 klotho タンパク質を抗原に'用いて、 klothoタンパク質に対する特異的抗体を作製した。  As shown in Example 10, a polyclonal antibody of the present invention was prepared by binding a chemically synthesized partial peptide of kloto protein to cysiglobulin, which is a carrier protein, to immunize egrets. Antibodies to each of the peptides induced by immunization were purified by an affinity column on which the peptide used for immunization was immobilized. The characteristics of the antibody thus obtained were examined by Western blotting, and the reactivity with the klotho protein was clarified. Further, as shown in Examples 22 and 23, a specific antibody against the klotho protein was prepared using the purified solubilized klotho protein as an antigen.
FGF23作用 FGF23 action
FGF23は生体内においてリンゃ 1, 25Dの代謝に重要な役割を有する。 例えば、 ADHRの患者において FGF-23遺伝子のミスセンス変異が低リン血症性疾患の惹起 に関与していることが示唆された (The ADHR Consortium. Autosomal dominant hypophosphatemic rickets is associated with mutations in FGF23. Nature Genet. 26: 345-348, 2000)。 さらに我々は腫瘍性骨軟化症において腫瘍が産生する疾患 惹起因子として FGF- 23を同定した(Shimada, T. , Mizutani, S. , Muto, Τ., Yoneya, T. , Hino, R. Takeda, S, Takeuchi, Y. , Fuj ita, T., Fukumoto, S and Yamashita, T. Cloning and chatacterizat ion of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl. Acad. Sci. 98:6500-6505, 2001)。 これらの研究により、 FGF-23が低リン血症性疾患やクル病 ·骨軟化症を伴う疾患 に関与している可能性があるることが示唆された。 FGF23 has an important role in the metabolism of phosphorus 1,25D in vivo. For example, It has been suggested that missense mutations in the FGF-23 gene are involved in the induction of hypophosphatemic disease in ADHR patients (The ADHR Consortium. Autosomal dominant hypophosphatemic rickets is associated with mutations in FGF23. Nature Genet. 26 : 345-348, 2000). In addition, we have identified FGF-23 as a tumor-inducing disease-causing factor in neoplastic osteomalacia (Shimada, T., Mizutani, S., Muto, Τ., Yoneya, T., Hino, R. Takeda, S, Takeuchi, Y., Fujita, T., Fukumoto, S and Yamashita, T. Cloning and chatacterization of FGF23 as a causative factor of tumor-induced osteomalacia. Proc Natl. Acad. Sci. 98: 6500-6505, 2001). These studies suggested that FGF-23 may be involved in hypophosphatemic diseases and diseases associated with cull and osteomalacia.
遺伝性低リン血症のなかで最も罹患率の高い疾患である XLHは約 2万人に 1人 の割合で存在するといわれている。 XLHの責任遺伝子は PHEXとして同定されてい るが、本遺伝子は 個のェクソンからなり、その遺伝子領域は 220kbにわたるた め遺伝病の原因となる変異解析は現在のところ診断目的では不可能な状況である。 突発的発症例や成人発症型の存在も示唆されている。 これまでのところ PHEX と FGF-23の関連は明らかにされていない。 XLHのモデル動物として自然発症変異マ ウスである Hypマウスが知られている。 このマウスでは PHEX遺伝子の 3'領域の 欠損が確認されており、 低リン血症、 クル病 ·骨軟化症を呈することが知られて いる。 我々は、 血中 FGF23測定系を用いて、 Hypマウスにおける FGF- 23の血中濃 度の測定を行い、 FGF- 23の濃度が著しく高いことを見出し、 Hypマウスの低リン 血漿、 クル病、 骨軟化症が FGF23の高値により引き起こされている可能性を見出 した (Increased circulatory level of biologically active full-length FGF-23 in patients with hypophosphatemic rickets/osteomalacia. J Clin Endocrinol Me tab. 2002 Nov;87(ll) :4957-60)。  XLH, the most prevalent of the inherited hypophosphatemia, is said to be present in about 1 in 20,000 people. Although the gene responsible for XLH has been identified as PHEX, this gene consists of a single exon, and its gene region spans 220 kb, so that mutation analysis that causes a genetic disease is currently impossible for diagnostic purposes. is there. Sudden cases and adult-onset cases have also been suggested. So far, the association between PHEX and FGF-23 has not been clarified. Hyp mice, which are spontaneously mutated mice, are known as model animals for XLH. In this mouse, the deletion of the 3 'region of the PHEX gene has been confirmed, and it is known that the mouse exhibits hypophosphatemia, cull disease and osteomalacia. We measured the blood concentration of FGF-23 in Hyp mice using the blood FGF23 measurement system, and found that the concentration of FGF-23 was extremely high. We found that osteomalacia may be caused by elevated levels of FGF23 (Increased circulatory level of biologically active full-length FGF-23 in patients with hypophosphatemic rickets / osteomalacia.J Clin Endocrinol Me tab. 2002 Nov; 87 ( ll): 4957-60).
以上のように FGF- 23は、血中のリン濃度と 1, 25D濃度を低下させる活性を有し ている。 FGF- 23活性の中和実験により、 我々は FGF- 23が健常時においてもリン や 1, 25Dの代謝バランスを維持する上で重要な役割を有することを明らかにした As described above, FGF-23 has an activity to reduce blood phosphorus concentration and 1.25D concentration. Experiments on neutralization of FGF-23 activity revealed that FGF-23 plays an important role in maintaining the metabolic balance of phosphorus and 1,25D even in normal health
(W003/057733)o 従って、 FGF- 23は、 リン代謝およびビタミン D代謝に関与して おり、 これらの関わる疾患、 例えば腫瘍性骨軟化症、 ADHR, XLH,腎性骨異栄養症、 透析骨症、 骨粗しょう症、 低リン血症、 クル病、 骨軟化症、 尿細管機能障害、 骨 減少症、 低カルシウム血症、 高リン血症、 高 1, 25D血症、 副甲状腺機能亢進症、 異所性石灰化、 搔痒、 骨硬化症、 パジェット病、 高カルシウム血症、 副甲状腺機 能低下症、 骨痛、 筋力低下、 骨格変形、 成長障害、 および低 1, 25D血症などの疾 患、を FGF- 23の作用をコントロールすることにより治療を可能とすることが考え られる。 また、 実施例 2 8に示すように、 FGF23と可溶化 klothoタンパク質の複 合体を添加することにより骨芽細胞由来細胞に刺激を加え、 細胞形態や細胞内の シグナル伝達系に影響を与えることが明らかとなった。 これらのことから、 FGF23 と klotho タンパク質は骨芽細胞などの骨代謝に関わる細胞に直接作用すること が示唆され、骨代謝に関わる疾患を FGF23の作用を klotho夕ンパク質ゃ抗 klo tho 抗体を用いてコントロールすることにより治療を可能とすることが考えられる。 (W003 / 057733) o Therefore, FGF-23 is involved in phosphorus metabolism and vitamin D metabolism, and is associated with diseases such as neoplastic osteomalacia, ADHR, XLH, renal osteodystrophy, Dialysis osteopathy, osteoporosis, hypophosphatemia, rickets, osteomalacia, renal tubular dysfunction, osteopenia, hypocalcemia, hyperphosphatemia, hyper1,25D blood disease, hyperparathyroidism Disease, ectopic calcification, pruritus, bone sclerosis, Paget's disease, hypercalcemia, hypoparathyroidism, bone pain, muscle weakness, skeletal deformity, growth disorders, and hypo1,25D Diseases may be treated by controlling the action of FGF-23. In addition, as shown in Example 28, the addition of a complex of FGF23 and solubilized klotho protein stimulates osteoblast-derived cells, which may affect cell morphology and intracellular signal transduction. It became clear. These results suggest that FGF23 and klotho protein directly act on cells related to bone metabolism such as osteoblasts, and that the effects of FGF23 on bone metabolism-related diseases can be measured using klotho protein-anti-klotho antibody. It is thought that treatment can be made possible by controlling the temperature.
FGF23と klothoタンパク質の複合体 Complex of FGF23 and klotho protein
本発明でいう FGF23と klothoタンパク質の複合体とは FGF23分子 (W002/14504 記載)と前記に定義したような kl othoタンパク質を混合することにより製造でき る。 本発明における FGF23分子は遺伝子組み換え技術の他、 化学的合成法、 細胞 培養方法等のような技術的分野において知られる公知の方法又はその修飾方法を 適宜用いることにより製造することができる。 例えば FGF23 は島田らの方法 {Endocrinology 143, 3179 (2002) } により製造することができる。 また、 本発 明における klothoタンパク質も同様に遺伝子組み替え技術の他、 化学的合成法、 細胞培養方法等のような技術的分野において知られる公知の方法又はその修飾方 法を適宜用いることにより製造することができる。  The complex of FGF23 and klotho protein in the present invention can be produced by mixing an FGF23 molecule (described in W002 / 14504) with a klotho protein as defined above. The FGF23 molecule in the present invention can be produced by appropriately using a known method known in the technical field such as a chemical synthesis method, a cell culture method, or a modification method thereof, in addition to a genetic recombination technique. For example, FGF23 can be produced by the method of Shimada et al {Endocrinology 143, 3179 (2002)}. Similarly, the klotho protein of the present invention is also produced by appropriately using a known method or a modification method known in the technical field such as a chemical synthesis method, a cell culture method, etc., in addition to the genetic recombination technique. be able to.
本発明の複合体は例えば下記のような製造方法によっても製造することができ る。すなわち、前述した FGF23と klothoタンパク質を混合し、数分〜数時間、 4°C 〜室温条件下で静置することにより作製することが可能である。 また、 複合体の 結 合 を よ り 強 固 に す る 為 に 、 Di succ inimidyl suberate , Bi s (sul fosucc inimidyl) suberate などの架橋剤を用いることも可能である。また、 FGF23と klothoタンパク質との結合遺伝子を遺伝子組み換え技術の他、化学的合 成法、 細胞培養方法等のような技術的分野において知られる公知の方法又はその 修飾方法を適宜用いることにより製造することができる。 また、 生体内に存在す る FGF23— klo tho タンパク質複合体を抗 FGF23 抗体結合ビーズ、 もしくは抗 kl o tho 夕ンパク質抗体結合ビーズを用いて精製することにより製造することも 可能である。 本発明の kl othoタンパク質を含む医薬組成物 The composite of the present invention can also be produced, for example, by the following production method. That is, it can be prepared by mixing the above-mentioned FGF23 and the klotho protein and leaving them to stand at 4 ° C. to room temperature for several minutes to several hours. In order to further strengthen the binding of the complex, a cross-linking agent such as di succ inimidyl suberate and bis (sul fosucc inimidyl) suberate can be used. Also, in addition to the gene recombination technique, a known method known in the technical field such as a chemical synthesis method, a cell culture method, or the like, may be used for the gene linked to the FGF23 and klotho protein. It can be produced by appropriately using a modification method. It can also be produced by purifying an FGF23-klo tho protein complex present in a living body using anti-FGF23 antibody-bound beads or anti-klo tho protein antibody-bound beads. Pharmaceutical composition comprising the klotho protein of the present invention
本発明の kl othoタンパク質は、血中リン酸濃度の上昇もしくは低下および Zま たは血中活性型ビ夕ミン D濃度の上昇もしくは低下、 または FGF23の作用の上昇 もしくは低下が好ましくない疾患に対して医薬組成物として用いることができる。 実施例 1 3、 2 8および 2 9のように本 kl o thoタンパク質もしくは本 kl o tho夕 ンパク質を含有する医薬組成物は血中リン、 血中活性型ビタミン D濃度もしくは FGF23作用を変化させる作用があるため、血中リン酸濃度、血中活性型ビタミン D 濃度、 FGF23作用の変化が引き起こされるような疾患、例えば高リン血症、高 1, 25D 血症、 腎性骨異栄養症、 透析骨症、 骨粗鬆症、 尿細管機能障害、 骨減少症、 低力 ルシゥム血症、 副甲状腺機能亢進症、 異所性石灰化、 接痒、 骨硬化症、 パジエツ ト病、 高カルシウム血症、 副甲状腺機能低下症、 骨痛、 筋力低下、 骨格変形、 お よび成長障害などの治療に利用することができる。 本発明の抗 kl othoタンパク質抗体を含む医薬組成物.  The klotho protein of the present invention is useful for increasing or decreasing blood phosphate concentration and increasing or decreasing Z or blood activated biminmin D concentration, or for diseases in which increasing or decreasing the action of FGF23 is undesirable. Can be used as a pharmaceutical composition. As in Examples 13, 28 and 29, a pharmaceutical composition containing the present klo tho protein or the present klo tho protein changes blood phosphorus, blood active vitamin D concentration or FGF23 action. A disease that causes changes in blood phosphate levels, blood active vitamin D levels, and FGF23 activity, such as hyperphosphatemia, hyper1,25D blood disease, renal osteodystrophy, Dialysis osteopathy, osteoporosis, renal tubular dysfunction, osteopenia, hypotensive lucidemia, hyperparathyroidism, ectopic calcification, pruritus, osteosclerosis, Paget's disease, hypercalcemia, vice It can be used to treat hypothyroidism, bone pain, muscle weakness, skeletal deformity, and growth disorders. A pharmaceutical composition comprising the anti-klotho protein antibody of the present invention.
実施例 1 4にあるように本発明の抗体は血中リン酸濃度および Zまたは血中活 性型ビタミン D濃度を上昇させる作用および体内での FGF 23の作用を阻害する作 用がある。 このことから血中リンおよび または血中活性型ビタミン D濃度の変 化が影響していると考えられる疾患、 例えば腫瘍性骨軟化症、 ADHR、 XLH などに 対して医薬組成物として用いることができ、 これらの疾患に共通して認められる 低リン血症、 骨石灰化不全、 骨痛、 筋力低下、 骨格の変形、 成長障害、 低 1, 25D 血症について改善効果が期待できる。また FGF-23が生理的条件下で重要な役割を 果たしており、 FGF- 23のリン代謝調節作用や FGF-23が調節するビタミン D代謝 を介したカルシウム代謝調節作用を本発明の抗体が制御することによって、 骨粗 鬆症、 クル病、高カルシウム血症、低カルシウム血症、 異所性石灰化、 骨硬化症、 パジェット病、 副甲状腺機能亢進症、 副甲状腺機能低下症、 搔痒等のミネラル代 謝やビタミン D代謝の異常に起因する疾患に対して治療的および予防的に医薬組 成物として用いることができる。 本発明の FGF23と klotho夕ンパク質の複合体を含む医薬品組成物 As described in Example 14, the antibody of the present invention has the effect of increasing the blood phosphate concentration and the concentration of Z or active vitamin D in the blood and the effect of inhibiting the action of FGF23 in the body. Therefore, it can be used as a pharmaceutical composition for diseases considered to be affected by changes in blood phosphorus and / or active vitamin D concentration, such as neoplastic osteomalacia, ADHR, and XLH. It can be expected to improve hypophosphatemia, bone calcification insufficiency, bone pain, muscle weakness, skeletal deformity, growth disorders, and low 1,25Demia, which are commonly found in these diseases. In addition, FGF-23 plays an important role under physiological conditions, and the antibody of the present invention controls the action of FGF-23 to regulate phosphorus metabolism and the action of FGF-23 to regulate calcium metabolism via vitamin D metabolism In some cases, minerals such as osteoporosis, rickets, hypercalcemia, hypocalcemia, ectopic calcification, sclerosis, Paget's disease, hyperparathyroidism, hypoparathyroidism, and pruritus Teens It can be used therapeutically and prophylactically as a pharmaceutical composition for diseases caused by abnormalities in vitamin X metabolism and vitamin D metabolism. Pharmaceutical composition comprising the complex of FGF23 and klotho protein of the present invention
FGF23と可溶化 klothoタンパク質は結合し (実施例 1 2 )、 klothoタンパク質 存在下でのみ FGF23添加により細胞内シグナル (ERKリン酸化亢進、 EGR1発現上 昇) が引き起こされること (実施例 6, 7、 8 ) や、 可溶化 klothoタンパク質発 現細胞の移植実験で血中リンおよび Zまたは血中活性型ビタミン Dの濃度が低下 すること (実施例 1 3 ) あるいは上昇すること (実施例 2 9 ) などから FGF23と klotho タンパク質とはなんらかの相互作用により血中リンおよび または血中 活性型ビタミン Dの濃度を変化させていることが考えられる。 ゆえに、 本発明の Klothoタンパク質と FGF23との複合体は、血中リン酸濃度の上昇もしくは低下お よび Zまたは血中活性型ビ夕ミン D濃度の上昇もしくは低下が好ましくない疾患 に対して医薬組成物として用いることができる。 すなわち血中リン酸濃度、 血中 活性型ビタミン!)濃度の変化が引き起こされるような疾患、 例えば高リン血症、 高 1, 25D血症、 腎性骨異栄養症、 透析骨症、 骨粗鬆症、 尿細管機能障害、 骨減少 症、 低カルシウム血症、 副甲状腺機能亢進症、 異所性石灰化、 搔痒、 骨硬化症、 パジェット病、 高カルシウム血症、 副甲状腺機能低下症、 骨痛、 筋力低下、 骨格 変形、 成長障害などの治療に利用することができる。 本発明の医薬組成物の使用  FGF23 binds to solubilized klotho protein (Example 12), and addition of FGF23 induces intracellular signals (enhanced ERK phosphorylation and increased EGR1 expression) only in the presence of klotho protein (Examples 6, 7, 8) or decreased (Example 13) or increased (Example 13) blood phosphorus and Z or active vitamin D levels in blood in transplantation experiments with solubilized klotho protein-expressing cells. This suggests that FGF23 and klotho protein may alter the concentration of blood phosphorus and / or active vitamin D in the blood by some interaction. Therefore, the complex of the Klotho protein and FGF23 of the present invention can be used as a pharmaceutical composition for diseases in which an increase or decrease in blood phosphate concentration and an increase or decrease in Z or blood activated biminin D concentration are undesirable. It can be used as a product. In other words, diseases that cause changes in blood phosphate concentration and blood active vitamin!) Concentration, such as hyperphosphatemia, hyper1,25D, renal osteodystrophy, dialysis osteopathy, osteoporosis, Renal tubular dysfunction, osteopenia, hypocalcemia, hyperparathyroidism, ectopic calcification, pruritus, bone sclerosis, Paget's disease, hypercalcemia, hypoparathyroidism, bone pain, muscle strength It can be used for treatment of depression, skeletal deformation, growth disorders, etc. Use of the pharmaceutical composition of the present invention
本発明の klothoタンパク質、抗 klothoタンパク質抗体、および klothoタンパ ク質と FGF23との複合体を有効成分として含む医薬組成物は、 医薬的に許容され る担体又は添加物を共に含むものであってもよい。 このような担体及び添加物の 例として、 水、 医薬的に許容される有機溶剤、 コラーゲン、 ポリビニルアルコー ル、ポリビニルピロリドン、力ルポキシビ二ルポリマ一、アルギン酸ナドリゥム、 水溶性デキストラン、 カルポキシメチルスターチナトリウム、 ぺクチン、 キサン タンガム、 アラビアゴム、 カゼイン、 ゼラチン、 寒天、 グリセリン、 プロピレン グリコール、 ポリエチレングリコール、 ワセリン、 パラフィン、 ステアリルアル コール、 ステアリン酸、 ヒト血清アルブミン、 マンニトール、 ソルビトール、 ラ ク卜ース、 医薬添加物として許容される界面活性剤などが挙げられる。 使用され る添加物は、 本発明の剤形に応じて上記の中から適宜又は組み合わせて選択され る。 The pharmaceutical composition of the present invention containing the klotho protein, the anti-klotho protein antibody, and the complex of the klotho protein and FGF23 as an active ingredient may contain a pharmaceutically acceptable carrier or additive. Good. Examples of such carriers and additives include water, pharmaceutically acceptable organic solvents, collagen, polyvinyl alcohol, polyvinylpyrrolidone, lipoxyvinyl polymer, nadridium alginate, water-soluble dextran, sodium carboxymethyl starch, Pectin, xanthan gum, gum arabic, casein, gelatin, agar, glycerin, propylene glycol, polyethylene glycol, petrolatum, paraffin, stearyl alcohol, stearic acid, human serum albumin, mannitol, sorbitol, la Kutose, a surfactant acceptable as a pharmaceutical additive, and the like. The additives to be used are appropriately or in combination selected from the above depending on the dosage form of the present invention.
本発明の klothoタンパク質、 抗 klothoタンパク質抗体、 klothoタンパク質と FGF23 との複合体を含む予防剤又は治療剤は、 経口的又は非経口的に投与するこ とができる。  The prophylactic or therapeutic agent containing the klotho protein, anti-klotho protein antibody, and complex of klotho protein and FGF23 of the present invention can be administered orally or parenterally.
Klothoタンパク質と FGF23との複合体を投与する代わりに、夫々のタンパク質 を同時に投与することも有効である。  Instead of administering a complex of Klotho protein and FGF23, it is also effective to administer each protein simultaneously.
経口的に投与する場合は、 それに適用される錠剤、 顆粒剤、 散剤、 丸剤などの 固形製剤、 あるいは液剤、 シロップ剤などの液体製剤等とすればよい。 特に顆粒 剤及び散剤は、 カプセル剤として単位量投与形態とすることができ、 液体製剤の 場合は使用する際に再溶解させる乾燥生成物にしてもよい。  In the case of oral administration, solid preparations such as tablets, granules, powders, and pills to be applied thereto, or liquid preparations such as liquids and syrups may be used. In particular, granules and powders can be in unit dosage form as capsules, or in the case of liquid preparations, as dry products to be re-dissolved when used.
これら剤形のうち経口用固形剤は、 通常それらの組成物中に製剤上一般に使用 される結合剤、賦形剤、滑沢剤、崩壊剤、湿潤剤などの添加剤を含有する。 また、 経口用液体製剤ば、 通常それらの組成物中に製剤上一般に使用される安定剤、 緩 衝剤、 矯味剤、 保存剤、 芳香剤、 着色剤などの添加剤を含有する。  Oral solid preparations of these dosage forms usually contain additives such as binders, excipients, lubricants, disintegrants, wetting agents and the like which are generally used in pharmaceutical preparations in their compositions. Oral liquid preparations usually contain additives such as stabilizers, buffering agents, flavoring agents, preservatives, fragrances and coloring agents which are generally used in preparations in their compositions.
非経口的に投与する場合は、 注射剤、 坐剤等とすることができる。  When administered parenterally, injections, suppositories and the like can be made.
注射の場合は、 通常単位投与量アンプル又は多投与量容器の状態で提供され、 使用する際に適当な担体、 例えば発熱物質不含の滅菌した水で再溶解させる粉体 であってもよい。 これらの剤形は、 通常それらの組成物中に製剤上一般に使用さ れる乳化剤、 懸濁剤などの添加剤を含有する。 注射手法としては、 例えば点滴静 脈内注射、 静脈内注射、 筋肉内注射、 腹腔内注射、 皮下注射、 皮内注射が挙げら れる。 また、その投与量は、投与対象の年齢、投与経路、投与回数により異なり、 広範囲に変えることができる。  For injections, they will usually be presented in unit dose ampoules or in multi-dose containers, which may be reconstituted in a suitable carrier, eg, sterile pyrogen-free water, for use. These dosage forms usually contain additives, such as emulsifiers and suspending agents, which are generally used in pharmaceutical compositions. Injection techniques include, for example, intravenous infusion, intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, and intradermal injection. The dose varies depending on the age of the subject, the route of administration, and the number of administrations, and can vary widely.
この場合、 本発明の klothoタンパク質、 抗 klothoタンパク質抗体、 klotho夕 ンパク質と FGF23との複合体の有効量と適切な希釈剤及び薬理学的に使 ¾し得る 担体との組合せとして投与される有効量は、 klothoタンパク質、 複合体にあって は、 一回につき体重 lkgあたり 0. 01〜100 g、 好ましくは 0. 5〜20 _ gである。 また、 抗体にあっては、 一回につき体重 1kgあたり 0. l g〜2mg、 好ましくは 1 〜500 /z gである。 スクリーニング系 In this case, an effective amount of a klotho protein, an anti-klotho protein antibody, a complex of klotho protein and FGF23 of the present invention, an appropriate diluent, and an effective amount administered as a combination with a pharmacologically usable carrier. The amount is 0.01 to 100 g, preferably 0.5 to 20 _g / kg body weight at a time for klotho protein or complex. In addition, in the case of an antibody, the dose is 0.1 lg to 2 mg, preferably 1 to 500 / zg per kg of body weight at a time. Screening system
血中リン濃度およぴ Zまたは血中活性型ビタミン D濃度の異常は様々な疾患、 例えば高リン血症、 高 1, 25D血症、 腎性骨異栄養症、 透析骨症、 骨粗鬆症、 低リ ン血症、 クル病、 骨軟化症、 尿細管機能障害、 骨減少症、 低カルシウム血症、 副 甲状腺機能亢進症、.異所性石灰化、 搔痒、 骨硬化症、 パジェット病、 高カルシゥ ム血症、副甲状腺機能低下症、骨痛、筋力低下、骨格変形、成長障害および低 1, 25D 血症など、 で確認されており、 血中リンおよび Zまたは血中活性型ビタミン D濃 度を調節するような物質はこれらのような疾患の治療に有用であると考えられる c 本発明による FGF23と kl othoタンパク質との相互作用を利用した系は上記疾患を 治療する物質(例えばタンパク質、 ペプチド、糖質、脂質、非ペプチド性化合物、 合成化合物、 発酵生産物、 生体物質) の探索に利用できる。 このような物質の探 索の指標としては、 FGF23— klo tho タンパク質相互作用による細胞内シグナルを 検出する系 (例えば細胞内タンパク質のリン酸化、 初期応答遺伝子の活性化、 細 胞内カルシウム、 cAMP , cGMP 等の細胞内情報伝達分子濃度の変化、 細胞膜電位 の変化、 細胞内 pHの変化、 細胞外情報伝達分子の遊離、 細胞の形態変化等) をあ げることができる。 Abnormalities in blood phosphorus and Z or active vitamin D levels can be caused by a variety of disorders, such as hyperphosphatemia, high 1.25D, renal osteodystrophy, dialysis osteopathy, osteoporosis, low Linemia, keratosis, osteomalacia, tubular dysfunction, osteopenia, hypocalcemia, hyperparathyroidism, ectopic calcification, pruritus, osteosclerosis, Paget's disease, hypercalcemia Blood serum and hypoparathyroidism, bone pain, muscle weakness, skeletal deformity, growth disorders and low 1,25D bloodemia, etc., and blood phosphorus and Z or blood active vitamin D levels It is believed that substances that regulate the disease are useful for the treatment of such diseases. C The system using the interaction between FGF23 and klotho protein according to the present invention is a substance that treats the above-mentioned diseases (eg, protein, peptide , Carbohydrates, lipids, non-peptidic compounds, Forming compounds, fermentation products, available to explore the biological material). Indicators for searching for such substances include systems that detect intracellular signals due to FGF23-klotho protein interaction (eg, phosphorylation of intracellular proteins, activation of early response genes, intracellular calcium, cAMP, changes in intracellular signaling molecules such as cGMP, changes in cell membrane potential, changes in intracellular pH, release of extracellular signaling molecules, changes in cell morphology, etc.).
例えば、全長 klothoタンパク質を発現させた細胞系に FGF23を作用させ、細胞 内 ERK リン酸化が亢進することを確認する系を利用することにより、 FGF23 と klo thoタンパク質相互作用を阻害する、 または増強するような物質 (低分子化合 物、 抗体、 など) を探索することが可能である。 また、 可溶化 klothoタンパク質 と FGF23とを様々な細胞系に添加することにより細胞内 E リン酸化が亢進する ことを確認する系を利用することにより FGF23— kl o tho相互作用 (例えば血中リ ン濃度変化もしくは血中活性型ビタミン D濃度変化) を阻害、 または増強するよ うな物質の探索をすることが可能である。  For example, FGF23 acts on a cell line expressing the full-length klotho protein and inhibits or enhances the interaction between FGF23 and klotho protein by using a system that confirms that intracellular ERK phosphorylation is enhanced. It is possible to search for such substances (small molecules, antibodies, etc.). In addition, by using a system that confirms that intracellular E phosphorylation is enhanced by adding solubilized klotho protein and FGF23 to various cell lines, the FGF23-klotho interaction (eg, It is possible to search for a substance that inhibits or enhances concentration changes or changes in blood active vitamin D concentration).
また、 例えば全長 klothoタンパク質を発現させた細胞系に FGF23を作用させ、 In addition, for example, FGF23 is allowed to act on a cell line expressing full-length klotho protein,
EGR1遺伝子の発現が上昇することを確認する系もしくは EGR1 プロモー夕が活性 化することを EGR1 プロモータールシフェラーゼレポーター遺伝子を用いて検出 する系など利用することにより、 FGF23と klo tho夕ンパク質相互作用を阻害する、 または増強するような物質 (低分子化合物、 抗体、 など) を探索することが可能 である。また、可溶化 kl otiioタンパク質と FGF23とを様々な細胞系に添加するこ とにより、 EGR1遺伝子の発現が上昇することを確認する系、 もしくは EGR1 プロ モータが活性化するごとを EGR1 プロモータ一ルシフェラーゼレポ一夕一遺伝子 を用いて検出する系、 を利用することにより FGF23— kl o thoタンパク質相互作用Inhibition of FGF23-klo tho protein interaction by using a system that confirms that EGR1 gene expression increases or a system that uses the EGR1 promoter luciferase reporter gene to detect activation of the EGR1 promoter. Search for substances (small molecules, antibodies, etc.) It is. In addition, a system that confirms that the expression of the EGR1 gene is increased by adding solubilized klotiio protein and FGF23 to various cell lines, or that the EGR1 promoter-luciferase reporter is activated every time the EGR1 promoter is activated. FGF23-klo tho protein interaction by using a detection system that uses the overnight gene
(例えば血中リン濃度変化もしくは血中活性型ビタミン D濃度変化) を阻害、 ま たは増強するような物質の探索をすることが可能である。 It is possible to search for a substance that inhibits or enhances (for example, a change in blood phosphorus level or a change in blood active vitamin D level).
また、 FGF23— kl otho タンパク質相互作用により遺伝子発現変化が引き起こさ れると考えられるビタミン]) 1 α水酸化酵素遺伝子、ビタミン D 2 4位水酸化酵素 遺伝子、 その他 FGF23— kl othoタンパク質相互作用により遺伝子変化が引き起こ されると考えられる遺伝子やそれら遺伝子のプロモー夕を利用したレポーター遺 伝子を利用することにより FGF23— kl o thoタンパク質相互作用 (例えば血中リン 濃度変化もしくは血中活性型ビタミン D濃度変化) を阻害、 または増強するよう な物質の探索をすることが可能である。  In addition, vitamins that are thought to cause gene expression changes by FGF23-klotho protein interaction]) 1α-hydroxylase gene, vitamin D2-4 hydroxylase gene, and other gene changes by FGF23-klotho protein interaction FGF23-klo tho protein interaction (eg, changes in blood phosphorus levels or active vitamin D levels) by using genes that are thought to cause Changes) can be searched for.
本発明は、 klo thoタンパク質を体内に投与して、 血中リン濃度および Zまたは 血中活性型ビタミン D濃度を低下または上昇させる方法、抗 klothoタンパク質抗 体を体内に投与して血中リン濃度およびノまたは血中活性型ビタミン D濃度を上 昇させる方法、 kl o thoタンパク質を体内に投与して FGF23の作用を増強または阻 害する方法、ならびに抗 k 101 hoタンパク質抗体を体内に投与して FGF 23の作用を 阻害する方法も包含する。 さらに、 本発明は血中リン濃度および または血中活 性型ビタミン D濃度を低下または上昇させるための、 あるいは体内での FGF23の 作用を増強または阻害するための医薬組成物製造のための kl otho タンパク質の 使用ならびに血中リン濃度および または血中活性型ビタミン])濃度を上昇させ るための、 あるいは体内での FGF23の作用を阻害するための医薬組成物製造のた めの抗 kl othoタンパク質抗体の使用をも包含する。 本明細書は本願の優先権の基礎である日本国特許出願 2003-137950号の明細書 および/または図面に記載される内容を包含する。 図面の簡単な説明  The present invention relates to a method for decreasing or increasing blood phosphorus concentration and Z or blood active vitamin D concentration by administering klo tho protein into the body, and a method for administering anti-klotho protein antibody to the body to reduce blood phosphorus concentration. A method to increase the concentration of active vitamin D in the blood or blood, a method to enhance or inhibit the action of FGF23 by administering klo tho protein to the body, and a method to increase FGF23 by administering anti-k101 ho protein antibody to the body Methods that inhibit the action of 23 are also included. Furthermore, the present invention relates to klotho for the production of a pharmaceutical composition for lowering or increasing blood phosphorus concentration and / or active vitamin D concentration or for enhancing or inhibiting the action of FGF23 in the body. Anti-klotho protein antibodies for protein use and for the production of pharmaceutical compositions for increasing blood phosphorus and / or blood active vitamin]) levels or for inhibiting the action of FGF23 in the body And the use of This description includes part or all of the contents as disclosed in the description and / or drawings of Japanese Patent Application No. 2003-137950, which is a priority document of the present application. BRIEF DESCRIPTION OF THE FIGURES
図 1は、 ヒト FGF23組換え体を単回投与してから 1時間後、 8時間後でのマウ ス腎臓における EGR1遺伝子の発現変化を RT- PCR法で解析した結果を示す写真で ある。 Figure 1 is a photograph showing the results of RT-PCR analysis of EGR1 gene expression changes in mouse kidney 1 hour and 8 hours after a single administration of human FGF23 recombinant. is there.
図 2は、 ヒト FGF23組換え体を単回投与してから 5分後、 1 0分後でのマウス 腎臓における ERK夕 パク質のリン酸化変化をウエスタンプロッティング法で解 祈した結果を示す写真である。  Figure 2 shows the results of Western blotting of the changes in ERK protein phosphorylation in mouse kidney at 5 and 10 minutes after a single dose of human FGF23 recombinant. It is.
図 3は、 全長 klothoタンパク質発現 PEAK細胞にヒト FGF23組換え体を添加し て 1 0分後の ERKタンパク質のリン酸化変化をウエスタンブロッテイング法で解 祈した結果を示す写真である。  FIG. 3 is a photograph showing the results of Western blotting analysis of changes in ERK protein phosphorylation 10 minutes after the addition of human FGF23 recombinant to PEAK cells expressing full-length klotho protein.
図 4は、 EGR1プロモー夕の下流にルシフェラーゼを結合したレポーター遺伝子 を kl otho 発現プラスミド共にトランスフエクションした PEAK 細胞でのヒト FGF23刺激時のルシフェラ一ゼ活性変化を調べた図である。  FIG. 4 shows changes in luciferase activity during stimulation of human FGF23 in PEAK cells transfected with a reporter gene linked to luciferase downstream of the EGR1 promoter together with a klotho expression plasmid.
図 5は、 PEAK細胞に可溶化マウス klotho タンパク質を安定発現する CHOras clone l細胞の培養上清とヒト FGF23を添加したときの PEAK細胞での ERKのリン 酸化亢進を調べた写真である。  FIG. 5 is a photograph showing the enhancement of ERK phosphorylation in PEAK cells when human FGF23 was added to the culture supernatant of CHOras clone l cells stably expressing soluble mouse klotho protein in PEAK cells.
図 6は、 可溶化マウス kl othoタンパク質一過性発現 PEAK rapid細胞上清サン プルおよびその対照として PEAK ra id細胞の培養上清サンプルを mklotho- 97抗 体、 mklotho- 118抗体、 mklot o-299抗体および mklotho- 941抗体を用いてウェス タンブロット法により解析した結果を示す写真である。  Figure 6 shows the results of the mklotho-97 antibody, mklotho-118 antibody, and mklot o-299, which were obtained by using a solubilized mouse klotho protein transiently expressing PEAK rapid cell supernatant sample and a PEAK raid cell culture supernatant sample as a control. 4 is a photograph showing the results of analysis by Western blotting using an antibody and the mklotho-941 antibody.
図 7中、 図 7 Bは、 可溶化マウス klothoタンパク質一過性発現 PEAK rap id細 胞上清サンプル、 その対照として PEAK rapid細胞の培養上清サンプル、 およびそ れらを FGF- 23固定化レジンに供した時の FGF- 23結合物サンプルを、 mkl otho-118 抗体または mkl otho- 941抗体を用いてウェスタンブロット法により解析した結果 を示す写真である。 図 7 A は同様なサンプルを、 銀染色法により染色した結果を 示す写真である。  In Fig. 7, Fig. 7B shows a sample of the supernatant of the solubilized mouse klotho protein transiently expressing PEAK rapid cells, a sample of the culture supernatant of PEAK rapid cells as a control, and the FGF-23-immobilized resin. 4 is a photograph showing the result of analyzing a sample of an FGF-23-conjugated substance subjected to the above by Western blotting using the mklotho-118 antibody or the mklotho-941 antibody. Figure 7A is a photograph showing the result of a similar sample stained by the silver staining method.
図 8 Aは、 細胞移植後 14日目における可溶化型 klothoタンパク質発現細胞移 植群 (klotho, n=5) および対照 CH0細胞移植群 (cont rol, n=5) の 1, 25ジヒド ロキシビタミン D ( 1, 25D)濃度を示したグラフである。結果は平均値士標準誤差と して表されており、 対照群との有意差検定を Student ' s tにより実施した。  Figure 8A shows 1,25-dihydroxyvitamin of the group transplanted with soluble klotho protein-expressing cells (klotho, n = 5) and the group transplanted with control CH0 cells (control, n = 5) on day 14 after cell transplantation. It is a graph which showed D (1, 25D) density | concentration. The results are expressed as the standard error of the mean, and a significant difference test with the control group was performed by Student's st.
図 8 Bは、 細胞移植後 27日目における可溶化型 kl othoタンパク質発現細胞移 植群 (klo tho, n=6) および対照 CH0細胞移植群 (cont rol, n = 6) の血清リン濃 度ならびに血清カルシウム濃度を示したグラフである。 結果は平均値土標準誤差 として表されており、 対照群との有意差検定を Student's tにより実施した。 図 9Aは、 抗 klothoタンパク質抗体 (P118)群および媒体投与群の投与 24時 間後の血清リン濃度を示した図である。 Figure 8B shows the serum phosphorus concentration of the group transplanted with the soluble klotho protein-expressing cells (klo tho, n = 6) and the group transplanted with the control CH0 cells (cont rol, n = 6) on day 27 after cell transplantation. 3 is a graph showing serum calcium concentrations. Results are mean soil standard error The significance test with the control group was performed by Student's t. FIG. 9A is a graph showing the serum phosphorus concentration of the anti-klotho protein antibody (P118) group and the vehicle administration group 24 hours after administration.
結果は平均値土標準誤差として表されており、 対照群との有意差検定を Student' s tにより'実施した。 The results are expressed as mean soil standard errors, and a significant difference test with the control group was performed by Student's st.
図 9 Bは、 抗 klothoタンパク質抗体(pi 18) 群および媒体投与群の投与 9時間 後の血清活性型ビタミン D濃度を示した図である。  FIG. 9B is a diagram showing serum active vitamin D concentrations 9 hours after administration in the anti-klotho protein antibody (pi 18) group and the vehicle administration group.
結果は平均値士標準誤差として表されており、 対照群との有意差検定を Student' s tにより実施した。 The results are expressed as the standard error of the mean, and a significant difference test with the control group was performed by Student's st.
図 1 0 Aは、 CHO- FGF23H細胞培養上清を SDS-ポリアクリルアミドゲル電気泳動 で分離し、抗 FGF- 23ポリクローナル抗体でウエスタンプロッティングにより組換 え体 FGF- 23タンパク質およびその代謝物を検出した写真である。培養上清中には 全長の FGF- 23Hタンパク質と配列番号 3 5のアミノ酸配列の配列番号 179番目と 180 番目の間で切断されたて生じた N 末側断片と C 末側断片が存在する。 hFGF23- 48抗体および hFGF23- 148抗体で、全長の FGF- 23Hタンパク質と N末側断 片ぺプチドが認識された。 N末側断片は小断片化されたぺプチドの存在が認めら れる。 抗 His6_tag抗体では、 全長タンパク質と C末側断片が検出された。  Figure 10A shows CHO-FGF23H cell culture supernatant separated by SDS-polyacrylamide gel electrophoresis and detection of recombinant FGF-23 protein and its metabolite by Western blotting with anti-FGF-23 polyclonal antibody It is a photograph taken. In the culture supernatant, there are a full-length FGF-23H protein and an N-terminal fragment and a C-terminal fragment generated by cleavage between SEQ ID NO: 179 and SEQ ID NO: 180 of the amino acid sequence of SEQ ID NO: 35. The hFGF23-48 antibody and the hFGF23-148 antibody recognized the full-length FGF-23H protein and the N-terminal fragment peptide. The presence of small fragmented peptides is observed in the N-terminal fragment. For the anti-His6_tag antibody, full-length protein and C-terminal fragment were detected.
図 1 0 Bは、 CHO-FGF23細胞培養上清から精製した FGF- 23全長タンパク質と N 末側断片および C末側断片を SDS-ポリアクリルアミドゲル電気泳動にて分離し、 CBB染色して検出した写真である。  Figure 10B shows that FGF-23 full-length protein purified from CHO-FGF23 cell culture supernatant, N-terminal fragment and C-terminal fragment were separated by SDS-polyacrylamide gel electrophoresis and detected by CBB staining. It is a photograph.
図 1 1は、 EGR1プロモータの下流にルシフェラーゼを結合したレポーター遺伝 子を klotho 発現プラスミド共にトランスフエクシヨンした PEAK細胞でのヒト FGF- 23刺激時に、 抗 FGF- 23抗体 ( 2 C3B) を添加した時のルシフェラーゼ活性変 化を調べた図である。 FGF- 23刺激により上昇するルシフェラーゼ活性が 2 C3B同 時添加により抑制されることが示された。  Figure 11 shows the results obtained when the anti-FGF-23 antibody (2C3B) was added during stimulation of human FGF-23 in PEAK cells transfected with a klotho expression plasmid and a reporter gene linked to luciferase downstream of the EGR1 promoter. FIG. 3 is a diagram showing a change in luciferase activity of the present invention. It was shown that luciferase activity increased by FGF-23 stimulation was suppressed by simultaneous addition of 2C3B.
図 1 2は、 CHOras clonel細胞に可溶化 klothoタンパク質発現 CHOras clonel 細胞の培養上清と FGF23を添加したときに引き起こされる ERKのリン酸化亢進を、 抗 FGF23抗体(2C3B) が抑制するかどうかを調べた結果を示す写真である。 FGF23 と可溶化 klothoタンパク質発現 CHOras clonel細胞培養上清を同時添加したとき の ERKリン酸化亢進は、 2C3B抗体濃度依存的に抑制されることが示された。 図 1 3は、 2C3B抗体、 抗 TPO抗体、 および媒体を投与後、 24時間目における血 清中リン酸、 血清中 1, 25-ジヒドロキシビタミン D濃度を示した図である。 Figure 12 shows whether anti-FGF23 antibody (2C3B) suppresses the increase in ERK phosphorylation caused by the addition of FGF23 and the culture supernatant of CHOras clonel cells, which express soluble klotho protein in CHOras clonel cells. 4 is a photograph showing the results of the measurement. It was shown that the enhancement of ERK phosphorylation when FGF23 and solubilized klotho protein-expressing CHOras clonel cell culture supernatant were added simultaneously was suppressed in a 2C3B antibody concentration-dependent manner. FIG. 13 is a diagram showing the concentrations of phosphoric acid in serum and 1,25-dihydroxyvitamin D in serum 24 hours after administration of the 2C3B antibody, anti-TPO antibody, and vehicle.
図 1 4は、 精製した可溶化 klothoを SDS-PAGE後銀染色および抗 klo tho抗体 (mklotho- 118抗体) にて検出した写真である。  FIG. 14 is a photograph showing the purified solubilized klotho after SDS-PAGE and silver staining and detection with an anti-klotho antibody (mklotho-118 antibody).
図 1 5は、 mskl抗体- 1および mskl抗体- 2にて可溶化 klothoタンパク質をゥ エスタンプロッティングで検出した写真である。  FIG. 15 is a photograph in which solubilized klotho protein was detected by mastl blotting using mskl antibody-1 and mskl antibody-2.
図 1 6は、 抗マウス klothoモノクローナル抗体を用いて精製可溶化] dothoを 免疫沈降し、沈降された可溶化 klothoを fflklo tho- 118抗体にてウエスタンブロッ ティングにて検出した写真である。  FIG. 16 is a photograph showing immunoprecipitation of purified solubilized dolot using an anti-mouse klotho monoclonal antibody, and detection of the precipitated solubilized klotho by Western blotting using the fflklotho-118 antibody.
図 1 7 Aは、 EGR1アツセィ系における mskl抗体- 1の FGF23作用に対する IS害 効果を調べた図である。  FIG. 17A is a graph showing the effect of mskl antibody-1 on the FGF23 action in the EGR1 Atsushi strain on IS activity.
図 1 7 Bは、 EGR1アツセィ系における mskl抗体- 2の FGF23作用に対する阻害 効果を調べた図である。  FIG. 17B is a diagram illustrating the inhibitory effect of the mskl antibody-2 on the FGF23 action in the EGR1 Atsushi system.
図 1 8 Aは、 mskl抗体- 1、 mskl抗体 _2のマウスへの投与による血清ジヒドロ キシビタミン Dに対する効果を調べた図である。  FIG. 18A shows the effect of administration of mskl antibody-1 and mskl antibody_2 to mice on serum dihydroxyvitamin D.
図 1 8 Bは、 mskl抗体- 1、 mskl抗体- 2のマウスへの投与による血清リン濃度 に対する効果を調べた図である。  FIG. 18B shows the effect of administration of mskl antibody-1 and mskl antibody-2 to mice on serum phosphorus concentration.
図 1 9は、 EGR1アツセィ系における抗 klo thoモノクローナル抗体の FGF23作 用に対する阻害効果を調べた図である。  FIG. 19 is a graph showing the effect of the anti-klotho monoclonal antibody on FGF23 action in the EGR1 Atsushi strain.
図 2 O Aは、抗 klothoモノクローナル抗体のマウスへの投与による血清ジヒド ロキシビタミン D濃度に対する効果を調べた図である。  FIG. 2 OA shows the effect of administration of an anti-klotho monoclonal antibody to mice on serum dihydroxyvitamin D concentration.
図 2 0 Bは、抗 klothoモノクローナル抗体のマウスへの投与による血清リン濃 度に対する効果を調べた図である。  FIG. 20B shows the effect of administration of an anti-klotho monoclonal antibody to mice on serum phosphorus concentration.
図 2 1は、 精製可溶化 klothoと FGF23の添加による HEK293細胞の ERKリン酸 化亢進に対する効果を調べた写真である。 '  FIG. 21 is a photograph showing the effect of adding purified solubilized klotho and FGF23 on the enhancement of ERK phosphorylation in HEK293 cells. '
図 2 2は、精製可溶化 klothoと FGF23の添加による MC3T3細胞の細胞形態変化 に対する効果を調べた写真である。  FIG. 22 is a photograph showing the effect of the addition of purified solubilized klotho and FGF23 on the cell morphology of MC3T3 cells.
図 2 3は、精製可溶化 klothoをマウスへ投与したときの血清ジヒドロキシビ夕 ミン D濃度に対する効果を調べた図である。 図 2 4 Aは、 HEK293細胞に発現させた klothoと FGF23 との細胞上での結合を FACSにて検出した図である。 FIG. 23 shows the effect of administering purified solubilized klotho to mice on serum dihydroxybiminin D concentration. FIG. 24A is a diagram in which binding between klotho expressed in HEK293 cells and FGF23 on cells was detected by FACS.
図 2 4 Bは、 細胞に klothoを発現させていないコントロールの結果を示す。  FIG. 24B shows the results of a control in which cells do not express klotho.
発明を実施するための最良の形態 BEST MODE FOR CARRYING OUT THE INVENTION
以下、 実施例により本発明をさらに具体的に説明する。 但し、 本発明はこれら 実施例にその技術的範囲が限定されるものではない。  Hereinafter, the present invention will be described more specifically with reference to examples. However, the technical scope of the present invention is not limited to these examples.
〔実施例 1〕 FGF23のマウスへの投与による腎臓での egrl発現上昇作用の確認 ヒ卜 FGF23のマウスへの投与時に腎臓で発現変動する遺伝子を探索する為にメ ンブレンアレイ At las mouse 1· 2k、 At las mouse 1. 2kII (米国 クロンテック社) を用いて採索を行なった。 メンブレンアレイは添付のプロトコールに従い行なつ た。 使用したサンプルはヒト FGF23 (5 / g/head)投与 1 時間後マウス腎臓、 ヒト [Example 1] Confirmation of the effect of increasing the expression of egrl in the kidney by administration of FGF23 to mice Human membrane array Atlas mouse 1.2k to search for genes whose expression fluctuates in the kidney when FGF23 is administered to mice , Atlas mouse 1.2kII (Clontech, USA). The membrane array was prepared according to the attached protocol. The samples used were human kidney, human kidney 1 hour after administration of FGF23 (5 / g / head), human
FGF23投与 8時間後マウス腎臓、 媒体投与マウス腎臓。 組み換えヒト FGF23の作 製および精製は島田らの方法 {Endocrinology 143, 3179 (2002) } に従って行つ た。 8 hours after administration of FGF23, mouse kidney, vehicle-administered mouse kidney. Production and purification of recombinant human FGF23 was performed according to the method of Shimada et al {Endocrinology 143, 3179 (2002)}.
マウスは BALB/c (雄 、 6週齢) を使用して各 n=2で行なった。 その結果、 ヒ ト FGF23投与後 1時間で発現が上昇する遺伝子として EGR1が見出された。  Mice were performed using BALB / c (male, 6 weeks old) at each n = 2. As a result, EGR1 was found as a gene whose expression increased one hour after administration of human FGF23.
この現象を確認するため、上述の 3種のマウス腎臓総 RNAを用いて First s trand cDNAを作製し、 RT-PCRを行なった。 Fi rs t s trand cDNAの合成は First s trand cDNA synthes is ki t (米国 インビトロゲン社) を用いて添付のプロトコールにしたが つて行なった。 Fi rst strand cDNA作製にはヒト FGF23投与 1時間後マウス腎臓、 ヒト FGF23投与 8時間後マウス腎臓、 媒体投与マウス腎臓より抽出した総 RNAを 用いた。 合成した。  In order to confirm this phenomenon, First strand cDNA was prepared using the above-mentioned three kinds of mouse kidney total RNA, and RT-PCR was performed. First strand cDNA was synthesized using First strand cDNA synthes is kit (Invitrogen, USA) according to the attached protocol. First strand cDNA was prepared using total RNA extracted from mouse kidney 1 hour after administration of human FGF23, mouse kidney 8 hours after administration of human FGF23, and mouse kidney after administration of vehicle. Synthesized.
First s trand cMAを鍀型に、 下記に示すマウス EGR1特異的なプライマー (配 列番号 4および 5 ) を用いて 94度 20秒、 68度 1分からなる工程を 1サイクルと して 25サイクルの PCRを実施した。  Using the first strand cMA as type I, and using the mouse EGR1-specific primers shown below (SEQ ID NOs: 4 and 5), 25 cycles of a cycle consisting of 94 ° C for 20 seconds and 68 ° C for 1 minute were performed for 25 cycles. Was carried out.
マウス EGR1 RT- PCR用 FWプライマー: CTTAATACCACCTACCAATCCCAGC (配列番号 4 ) マウス EGR1 RT- PCR用 RVプライマー: GTTGAGGTGCTGAAGGAGCTGCTGA (配列番号 5 ) 図 1に示したように、 これら PCR産物を 2 %ァガロースゲルにて電気泳動した ところヒト FGF23投与後 1時間のマウス腎臓でのみ EGR1の発現上昇が確認された。 一方、 対象とした G3PDHプライマーを用いた場合ではどのサンプルも同程度の発 現が認められた。 この結果から、 FGF23投与後 1時間でマウス腎臓において EGR1 の発現上昇することが確認された。 FW primer for mouse EGR1 RT-PCR: CTTAATACCACCTACCAATCCCAGC (SEQ ID NO: 4) RV primer for mouse EGR1 RT-PCR: GTTGAGGTGCTGAAGGAGCTGCTGA (SEQ ID NO: 5) As shown in FIG. 1, these PCR products were electrophoresed on a 2% agarose gel. However, an increase in EGR1 expression was confirmed only in the mouse kidney one hour after administration of human FGF23. On the other hand, when the target G3PDH primer was used, the same level of expression was observed in all samples. From these results, it was confirmed that 1 hour after the administration of FGF23, EGR1 expression was increased in mouse kidney.
〔実施例 2〕 FGF23のラッ卜への投与による腎臓での ERKリン酸化亢進  [Example 2] Administration of FGF23 to rats enhances ERK phosphorylation in kidney
EGR1発現上昇は MAPキナーゼカスケ一ドの活性化により引き起こされることが 知られている。 そこで、 ヒト FGF23投与時のラット腎臓における ERKリン酸化亢 進が引き起こされるかどうかを調べた。 ラット (7週齢、 S D , 雄) にヒト FGF23 を 5 g/head投与し、 5分後、 10分後の腎臓を摘出。 腎臓皮質部のみをかみそり にて切断した。 切断したラット腎臓皮質部は液体窒素にて凍結した。 凍結した腎 臓は抽出バッファー (20mM Hepes (pH7. 5) , 150πιΜ NaCl , 1¾ NP-40 , lmM EDTA , 0. 1% SDS , 50niM sodium f luoride , 2mM sodium Vanadate , 1 tab. /7ml COMPLETE (米 国 ロシュ社)) にてポリトロンホモジナイザーにてホモジナイズを行なった。 2 時間氷上で静置後 1000Gにて 1 0分間遠心を行ない、 上清画分をその後の実験に 用いた。上清画分のタンパク質濃度を BCAキット(米国 PIERCE社)にて測定し、 全てのサンプルを 22. 7mg/mlのタンパク質濃度に抽出バッファーで調製した。 こ のサンプルを抗リン酸化 ERK抗体 (米国 サンタクルーズ社) を用いてウェス夕 ンブロッテイングを行なった。 図 2に示した様に、 ヒト FGF23を投与したラット では、 5分後、 1 0分後ともに ERKリン酸化が宂進していることが確認された。 〔実施例 3〕 マウス klotho遺伝子のクローン化  It is known that elevated EGR1 expression is caused by activation of the MAP kinase cascade. Therefore, it was examined whether administration of human FGF23 would cause enhanced phosphorylation of ERK in rat kidney. Rats (7 weeks old, SD, male) were administered human FGF23 at a dose of 5 g / head, and the kidneys were removed 5 minutes and 10 minutes later. Only the kidney cortex was cut with a razor. The cut rat kidney cortex was frozen in liquid nitrogen. The frozen kidney was extracted with extraction buffer (20 mM Hepes (pH 7.5), 150πιπ NaCl, 1 NP-40, lmM EDTA, 0.1% SDS, 50niM sodium fluoride, 2mM sodium Vanadate, 1 tab./7ml COMPLETE (US (Roche Co., Ltd.)) and homogenized with a Polytron homogenizer. After standing on ice for 2 hours, the mixture was centrifuged at 1000 G for 10 minutes, and the supernatant fraction was used for subsequent experiments. The protein concentration of the supernatant fraction was measured with a BCA kit (PIERCE, USA), and all samples were prepared with an extraction buffer to a protein concentration of 22.7 mg / ml. This sample was subjected to Wes evening blotting using an anti-phosphorylated ERK antibody (Santa Cruz, USA). As shown in FIG. 2, in the rats to which human FGF23 was administered, it was confirmed that ERK phosphorylation was enhanced after 5 minutes and 10 minutes. [Example 3] Cloning of mouse klotho gene
マウス kl 01 ho遺伝子の全長 cDNAクローニングは PCR法にて行なった。 GeneBank のデータ (Accession No. AB005141) を元に以下のプライマーを合成した。  The full-length cDNA cloning of the mouse kl 01 ho gene was performed by PCR. The following primers were synthesized based on GeneBank data (Accession No. AB005141).
マウス klotho遺伝子クローニング FWプライマー: Mouse klotho gene cloning FW primer:
CCCGAATTCGCAGCATGCTAGCCCGCGCCCCTCCTC (配列番号 6 )  CCCGAATTCGCAGCATGCTAGCCCGCGCCCCTCCTC (SEQ ID NO: 6)
マウス klotho遺伝子クローニング RV1プライマー: Mouse klotho gene cloning RV1 primers:
ATTTGCGGCCGCTTACTTATAACTTCTCTGGCCTTTCTT (配列番号 7 )  ATTTGCGGCCGCTTACTTATAACTTCTCTGGCCTTTCTT (SEQ ID NO: 7)
配列番号 6のプライマーはマウス klotho遺伝子の 5'側のプライマー i?、 開始 コドンとその上流 5塩基のコザック配列部分を含む。 さらに EcoRIサイ卜が付加 されている。  The primer of SEQ ID NO: 6 contains a primer i? On the 5 'side of the mouse klotho gene, an initiation codon and a Kozak sequence portion of 5 bases upstream thereof. In addition, an EcoRI site has been added.
配列番号 7のプライマ一はマウス klotho遺伝子の 3'側のプライマーで、 終始 コドンを含み、 さらに Not Iサイトが付加されている。 錶型にはマウス腎臓 cDNA を使用した。マウス腎臓 cDNAの作製は実施例 1の方法にしたがつて行なつた。 PCR 反応は LATaQwithGCBuifer (日本国 夕カラ社) を使用した。すなわち、 TaKaRaThe primer of SEQ ID NO: 7 is a primer on the 3 'side of the mouse klotho gene, which contains a termination codon and a Not I site. Mouse kidney cDNA for type 錶 It was used. Preparation of mouse kidney cDNA was performed according to the method of Example 1. The PCR reaction used LATaQwithGCBuifer (Yukara, Japan). That is, TaKaRa
La Taq polymerase 0.5/^1 , 2XGC buffer 25 1 , dNTP Mix 8 1 (2.5mM) , 配列 番号 6のプライマー (1 / M)、 配列番号 7のプライマー (1 M)、 上述のマウス 腎臓 cDNAl U、 水を最終的に 50 \になるように加え、 以下の条件で PCR反応 を行なった。 一次変性は 94°Cで 5分、 次に 94 20秒、 55でで 30秒、 7La Taq polymerase 0.5 / ^ 1, 2XGC buffer 25 1, dNTP Mix 81 (2.5 mM), primer of SEQ ID NO: 6 (1 / M), primer of SEQ ID NO: 7 (1 M), mouse kidney cDNA Water was added to a final volume of 50 \, and PCR was performed under the following conditions. Primary denaturation at 94 ° C for 5 minutes, then 94 20 seconds, 55 at 30 seconds, 7
2 °Cで 4分を 1サイクルとして 35サイクル行い、 最後に伸長反応を 72°Cで 7 分行った。 PCRによる増幅産物は 0. 8 %ァガロース電気泳動にて確認した結果、 約 3kbp付近に単一バンドが確認されたため、ゲルからバンド部分の切り出しを行 ない MinElute Gel extraction kit (ドイツ国 キアゲン社) を用いてゲルから の精製を行なった。精製したマウス klotho cDNA断片を EcoRI、NotIにて切断し、One cycle was performed at 2 ° C for 4 minutes for 35 cycles, and finally the extension reaction was performed at 72 ° C for 7 minutes. The amplified product by PCR was confirmed by 0.8% agarose electrophoresis. As a result, a single band was detected at about 3 kbp. The band was cut out from the gel, and a MinElute Gel extraction kit (Qiagen, Germany) was used. Was used to purify the gel. Cut the purified mouse klotho cDNA fragment with EcoRI and NotI,
EcoRI.NotI切断した PEAK8ベクター(米国 エッジバイオシステム社)にライゲ ーシヨン反心を行なった。 ライケーシヨン反応は TaKaRa Ligation Kit versionLigation was performed on the PEAK8 vector (Edge Biosystems, USA) cut with EcoRI.NotI. The reaction is TaKaRa Ligation Kit version
2(日本国 夕カラ社)を用い、添付のプロトコ一ルに準じて行なった。 クローン化 されたマウス k 101 ho cDNAの塩基配列を確認する為にマウス k 101 ho遺伝子配列の2 (Yukara, Japan) was performed according to the attached protocol. In order to confirm the nucleotide sequence of the cloned mouse k101ho cDNA,
GeneBankデ一夕 (Accession No. AB005141) を基に以下のマウス klotho遺伝子 配列確認用プライマーを合成した。 Based on GeneBank Data (Accession No. AB005141), the following primers for mouse klotho gene sequence confirmation were synthesized.
ACTACCGCTTCTCCATATCGTG (配列番号 8 )  ACTACCGCTTCTCCATATCGTG (SEQ ID NO: 8)
TCGCCTTAAGCTCCCATTGGAT (配列番号 9 )  TCGCCTTAAGCTCCCATTGGAT (SEQ ID NO: 9)
ATGGGGTCGACGTCATTGGGTACA (配列番号 10 )  ATGGGGTCGACGTCATTGGGTACA (SEQ ID NO: 10)
TCTTGCCTCTGGGTAACCAGAC (配列番号 1 1 )  TCTTGCCTCTGGGTAACCAGAC (SEQ ID NO: 11)
CTTGCAGGCTGACTGGATAGA (配列番号 12 )  CTTGCAGGCTGACTGGATAGA (SEQ ID NO: 12)
AGCAAGACTCACTGAGGATCTA (配列番号 13 )  AGCAAGACTCACTGAGGATCTA (SEQ ID NO: 13)
CACGATATGGAGAAGCGGTAGT (配列番号 14 )  CACGATATGGAGAAGCGGTAGT (SEQ ID NO: 14)
ATCCAATGGGAGCTTAAGGCGA (配列番号 15)  ATCCAATGGGAGCTTAAGGCGA (SEQ ID NO: 15)
TGTACCCAATGACGTCGACCCCAT (配列番号 16)  TGTACCCAATGACGTCGACCCCAT (SEQ ID NO: 16)
GTCTGGTTACCCAGAGGCAAGA (配列番号 17 )  GTCTGGTTACCCAGAGGCAAGA (SEQ ID NO: 17)
TCTATCCAGTCAGCCTGCAAG (配列番号 18 )  TCTATCCAGTCAGCCTGCAAG (SEQ ID NO: 18)
TAGATCCTCAGTGAGTCTTGCT (配列番号 19 )  TAGATCCTCAGTGAGTCTTGCT (SEQ ID NO: 19)
これらのプライマーを用いクローン化されたマウス klotho cDNAの塩基配列解 析を行なった結果、 マウス klotho遺伝子と完全に一致し、 マウス klotho遺伝子 のクローン化を完了した。 以下このプラスミドを mklotho/pEAK8と記す。 また、 さらに mklotho/pEAK8 'ベクターを EcoRI、 Notlで切断して得られたマウス klotho cDNAを、 プラスミド pEAK8に分子内リボゾームエントリー部位 (IRES) および増 強型緑色蛍光タンパク質 (EGFP) を連結することにより作製したベクター IRES-EGFP-PEAK8 の EcoRI 、 Notl 部位 に 挿 入 す る こ と に よ っ て mklotho/IRES-EGFP-pEAK8を得た。 Nucleotide sequence analysis of mouse klotho cDNA cloned using these primers As a result of the analysis, it completely matched the mouse klotho gene, and the cloning of the mouse klotho gene was completed. Hereinafter, this plasmid is referred to as mklotho / pEAK8. In addition, mouse klotho cDNA obtained by digesting the mklotho / pEAK8 'vector with EcoRI and Notl is ligated to plasmid pEAK8 by linking an intramolecular ribosome entry site (IRES) and enhanced green fluorescent protein (EGFP). Mklotho / IRES-EGFP-pEAK8 was obtained by inserting it into EcoRI and Notl sites of the prepared vector IRES-EGFP-PEAK8.
〔実施例 4〕 マウス可溶化 klothoタンパク質をコードする遺伝子のクローン化 実施例 3で作製したマウス kl 01 ho遺伝子を铸型に可溶化 kl o t hoタンパク質を作 製した。 作製の為に以下のプライマーを合成した。  [Example 4] Cloning of gene encoding mouse-solubilized klotho protein The mouse kl01ho gene prepared in Example 3 was solubilized into type II to produce a klotho protein. The following primers were synthesized for production.
マウス可溶化 klothoタンパク質コード遺伝子クローニング用 RV3プライマー:RV3 primer for cloning mouse-solubilized klotho protein-encoding gene:
ATTTGCGGCCGCTTAGGTTTGAAAAAATCCACATTCGGTGCA (配列番号 2 0 ) ATTTGCGGCCGCTTAGGTTTGAAAAAATCCACATTCGGTGCA (SEQ ID NO: 20)
配列番号 20のプライマーはマウス klotho遺伝子の 2940塩基までを含み、 TAA 終始コドンを付加し、 さらに Notlサイトを付加している。 PCR反応は配列番号 8 と配列番号 2 0のプライマーと Pyrobest Polymerase (日本国 夕カラ社)を用い て行なった。すなわち、 TaKaRa Pyrobest polymerase 0.5 10X Pyrobest Buffer The primer of SEQ ID NO: 20 contains up to 2940 bases of the mouse klotho gene, adds a TAA termination codon, and further adds a Notl site. The PCR reaction was carried out using the primers of SEQ ID NO: 8 and SEQ ID NO: 20 and Pyrobest Polymerase (Yukara, Japan). That is, TaKaRa Pyrobest polymerase 0.5 10X Pyrobest Buffer
5 1、 dNTPMi 4 l (1.25mM) , 配列番号 8プライマー 1 M、 配列番号 2 0プライ マ一 1 M、 マウス klotho cDNA 10ng、 を加え最終液量 5 0 1になるように水を 加えた。 PCR反応は以下の通りに行なった。一次変性は 94 °Cで 5分、'次に 94°C51, dNTPMi 4 l (1.25 mM), SEQ ID NO: 8 primer 1 M, SEQ ID NO: 20 primer 1 M, mouse klotho cDNA 10 ng were added, and water was added to a final volume of 501. The PCR reaction was performed as follows. Primary denaturation at 94 ° C for 5 minutes, then at 94 ° C
2 0秒、 55 °Cで 3 0秒、 7 2でで 4分を 1サイクルとして 3 5サイクル行い、 最後に伸長反応を 7 2°Cで 7分行った。 PCR による増幅産物は 0. 8 %ァガロー ス電気泳動にて確認した結果、約 1.7kbp付近に単一バンドが確認されたため、ゲ ルからパンド部分の切り出しを行ない MinElute Gel extraction kit (ドイツ国 キアゲン社) を用いてゲルからの精製を行なった。 精製したマウス klotho cDNA 断片を Kpnl、 Notlにて切断し、 実施例 3で作製したマウス klotho-PEAK8ベクタOne cycle was 20 seconds, 30 seconds at 55 ° C, and 4 minutes at 72, and 35 cycles were performed. Finally, the extension reaction was performed at 72 ° C for 7 minutes. The amplified product by PCR was confirmed by 0.8% agarose gel electrophoresis.As a result, a single band was observed at about 1.7 kbp.Pand was cut out from the gel, and MinElute Gel extraction kit (Qiagen, Germany) was used. ) Was used for purification from the gel. The purified mouse klotho cDNA fragment was digested with Kpnl and Notl, and the mouse klotho-PEAK8 vector prepared in Example 3 was cut.
—を KpnI,NotI切断したものににライゲ一シヨン反応を行なった。 塩基 SB列の確 認は実施例 3に準じて行ない、可溶化マウス klothoタンパク質の作製を完了した。 以下このプラスミドを soluble-mldotho/pEAK8と記す。 この可溶化マウス klotho タンパク質はアミノ酸番号 1から 980までをコードする。 さらに、 得られたマウ ス可溶化 klotho cDNAを EcoRI, Notlで切断し、 プラスミド pEAK8 (米国 エッジ バイオシステム社) に分子内リボゾームエントリー部位 (IRES) および増強型緑 色蛍光タンパク質 ( EGFP ) を連結する ことによ り作製したベクター I ES-EGFP-PEAK8の EcoRI. Not I部位に挿入することによって solubl e- mklotho/ IRES-EGFP-PEAK8を得た。 -Was digested with KpnI and NotI, and subjected to a ligation reaction. Confirmation of the base SB sequence was performed according to Example 3, and the production of the solubilized mouse klotho protein was completed. Hereinafter, this plasmid is referred to as soluble-mldotho / pEAK8. This solubilized mouse klotho protein encodes amino acids 1 to 980. Furthermore, the obtained mouse solubilized klotho cDNA was digested with EcoRI and Notl, and plasmid pEAK8 (US Edge) BioSystems Inc.) into the EcoRI.NotI site of the vector IES-EGFP-PEAK8, which was created by ligating an intramolecular ribosome entry site (IRES) and enhanced green fluorescent protein (EGFP). A solubl e-mklotho / IRES-EGFP-PEAK8 was obtained.
〔実施例 5〕 マウス可溶化 klothoタンパク質安定発現細胞の作製  [Example 5] Production of mouse solubilized klotho protein stably expressing cells
Trans IT LT1 (米国 MIRUS社) を用い、 添付文書にしたがって CHOras c lone l 細胞に solubl e-mklotho/ IRES- EGFP- pEAK8プラスミドを導入した。 5 g/ml ピュ 一口マイシン、 1 0 %FBS を含む ΜΕΜ α培地にて薬剤耐性細胞を選択し、 FACS vantage (米国 べクトンディッキンソン社) によって EGFP (Green Fluorescent Prote in)の発光強度が強い細胞をソ一ティングによりクローン化した。 クローン 化した細胞がコンフルェントになったところで、 血清を含まない DF (DMEM/F-12) 培地に置換し 2日後に上清を回収した。回収した上清を抗 klothoタンパク質抗体 Using Trans IT LT1 (MIRUS, USA), the plasmid-mklotho / IRES-EGFP-pEAK8 plasmid was introduced into CHOras clone cells according to the package insert. G Select drug-resistant cells in α medium containing 5 g / ml pure bitemycin and 10% FBS, and use FACS vantage (Becton Dickinson, USA) to select cells with strong EGFP (Green Fluorescent Protein) emission. Cloned by sorting. When the cloned cells became confluent, the medium was replaced with serum-free DF (DMEM / F-12) medium, and the supernatant was recovered 2 days later. Collect the supernatant using anti-klotho protein antibody
(米国 ALPHA DIAGNOSTIC 社) を用いてウエスタンブロッテイングを行ない、 130kDa付近の klothoタンパク質のバンドの強いクローンを選択した。 Western blotting was performed using ALPHA DIAGNOSTIC (USA) to select clones with a strong klotho protein band around 130 kDa.
〔実施例 6〕 マウス全長 klothoタンパク質の一過性発現による FGF23刺激での ERKリン酸化亢進  [Example 6] Transient expression of mouse full-length klotho protein enhances ERK phosphorylation upon FGF23 stimulation
ERKリン酸化亢進確認 Confirmation of enhanced ERK phosphorylation
実施例 3で作製したマウス全長 klotho タンパク質発現プラスミドを用いてマ ウス全長 klothoタンパク質を PEAKrapid細胞 (米国 エッジバイオシステム社) に一過性に発現させて FGF23で刺激を加えたときの細胞内 ERKリン酸化の亢進を 検証した。 1 2ゥエルプレートに PEAKrapid細胞に添付のプロトコールに従い実 施例 3で作製した mklotho/pEAK8プラスミドを一過性にトランスフエク卜した。 トランスフエクト 2 4時間後に血清を含まない DMEM培地に培地交換し、さらに 2 4時間(;02ィンキュベ一夕一内でィンキュベートを行なった。へパリン(米国 シ ダマ社) を 1 O g /mlになるように添加した後に、 FGF23、 bFGF、 媒体を添加し た。 1 0分 3 7 °Cでインキュベートを行なった後に上清を吸引し PBSで洗浄を行 なった後 SDS-PAGEサンプルバッファ一(日本国 第一化学社)にて細胞を溶解し た。このサンプルについて実施例 2と同様にウエスタンブロッテイングを行った。 図 3に示すようにマウス全長 klothoタンパク質発現 PEAKrapid細胞のみで FGF23 による ERKリン酸化亢進が確認された。 〔実施例 7〕 マウス全長 klothoタンパク質の一過性発現による FGF23刺激での egr l発現上昇の確認 The mouse full-length klotho protein was transiently expressed in PEAKrapid cells (Edge Biosystems, USA) using the mouse full-length klotho protein expression plasmid prepared in Example 3, and the intracellular ERK phosphorus was stimulated with FGF23. The enhanced oxidation was verified. The mklotho / pEAK8 plasmid prepared in Example 3 was transiently transfected into a 12-well plate according to the protocol attached to the PEAKrapid cells. Trans Hue transfected 2 4 hours later the medium was replaced with DMEM medium without serum, additional 2 4 h (;. 0 2 Inkyube Isseki was performed Inkyubeto within one heparin (U.S. sheet Damasha) the 1 O g / ml Then, FGF23, bFGF, and medium were added for 10. After incubation at 37 ° C for 10 minutes, the supernatant was aspirated, washed with PBS, and then washed with SDS-PAGE sample buffer. The cells were lysed at (Daiichi Kagaku, Japan) and Western blotting was performed on this sample in the same manner as in Example 2. As shown in Fig. 3, EGF by FGF23 was used only in mouse full-length klotho protein-expressing PEAKrapid cells. Enhanced phosphorylation was confirmed. [Example 7] Confirmation of increase in egrl expression by FGF23 stimulation by transient expression of mouse full-length klotho protein
マウス全長 kl othoタンパク質を PEAKrapid細胞(米国 エッジバイオシステム 社)へ一過性発現させたときの FGF23刺激による EGR1の発現上昇を検証した。 PEAK rap id 細胞へ実施例 6と同様にトランスフエクシヨンを行なった。 トランスフエ クシヨン 4 8時間後にへパリン 1 0 g/mlになるように加え、さらにヒト FGF23 The increase in EGR1 expression induced by FGF23 stimulation when the full-length mouse klotho protein was transiently expressed in PEAKrapid cells (Edge Biosystems, USA) was examined. Transfection was performed on PEAK rap id cells in the same manner as in Example 6. Transfection 48 8 hours later, add heparin to 10 g / ml, and add human FGF23
( 1 0 O ng/ml ) にて刺激を加えた。 刺激 3 0分後に細胞を PBS で洗浄を行い IS0GEN (日本国 二ツボンジーン社) を用いて懸濁させた。 添付文書に従いトー タル RNAを調製し、実施例 1と同様に RT- PCRを行なった。実施例 1の結果と同様 にマウス全長 klothoタンパク質発現 PEAKrapid細胞では FGF23 lOOng/ml刺激で EGR1の有意な発現上昇が確認された。 (10 O ng / ml). Thirty minutes after the stimulation, the cells were washed with PBS and suspended using IS0GEN (Nitsubon Gene, Japan). Total RNA was prepared according to the package insert, and RT-PCR was performed in the same manner as in Example 1. Similar to the results of Example 1, in PEAKrapid cells expressing mouse full-length klotho protein, a significant increase in EGR1 expression was confirmed upon stimulation with FGF23 lOOng / ml.
〔実施例 8〕 マウス可溶化 kl 0 tho夕ンパク質発現細胞上清と FGF23添加による CHOras clone l細胞での ERKリン酸化亢進確認  [Example 8] Confirmation of enhanced ERK phosphorylation in CHOras clone l cells by addition of mouse solubilized kl 0 tho protein-expressing cell supernatant and FGF23
8- (1) マウス可溶化 klothoタンパク質発現細胞の培養上清の調製 8- (1) Preparation of culture supernatant of mouse solubilized klotho protein-expressing cells
実施例 5で作製したマウス可溶化 klotho夕ンパク質発現 CHOras clone l細胞を 組織培養用フラスコ (225cm2、 米国 コーニング社) に播種した。 2 4時間培養 後血清を含まない ΜΕΜ α培地に置換し、 5日間 3 7でにて培養し、 その培養上清 を回収した。 The mouse solubilized klotho protein-expressing CHOras clonel cells prepared in Example 5 were seeded in a tissue culture flask (225 cm 2 , Corning, USA). After culturing for 24 hours, the medium was replaced with a serum-free ΜΕΜα medium, and the cells were cultured at 37 for 5 days, and the culture supernatant was collected.
8 -(2)マウス可溶化 klotho タンパク質発現細胞の培養上清と FGF23 添加による CHOras clonel細胞での ERKリン酸化亢進確認  8-(2) Enhanced ERK phosphorylation in CHOras clonel cells by adding FGF23 and culture supernatant of mouse solubilized klotho protein-expressing cells
CHOras c l one l細胞、 および HEK293細胞を 24ゥエルプレート (米国 コ一二 ング社) に播種した。 2 4時間培養後、 8- (1)で調製したマウス可溶化 klotho夕 ンパク質発現 CHOras c l onel細胞培養上清に置換しさらに 4時間培養を行なった。 続いてへパリン 10 g/ml を添加したのちにヒト FGF23 ( 100ng/ml) を添加した。 1 0分 3 7ででィンキュベ一トを行なった後に上清を吸引し PBSで洗浄を行なつ た後 SDS- PAGEサンプルバッファー (日本国 第一化学社) にて細胞を溶解した。 このサンプルについて実施例 2と同様に抗 ERK抗体 (米国 サンタクルーズ社) を用いてウェスタンプロッティングを行った。 図 5に示すようにマウス可溶化 klothoタンパク質を添加した細胞のみで FGF23による ERKリン酸化亢進が確認さ れた。 〔実施例 9〕 EGR1プロモー夕 ·ルシフェラ一ゼを用いた klothoタンパク質発 現 PEAK細胞でのアツセィ CHOras clone cells and HEK293 cells were seeded on 24-well plates (Coming, USA). After culturing for 24 hours, the culture medium was replaced with the mouse-soluble solubilized klotho protein-expressing CHOras clone cell culture supernatant prepared in 8- (1), and culturing was further performed for 4 hours. Subsequently, 10 g / ml of heparin was added, and then human FGF23 (100 ng / ml) was added. After performing incubation at 10 minutes 37, the supernatant was aspirated, washed with PBS, and lysed with SDS-PAGE sample buffer (Daiichi Kagaku, Japan). Western blotting was performed on this sample in the same manner as in Example 2 using an anti-ERK antibody (Santa Cruz, USA). As shown in FIG. 5, the enhancement of ERK phosphorylation by FGF23 was confirmed only in the cells to which the mouse soluble klotho protein was added. [Example 9] EGR1 Promoter · klotho protein expression using luciferase Acetase in PEAK cells
9-(1) マウス EGR1プロモー夕 ·ルシフェラーゼ融合遺伝子の作製  9- (1) Mouse EGR1 Promoter / Luciferase fusion gene construction
マウス EGR1の遺伝子発現上昇を調べる為、 マウス EGR1プロモータ ·ルシフエ ラーゼ融合遺伝子の作製を行なった。マウス EGR1プロモータ部分のクローニング は PCR法により行なった。 プライマーは文献 (Barbara, A. et al. , Proc. Natl. Acad. Sci. USA Vol.85, pp.7857-7861) を元に以下の様に作製した。  In order to examine the increase in mouse EGR1 gene expression, a mouse EGR1 promoter-luciferase fusion gene was prepared. Cloning of the mouse EGR1 promoter was performed by PCR. Primers were prepared as follows based on the literature (Barbara, A. et al., Proc. Natl. Acad. Sci. USA Vol. 85, pp. 7857-7861).
マウス EGR1プロモータクローニング用 FWプライマー: FW primer for mouse EGR1 promoter cloning:
GGGGTACCAACAGATCCTGGCGGGGACTTAGGAC (配列番号 2 1 )  GGGGTACCAACAGATCCTGGCGGGGACTTAGGAC (SEQ ID NO: 21)
マウス EGR1プロモータクローニング用' RVプライマー: 'RV primers for mouse EGR1 promoter cloning:
CCCAAGCTTTCGCGACTCCCCGAATCGGCCTCTATT (配列番号 2 2 )  CCCAAGCTTTCGCGACTCCCCGAATCGGCCTCTATT (SEQ ID NO: 22)
配列番号 2 1のプライマーはマウス EGR 1プロモー夕部分の- 1023塩基部分から 開始するプライマ一でさらに Hindlllサイトが付加されている。 配列番号 2 2の プライマーはマウス EGR1 mRNA開始部位 (+1) までを含むプライマーで Kpnlサイ トが付加されている。 錡型にはマウス Genomic DNA (米国 クロンテック社)を用い た。 PCR反応は LA Taq GC Buffer (日本国 宝酒造社) を使用し、 添付のマニュ アルにしたがって行なった。 PCR の反応条件は 94°Cで 5分を一次変性とし、 次 に 94で 2 0秒、 6 0°C 30秒、 7 2 °C 1分を 1サイクルとして 3 0サイクル行 い、 最後に伸長反応を 72 °Cで 7分行なった。 PCR による増幅産物は 2 %ァガロ ースゲルによる電気泳動で確認した結果、約 lkbp付近に単一バンドが確認された 為、 ゲルからバンド部分の切り出しを行い、 GeneClean キット (米国 バイオ 1 The primer of SEQ ID NO: 21 is a primer starting from the −1023 base portion of the mouse EGR1 promoter, and a Hindlll site is further added. The primer of SEQ ID NO: 22 is a primer including the mouse EGR1 mRNA start site (+1) and has a Kpnl site added thereto. For mouse type マ ウ ス, mouse Genomic DNA (Clontech, USA) was used. The PCR reaction was performed using LA Taq GC Buffer (Takara Shuzo, Japan) according to the attached manual. The PCR reaction conditions were as follows: primary denaturation at 94 ° C for 5 minutes, followed by 30 cycles of 94 ° C for 20 seconds, 60 ° C for 30 seconds, and 72 ° C for 1 minute, and finally elongation. The reaction was performed at 72 ° C for 7 minutes. The amplified product by PCR was confirmed by electrophoresis on a 2% agarose gel. As a result, a single band was detected at about lkbp. The band was cut out from the gel, and the GeneClean kit (US Bio 1) was used.
0 1社) にて精製を行なった後、 Hindlll, Kpnl にて切断を行なった。 再度(1 company) and then cut with Hindlll and Kpnl. again
GeneCleanキット(米国 バイオ 1 0 1社)にて精製を行い、同様に Hindlll, Kpnl にて切断した pGL3- Basic (米国 プロメガ社) にライゲーシヨンを行なった。 ラ ィゲーシヨン反応は TaKaRa Ligation kid version 2(日本国 宝酒造社)を用い て添付のプロトコールに準じて行なった。クローン化されたマウス EGR1プロモー タは塩基配列の確認を行ないマウス EGR1プロモータ'ルシフェラ一ゼ融合遺伝子 の作製を完了した。以下このプラスミドを mEGRlpromoter_lu / pGL3Basicと記す。Purification was performed using a GeneClean kit (U.S.A., Bio 101), and ligation was similarly performed with pGL3-Basic (Promega, U.S.A.) cut with Hindlll and Kpnl. The ligation reaction was performed using TaKaRa Ligation kid version 2 (Takara Shuzo, Japan) according to the attached protocol. The nucleotide sequence of the cloned mouse EGR1 promoter was confirmed, and the production of the mouse EGR1 promoter 'luciferase fusion gene was completed. Hereinafter, this plasmid is referred to as mEGRlpromoter_lu / pGL3Basic.
9- (2) マウス EGR1 プロモー夕 'ルシフェラーゼレポ一夕一遺伝子を用いた klotho発現 PEAK細胞におけるヒト FGF23刺激時のルシフェラーゼ活性の上昇 上記 9- (1)で作製したマウス EGR1プロモータつレシフェラ一ゼレポーター遺伝 子を用いてマウス klotho発現 PEAK細胞での EGR1の FGF23刺激時の EGR1発現変 化を調べた。 PEAK rapid細胞 (米国 エッジバイオシステム社) に添付のプロト コールに準じて実施例 3で作製した mklotho/pEAK8 と実施例 9- (1)で作製した iEGRlpromoter-luc: /pGL3Basicを同時にトランスフエクシヨンした。 トランスフ ェクシヨンは 10cm径の培養ディッシュ(米国 べクトンディッキンソン社) に行 なった。 トランスフヱクシヨン 4時間後に細胞を 0.53mM EDTA含有 PBSにて剥が し、 白色 9 6ゥエルプレート (米国 コーニング社) に播種した。 24時間培養 後、 ヒト FGF23を 0〜 1 0 Ong/mlで添加し、 へパリン (米国 シグマ社) も 1 0 g/ml になるように同時に添加した。 さらに 2 4時間 3 7°Cにて静置後、 Steady-Glo Luciferase assay system (米国 プロメガ社) を用いてプロトコ一 ルに準じてルシフェラーゼ活性を測定した。測定はトップカウント(米国 パッ力 ―ド社)を用いて行なった。 結果は図 4に示す。 9- (2) Increase in luciferase activity of human EGF1 promoter-stimulated human FGF23 in klotho-expressing PEAK cells using the luciferase repo overnight gene Using the mouse EGR1 promoter and the reporterase reporter gene prepared in 9- (1) above, changes in EGR1 expression of FGR23-stimulated EGR1 in mouse klotho-expressing PEAK cells were examined. Mklotho / pEAK8 prepared in Example 3 and iEGRlpromoter-luc: / pGL3Basic prepared in Example 9- (1) were transfected simultaneously according to the protocol attached to PEAK rapid cells (Edge Biosystems, USA) . The transfection was performed in a 10 cm-diameter culture dish (Becton Dickinson, USA). Four hours after transfection, the cells were detached with PBS containing 0.53 mM EDTA, and seeded on a white 96-well plate (Corning, USA). After culturing for 24 hours, human FGF23 was added at 0 to 10 Ong / ml, and heparin (Sigma, USA) was also added so as to be 10 g / ml. After standing still at 37 ° C for 24 hours, luciferase activity was measured using a Steady-Glo Luciferase assay system (Promega, USA) according to the protocol. The measurement was performed using a TopCount (US-based Pacikido). The results are shown in FIG.
〔実施例 1 0〕 抗マウス klothoタンパク質部分ペプチドポリクローナル抗体の 作製  [Example 10] Preparation of anti-mouse klotho protein partial peptide polyclonal antibody
10- (1) FGF- 23部分配列に相当するペプチドの合成  10- (1) Synthesis of peptide corresponding to FGF-23 partial sequence
配列番号 23 (マウス全長 klotho タンパク質) のタンパク質の部分について DNASIS Macヴァージョン 7. 2の計算機能を用いて、 疎水性度を予測し、 ぺプチ ド抗体作製に適した部位を予測した。 親水性の程度が高く、 かつ糖鎖修飾やリン 酸化部位となりうる部位以外という観点からさらに抗体作製に適切と考えられる 部分配列を抽出した。その結果、配列番号 2 3の残基番号 97番目ヒスチジンから 始まる 16残基のアミノ酸からなるペプチドの C末端にシスティン残基を付加した ペプチド mklotlio-97(配列番号 24)、 118番目プロリンから始まる 21アミノ酸残 基からなるペプチドの N 末端にシスティ ン残基を付加したペプチド mklotho - 118(配列番号 2 5)、 299番目ァラニンから始まる 20アミノ酸残基から なるペプチド mklotho- 299(配列番号 2 6)、 941番目プロリンから始ま ¾ 25アミ ノ酸残基からなるペプチド mklotho-941 (配列番号 2 7)、 を抗原として選択し化 学合成した。  For the protein part of SEQ ID NO: 23 (mouse full-length klotho protein), using the calculation function of DNASIS Mac Version 7.2, the hydrophobicity was predicted, and a site suitable for peptide antibody production was predicted. From the viewpoint that the degree of hydrophilicity is high and the site is not a site that can be a sugar chain modification or phosphorylation site, a partial sequence that is considered to be more suitable for antibody production was extracted. As a result, peptide mklotlio-97 (SEQ ID NO: 24) with a cysteine residue added to the C-terminal of a peptide consisting of 16 amino acids starting from histidine at residue number 97 in SEQ ID NO: 23, starting from proline at position 118 21 Peptide mklotho-118 (SEQ ID NO: 25) with a cysteine residue added to the N-terminus of the peptide consisting of amino acid residues, peptide mklotho-299 (SEQ ID NO: 26) consisting of 20 amino acid residues starting from alanine at position 299, A peptide mklotho-941 (SEQ ID NO: 27) consisting of 25 amino acid residues starting from the 941th proline was selected as an antigen and chemically synthesized.
mklotho- 97: HHSGAAPSDSPIVVAPC (配列番号 24) mklotho-97: HHSGAAPSDSPIVVAPC (SEQ ID NO: 24)
mklotho- 118: CPPLSSTGDVASDSYNNVYRDT (配列番号 2 5) mkl ot o-299 : ALSS而 INPR履 TDYNIREC (配列番号 2 6 ) mklotho- 118: CPPLSSTGDVASDSYNNVYRDT (SEQ ID NO: 25) mkl ot o-299: ALSS-INPR TDYNIREC (SEQ ID NO: 26)
mkl ot o-941 : PSMKHYRKI IDSNGFLGSGTLGRFC (配列番号 2 7 ) mkl ot o-941: PSMKHYRKI IDSNGFLGSGTLGRFC (SEQ ID NO: 27)
10— (2) 抗 FGF - 23部分べプチドに対するポリクローナル抗体の調製  10— (2) Preparation of polyclonal antibody against anti-FGF-23 partial peptide
上述の全てのペプチドはそれぞれの持つシスティン残基を介してキヤリァ夕ン パク質であるゥシサイログロブリンと結合させ、 免疫に供した。 免疫は一つの抗 原ペプチドに対して 2羽のゥサギを用いた。 初回免疫は、 1羽あたり lOO gのキ ャリァ蛋白に結合したペプチドをフロイト完全アジュバントでェマルジヨンとし たものをゥサギの皮内または皮下に投与した。初回免疫の 1週間後に lOO gのキ ャリア蛋白に結合したペプチドをフロイ卜不完全アジュバントでェマルジョンと したものを同様に投与した。 これと同じ投与を 2週間間隔で 6回実施し、 最終投 与の 1週間後に全採血を行ない、 抗血清を調製した。  All of the above-mentioned peptides were bound to carrier protein ゥ -thyroglobulin via their cysteine residues and subjected to immunization. Immunization was performed using two egrets for one antigen peptide. In the first immunization, a peptide conjugated to lOO g of a carrier protein per bird was emulsified with Freud's complete adjuvant and administered intradermally or subcutaneously to a egret. One week after the first immunization, a peptide conjugated to lOO g of the carrier protein was emulsified with incomplete Freund's adjuvant and administered similarly. The same administration was performed six times at two-week intervals, and one week after the final administration, whole blood was collected to prepare an antiserum.
ゥサギ血清からの抗マウス klotho タンパク質の部分ペプチドに対する抗体を 精製するためのァフィ二ティカラムを作製するために、 免疫に用いたそれぞれの ペプチドを Sul foLink Ki t (米国、 PIERCE社) を用いてゲル上に固相化した。 吸 着緩衝液として PBS (-)を用いて抗血清をこのカラムに添加し、免疫に用いたぺプ チドと結合する抗体をカラムに保持した。次に溶出緩衝液として 0. 1Mグリシン緩 衝液 (pH2. 8) を用いてカラムに結合した抗体を溶出させ、 回収した。 溶出画分は lM Tr is-HCl ( H 9. 0)を添加して pH7. 2付近に調整した。調製された抗体溶液は、 ゲルろ過カラム NAP25 (米国、 Amers am Pharmac i a Biotech社)に供して PBS (-)に 緩衝液を置換した後、 孔径 0. のメンブランフィルター MILLEX-GV (米国、 Mi l l ipore 社) でろ過滅菌し、 各ペプチドに対する抗体を得た。 精製抗体の濃度 は 280皿の吸光度を測定し、 lmg/mlを 1. 350Dとして算出した。 このようにして 得られた各精製抗体を、 免疫に用いたペプチドの名前を用いて示すことにする。 例えば配列番号 2 4の mklo tho-97 ペプチドを免疫して得られた抗体に関しては mklotho-97抗体と記載することにする。  ゥ To prepare an affinity column for purifying antibodies against partial peptides of the anti-mouse klotho protein from heron serum, each peptide used for immunization was gel-prepared using SulfoLink Kit (PIERCE, USA). Was immobilized. Antiserum was added to this column using PBS (-) as an adsorption buffer, and the antibody that binds to the peptide used for immunization was retained on the column. Next, the antibody bound to the column was eluted and recovered using a 0.1 M glycine buffer (pH 2.8) as an elution buffer. The eluted fraction was adjusted to around pH 7.2 by adding 1M Tris-HCl (H 9.0). The prepared antibody solution was applied to a gel filtration column NAP25 (Amersham Pharmacia Biotech, USA) to replace the buffer with PBS (-), and then a membrane filter with a pore size of 0.MILLEX-GV (Mill, USA) The solution was sterilized by filtration using ipore) to obtain antibodies against each peptide. The concentration of the purified antibody was calculated by measuring the absorbance of 280 dishes and setting lmg / ml to 1.350D. Each of the purified antibodies thus obtained will be indicated using the name of the peptide used for immunization. For example, an antibody obtained by immunizing the mklotho-97 peptide of SEQ ID NO: 24 will be referred to as a mklotho-97 antibody.
10— (3) 抗マウス klothoタンパク質部分ペプチド抗体によるマウス kl othoタン パク質の認識  10— (3) Recognition of mouse kl otho protein by anti-mouse klotho protein partial peptide antibody
上述の方法により取得した mklotho-97抗体、 mklotho-118抗体、 mklotho-299 抗体および mklotlio_941抗体を用いて、 実施例 8によって作製したマウス可溶化 klot o夕ンパク質一過性発現 PEAK rapid細胞上清中のマウス可溶化 kl otho夕ン パク質をウェスタンプロッティング法により解析を行った。 図 6に示すように、 いずれの抗体でも 130 kDa付近のマウス可溶化 kl othoタンパク質が検出された。 したがって、これら 4種類の抗体はマウス klo thoを認識し得る抗体であることが 判明した。 Using the mklotho-97 antibody, mklotho-118 antibody, mklotho-299 antibody, and mklotlio_941 antibody obtained by the above method, the mouse solubilized klot o protein transiently expressed PEAK rapid cell supernatant prepared in Example 8 Mouse solubilization in mouse The protein was analyzed by Western plotting. As shown in FIG. 6, mouse-solubilized k-lotho protein near 130 kDa was detected for all antibodies. Therefore, these four types of antibodies were found to be antibodies that can recognize mouse klotho.
〔実施例 1 1〕 ピオチン標識抗体の作製  Example 11 Preparation of Piotin-Labeled Antibody
上述の精製した 4種のマウス klothoタンパク質の部分ペプチドに対するポリク ローナル抗体を用いてピオチン標識を行った。 50mM 炭酸水素ナトリウム溶液で lmg/mlの濃度に希釈した抗体溶液 1mlに、 Biot in- AC5- Osu (日本、 同仁化学社)を ジメチルホルムアミドに 1. 82mg/ilの濃度で溶解した溶液を 10 l添加し、 4°Cに おいて 2時間転倒混和した。 その後、 この反応液を NAP10カラム(米国、 Amersham Pharmac ia Biotech社)に供し、 未反応の B iot in- AC5- Osuを除き、 溶媒を PBS (-) に置換し、 4種類のピオチン標識抗 FGF23部分べプチドポリクローナル抗体を得 た。  Piotin labeling was performed using polyclonal antibodies against the above-mentioned purified four partial peptides of the mouse klotho protein. To 1 ml of antibody solution diluted to lmg / ml with 50mM sodium bicarbonate solution, add 10l of a solution of Biotin-AC5-Osu (Dohin Chemical Co., Japan) dissolved in dimethylformamide at a concentration of 1.82mg / il. The mixture was added and mixed by inversion at 4 ° C for 2 hours. Then, this reaction solution was applied to a NAP10 column (Amersham Pharmacia Biotech, USA) to remove unreacted Biotin-AC5-Osu, and the solvent was replaced with PBS (-). A partial peptide polyclonal antibody was obtained.
〔実施例 1 2〕 FGF-23と klothoタンパク質の結合実験  [Example 1 2] Binding experiment between FGF-23 and klotho protein
FGF-23タンパク質と klothoタンパク質が結合することを以下の実験によって 確かめた。  The binding of the FGF-23 protein to the klotho protein was confirmed by the following experiment.
12- (1) FGF- 23固定化レジンの作製 - 組換え FGF- 23RQH の作製および精製は後述の実施例 1 5に従って行った。本精 製物を NHS活性化セファロ一ス 4FF (米国、 Amersliam Pharmacia Bio tech社) に 常法に従いレジン 1mlあたり 3. 3mg固定化した。  12- (1) Preparation of FGF-23-immobilized resin-Preparation and purification of recombinant FGF-23RQH were performed according to Example 15 described later. This purified product was immobilized on NHS-activated Sepharose 4FF (Amersliam Pharmacia Biotech, USA) in a usual manner at 3.3 mg per 1 ml of resin.
12- (2) FGF-23RQH固定化レジンへの可溶化 klothoタンパク質の結合  12- (2) Binding of solubilized klotho protein to FGF-23RQH-immobilized resin
実施例 8で作製した 8ml の可溶化マウス klotho タンパク質一過性発現 PEAK rap id細胞上清もしくは対照として親株の PEAK rap id細胞の上清に対して、 上記 8 ml of the solubilized mouse klotho protein transiently-expressed PEAK rap id cell supernatant prepared in Example 8 or the parental PEAK rap id cell supernatant as a control,
FGF-23固定化レジン 50 ^ 1を添加し、 4°Cにおいて 1時間転倒混和して可溶化マ ウス klothoタンパク質と FGF- 23を反応させた。 その後 PBSでレジンを 4回洗浄 し、未反応物を除去した。それぞれのレジンに 300 1のサンプルバッファー( 50mMFGF-23-immobilized resin (50 ^ 1) was added, and the mixture was inverted at 4 ° C for 1 hour to react the solubilized mouse klotho protein with FGF-23. Thereafter, the resin was washed four times with PBS to remove unreacted substances. 300 1 sample buffer (50 mM
Tr i s-HCl pH6. 8、 0. 4% SDS、 6%グリセロール、 0. 002%ブロムフエノールブルー、Tris-HCl pH 6.8, 0.4% SDS, 6% glycerol, 0.002% bromphenol blue,
2% i3メルカプトエタノール) を添加し、 95°Cで 5分間加熱したのち、 遠心分離し て得た上清を回収した。 これらの上清および細胞上清を実施例 1 0で取得した mklotho- 118抗体および mklotho- 941抗体を用いて、 ウェスタンブロッティング 法により解析を行った。 その結果を図 7に示す。 また同様のサンプルを 2D-銀染 色試薬 · II 「第一」 (日本国、 第一化学薬品株式会社) を用いて銀染色した結果に ついても図 7に示す。'これらの結果より、 FGF- 23RQH 固定化カラムに可溶化マウ ス kl otho タンパク質が結合し、 濃縮されることが示された。 よって FGF- 23 と klothoタンパク質が結合することが明らかにされた。 After adding 2% i3 mercaptoethanol) and heating at 95 ° C for 5 minutes, the supernatant obtained by centrifugation was collected. Western blotting of these supernatants and cell supernatants using the mklotho-118 and mklotho-941 antibodies obtained in Example 10 Analysis was performed by the method. Figure 7 shows the results. Fig. 7 also shows the results of silver staining of a similar sample using 2D-silver staining reagent II II (Daiichi Chemical Co., Ltd., Japan). 'These results indicated that the solubilized mouse klotho protein was bound to the FGF-23RQH-immobilized column and concentrated. Therefore, it was clarified that FGF-23 binds to klotho protein.
〔実施例 1 3〕 可溶化 klothoタンパク質発現 CH0細胞の移植試験  [Example 13] Transplantation test of solubilized klotho protein-expressing CH0 cells
13- (1) 血清 1, 25ジヒドロキシビタミン D濃度の検討 13- (1) Examination of serum 1,25 dihydroxyvitamin D concentration
可溶化 klothoタンパク質組換え体の生物活性を検討するため、実施例 5で得ら れた可溶化 klothoタンパク質安定発現 CH0細胞をヌードマウスへ皮下移植するこ とにより、可溶化 klothoタンパク質組換え体を恒常的に分泌する移植細胞由来の 腫瘍を保持した可溶化 klothoタンパク質高発現モデルマウスを作成した。実験動 物には 7週齢の雄性 BALB/cヌ一ドマウスを用い、細胞移植は以下の要領で実施し た。 マウスはその入荷より解剖に到る全ての期間中、 プラスチックケージにて飼 育し市販のげつ歯類用固形餌ならびに水道水を自由摂取させた。  In order to examine the biological activity of the solubilized klotho protein recombinant, the solubilized klotho protein recombinant stably expressed CH0 cells obtained in Example 5 were subcutaneously implanted into nude mice to obtain the solubilized klotho protein recombinant. A mouse model for high expression of solubilized klotho protein, which retains tumors derived from transplanted cells that secrete constantly, was created. Seven-week-old male BALB / c nude mice were used as experimental animals, and cell transplantation was performed as follows. Mice were kept in plastic cages throughout the period from their arrival to dissection and were allowed free access to commercially available rodent chow and tap water.
可溶化型 klotho夕ンパク質発現 CH0細胞ならびに対照として親株 CH0細胞をそ れぞれ 1 X 108 ce l l s/mL となるよう PBSバッファーに分散しこれら細胞懸濁液を ヌードマウスの背部皮下二箇所へそれぞれ 0. lmLずつ注入した (対照 CH0細胞移 植群 n=5、 可溶化型 klothoタンパク質発現 CHO細胞移植群 n=5)。 細胞移植後 14 日目において、 エーテル麻酔下、 心採血により血液を採取し、 マイクロティナ (Bec ton Dickinson, 米国) を用いて血清を調製した。 得られた血清中の 1, 25 ジヒドロキシビタミン D濃度を 1, 25 (0H) 2D RIAキット (TFB、 米国) を用いて測 定した。 Solubilized klotho protein-expressing CH0 cells and the parental CH0 cells as a control were each dispersed in PBS buffer at 1 × 10 8 cells / mL, and these cell suspensions were subcutaneously subcutaneously at the back of nude mice at two locations. 0.1 mL each was injected into the control group (control CH0 cell transplantation group n = 5, solubilized klotho protein-expressing CHO cell transplantation group n = 5). On day 14 after cell transplantation, blood was collected by cardiac blood sampling under ether anesthesia, and serum was prepared using Microtina (Becton Dickinson, USA). The 1,25-dihydroxyvitamin D concentration in the obtained serum was measured using a 1,25 (0H) 2D RIA kit (TFB, USA).
その結果、 可溶化型 klothoタンパク質発現細胞移植群の 1, 25ジヒドロキシビ 夕ミン!)濃度は、 対照 CH0細胞移植群のそれに比し有意に低下していた (図 8 )。 13- (2) 血清リン濃度の検討  As a result, the concentration of 1,25-dihydroxyvinylamine in the group transplanted with the soluble klotho protein-expressing cells was significantly lower than that in the group transplanted with the control CH0 cells (FIG. 8). 13- (2) Examination of serum phosphorus concentration
上述の方法と同様の方法で、可溶化 klotho夕ンパク質発現 CH0細胞ならびに対 照 CH0細胞を 5週齢の雄性 BALB/cヌードマウスに皮下移植し(対照 CH0細胞移植 群 n=6、 可溶化 kl othoタンパク質発現 CHO細胞移植群 n=6)、 27日目における血 清リン濃度をピ一テストヮコー (和光純薬、 日本) を用いて測定した。  In the same manner as above, solubilized klotho protein-expressing CH0 cells and control CH0 cells were subcutaneously transplanted into 5-week-old male BALB / c nude mice (control CH0 cell transplantation group n = 6, solubilized The serum phosphorus concentration on day 27 of the klotho protein-expressing CHO cell transplant group n = 6) was measured using Pitest Corporation (Wako Pure Chemical, Japan).
その結果、図 8に示すように、可溶化 klothoタンパク質発現細胞移植群の血清 リン濃度は、 対照 CH0細胞移植群のそれに比し有意に低— As a result, as shown in FIG. 8, the sera of the solubilized klotho protein-expressing cell transplant group Phosphorus concentration is significantly lower than that of control CH0 cell transplant group.
以上の結果から、可溶化 klo thoタンパク質組換え体は、血清リン濃度ならびに 1, 25ジヒドロキシビタミン D濃度を低下させる生理活性を有していることが証明 された。 この活性はすでに報告されている FGF-23のそれと同様のものである。 〔実施例 1 4〕 抗 klothoタンパク質抗体のマウス投与によるリン、 ビタミン D 上昇作用  From the above results, it was proved that the solubilized klo tho protein recombinant had a physiological activity to reduce serum phosphorus concentration and 1,25 dihydroxyvitamin D concentration. This activity is similar to that of previously reported FGF-23. [Example 14] Phosphorus and Vitamin D Elevating Effect by Administration of Anti-klotho Protein Antibody to Mice
BALB/c, 雄、 7週齢の正常マウスに実施例 1 0で取得した抗 klotho夕ンパク質 抗体 P118を腹腔内へ 1匹あたり 200 g/O. 2mLずつ単回投与した。 対照群へは、 溶媒 (PBS)を 0. 2mLずつ腹腔内へ投与した。投与 9時間後にガラス製キヤピラリを 用いて眼窩より採血し、 マイクロティナ (べクトンディツキソン社、 米国) を用 いて血清を分離、 さらに 24時間後に腹大静脈より採血し、 同じくマイクロティナ を用いて血清を分離した。 得られた 9時間後の血清で 1, 25-ジヒドロキシビタミ ン D濃度を 1, 25 (0H) 2D RIAキット 「TFB」 (ティエフビー社、 日本国)を用いて測 定した。また 24時間後の血清でリン酸濃度をリン-テストヮコー(和光純薬、 日本 国)を用いて添付文書に従い、 決定した。 抗 klothoタンパク質抗体および対照群 はそれぞれ 6匹のマウスから構成され、水道水および 1. 03 %の無機リン酸および 1. 18%のカルシウムを含んだ固形食 CE2 (日本クレア社、 日本国)を自由摂取させ た。  BALB / c, male, 7-week-old normal mice were intraperitoneally administered once with the anti-klotho protein antibody P118 obtained in Example 10 at 200 g / O. 2 mL / mouse. To the control group, 0.2 mL of the solvent (PBS) was intraperitoneally administered. Nine hours after administration, blood was collected from the orbit using a glass capillary, serum was separated using Microtina (Becton Dickson, USA), and 24 hours later, blood was collected from the abdominal vena cava, using Microtina again. To separate the serum. Nine hours after the obtained serum, the concentration of 1,25-dihydroxyvitamin D was measured using a 1,25 (0H) 2D RIA kit “TFB” (TFB, Japan). The serum phosphate concentration 24 hours later was determined using Phosphor-Test II Co. (Wako Pure Chemical Industries, Japan) according to the package insert. The anti-klotho protein antibody and the control group consisted of 6 mice each, and were supplied with tap water and a solid diet containing 1.03% inorganic phosphate and 1.18% calcium CE2 (CLEA Japan, Japan). They were allowed free intake.
その結果、 P118抗体投与 9時間後の 1, 25-ジヒドロキシビタミン ΰ、 24時間後 の血清リン酸濃度ともに媒体投与群に比べて有意に上昇していた (図 9 )。  As a result, the 1,25-dihydroxyvitamin 9 9 hours after administration of the P118 antibody and the serum phosphate concentration 24 hours after administration were significantly higher than those in the vehicle-administered group (FIG. 9).
以上の結果より、抗 klothoタンパク質抗体は血清リン濃度、血清活性型ビタミ ン D濃度を上昇しうることが示された。  From the above results, it was shown that the anti-klotho protein antibody can increase the serum phosphorus concentration and the serum activated vitamin D concentration.
〔実施例 1 5〕 FGF- 23、 FGF-23H, FGF-23RQHタンパク質タンパク質の取得 Example 15 Acquisition of FGF-23, FGF-23H, and FGF-23RQH Proteins
15 - (1) FGF-23H夕ンパク質発現べクタ一の構築 15-(1) Construction of FGF-23H protein expression vector
FGF-23をコ一ドする cDNAは、腫瘍性骨軟化症の責任腫瘍の cDNAライブラリ一 を铸型とし、 F lEcoRIプライマー (配列番号 2 8 ) と LHi sNot プライマー (配列 番号 2 9 ) と LA- Tad DNA polymeraseを用いて 96°Cで 1分間保温した後、 96°Cで The cDNA coding for FGF-23 was derived from a cDNA library of the tumor responsible for neoplastic osteomalacia, type III, using the FlEcoRI primer (SEQ ID NO: 28), the LHisNot primer (SEQ ID NO: 29), and LA- After incubating at 96 ° C for 1 minute using Tad DNA polymerase,
30秒、 55でで 30秒、 72°Cで 30秒からなる工程を 1サイクルとした 35サイクル の PCRを実施することにより増幅した。 F lEcoRIプライマーは FGF- 23をコードす る塩基配列のさらに 5'側上流に存在する配列にァニールし、 その増幅断片の FGF-23をコードする領域の 5'側に EcoRI制限酵素部位を付加する。 LHisNotブラ イマ一は FGF- 23をコードする配列の終始コドンの 5'側の配列とァニールする配 列と His6_tag配列 (His - His- His - His- His- His) をコードする配列に続く終始コ ドンと Notl制限酵素配列を含む。 その結果、 増幅断片は FGF-23タンパク質タン パク質の C末端に His6-tag配列を付加したものをコードすることになり、その下 流に Notl制限酵素部位を有する。 この増幅断片を EcoRIと Notlで消化し、 同様 に EcoRI と Notl で消化した動物細胞発現ベクターである pcDNA3. lZeo (米国 Invitrogen社)と連結した。このように作製した発現ベクターをクローニングし、 塩基配列を決定して目的の H i s 6- 1 ag配列が付加された FGF- 23タンパク質タンパ ク質をコ一ドしていることを確認した。 このベクターを pcDNAFGF- 23Hと称す。 FlEcoRI: CCGGAATTCAGCCACTCAGAGCAGGGCACG (配列番号 2 8 ) Amplification was performed by performing 35 cycles of PCR, in which a cycle consisting of 30 seconds at 55, 30 seconds at 55, and 30 seconds at 72 ° C was one cycle. The FlEcoRI primer anneals to a sequence located further 5 ′ upstream of the nucleotide sequence encoding FGF-23, and Add an EcoRI restriction enzyme site 5 'to the region encoding FGF-23. The LHisNot primer is a sequence that anneals to the sequence 5 'of the stop codon of the sequence encoding FGF-23 and the sequence following the sequence that encodes the His6_tag sequence (His-His-His-His-His-His-His). Contains Don and Notl restriction enzyme sequences. As a result, the amplified fragment encodes the FGF-23 protein with a His6-tag sequence added to the C-terminus, and has a Notl restriction enzyme site downstream. This amplified fragment was digested with EcoRI and Notl, and ligated to pcDNA3.lZeo (Invitrogen, USA), which was an animal cell expression vector similarly digested with EcoRI and Notl. The expression vector thus prepared was cloned, and its nucleotide sequence was determined. It was confirmed that the FGF-23 protein protein to which the desired His6-1-ag sequence had been added was encoded. This vector is called pcDNAFGF-23H. FlEcoRI: CCGGAATTCAGCCACTCAGAGCAGGGCACG (SEQ ID NO: 28)
LHisNot: ATAAGAATGCGGCCGCTCAATGGTGATGGTGATGATGGATGAACTTGGCGAA (配列番号 2 9) LHisNot: ATAAGAATGCGGCCGCTCAATGGTGATGGTGATGATGGATGAACTTGGCGAA (SEQ ID NO: 29)
15- (2) FGF - 23タンパク質発現ベクターの構築  15- (2) Construction of FGF-23 protein expression vector
pcDNA/FGF- 23Hを錶型として FlEcoRIプライマーと LNotプライマー(配列番号 3 0) と LA-Tan DNA polymeraseを用いて 94°Cで 1分間保温した後、 94°Cで 30 秒、 55°Cで 30秒、 72 で 1分からなる工程を 1サイクルとした 25サイクルの PCR を実施することにより増幅した。反応終了後、 PCR産物の末端を T4 DNA polymerase (スイス、 Roche社)により平滑化したのち、 Polynucleotide kinase' (スイス、 Roche社)を用いて DNA末端のリン酸化を行うことで FGF- 23タンパク質をコード する cDNA断片を調製した。 発現べクタ一 pCAGGS (Niwa H, et al. , Gene. 1991, 108:193-199) を EcoRIで消化し Klenow断片 (スイス、 Roche社) を用いて末端 を平滑化し、 ゥシ小腸アルカリフォスファターゼ (日本、 宝酒造社) を用いて脱 リン酸化を行った。 このように調製した FGF- 23をコードする cDNA断片と pCAGGS ベクターを連結した。 このように作製した発現べクタ一をクローニングし、 塩基 配列を決定して目的の FGF - 23 タンパク質をコードする配列が的確に挿入されて いることを確認した。 このべクタ一を pCAGGS/FGF- 23と称す。  After incubating pcDNA / FGF-23H for type I with FlEcoRI primer, LNot primer (SEQ ID NO: 30) and LA-Tan DNA polymerase for 1 minute at 94 ° C, 30 seconds at 94 ° C and 55 ° C Amplification was performed by performing 25 cycles of PCR, with one cycle consisting of 30 seconds and 72 minutes 1 minute. After completion of the reaction, the end of the PCR product is blunted with T4 DNA polymerase (Roche, Switzerland), and then the FGF-23 protein is phosphorylated with Polynucleotide kinase '(Roche, Switzerland) to convert the FGF-23 protein. An encoding cDNA fragment was prepared. Expression vector pCAGGS (Niwa H, et al., Gene. 1991, 108: 193-199) was digested with EcoRI, blunt-ended with Klenow fragment (Roche, Switzerland), and the small intestine alkaline phosphatase ( Dephosphorylation was performed using Takara Shuzo, Japan. The thus prepared cDNA fragment encoding FGF-23 was ligated to the pCAGGS vector. The expression vector thus prepared was cloned and its nucleotide sequence was determined to confirm that the sequence encoding the desired FGF-23 protein was correctly inserted. This vector is called pCAGGS / FGF-23.
また上述の FlEcoRIプライマーと LNotプライマーで増幅した FGF- 23をコード する断片を EcoRIと Notlで消化したのち精製した。 これを、発現ベクターである The FGF-23-encoding fragment amplified with the FlEcoRI and LNot primers was digested with EcoRI and Notl and purified. This is the expression vector
PEAK8 (米国、 Edge Biosystem社) に分子内リボゾームエントリー配列 (IRES) と増強型緑色蛍光タンパク質 (EGFP) を連結した PEAK8/IRES/EGFP ベクターの EcoRIと Not l制限酵素部位に挿入してクロ一ニングした。取得したプラスミドの 塩基配列を決定し、 FGF- 23タンパク質をコードしていることを確認した。 このべ クタ一を PEAK8/IRES/EGFP/FGF- 23と称す。 Intramolecular ribosome entry sequence (IRES) in PEAK8 (Edge Biosystem, USA) And the enhanced green fluorescent protein (EGFP) were ligated and inserted into the EcoRI and Notl restriction enzyme sites of the PEAK8 / IRES / EGFP vector and cloned. The nucleotide sequence of the obtained plasmid was determined, and it was confirmed that it encodes the FGF-23 protein. This vector is called PEAK8 / IRES / EGFP / FGF-23.
LNot: ATAAGAATGCGGCCGCTCAGATGAACTTGGCGAA (配列番号 3 0 ) LNot: ATAAGAATGCGGCCGCTCAGATGAACTTGGCGAA (SEQ ID NO: 30)
pCAGGS/FGF-23を EcoRIで消化して直鎖化したのち、 Kl enow断片(スイス、 Roche 社) を用いて末端を平滑化した。 これをさらに BamHIで消化して FGF-23の cDNA を含む DNA断片をァガロース電気泳動で分離精製した。また、発現ベクター INPEP4 を Bgl l lで消化した後、 Klenow断片 (スイス、 Roche社) を用いて末端を平滑化 した。 さらに BamHIで消化したのちァガロース電気泳動でベクターを精製した。 これらの FGF-23 cDNAを含む断片とベクターを連結した。 このように作製した発 現ベクターをクローニングし、塩基配列を決定して目的の FGF-23タンパク質をコ 一ドする配列が的確に挿入されていることを確認した。 このベクターを INPEP4/FGF-23と称す。  After pCAGGS / FGF-23 was linearized by digestion with EcoRI, the ends were blunted using a Klenow fragment (Roche, Switzerland). This was further digested with BamHI, and a DNA fragment containing FGF-23 cDNA was separated and purified by agarose electrophoresis. After digesting the expression vector INPEP4 with Bglll, the ends were blunt-ended using a Klenow fragment (Roche, Switzerland). After digestion with BamHI, the vector was purified by agarose gel electrophoresis. The fragment containing the FGF-23 cDNA was ligated to a vector. The expression vector thus prepared was cloned and its nucleotide sequence was determined to confirm that the sequence encoding the desired FGF-23 protein was correctly inserted. This vector is called INPEP4 / FGF-23.
15- (3) FGF-23RQHタンパク質発現べク夕一の構築  15- (3) Construction of FGF-23RQH protein expression vector
FGF- 23タンパク質は、 179番目の Arg残基と 180番目の Ser残基の間でタンパ ク質切断を受けやすいことが見出された。 この切断部位の N末側アミノ酸配列は タンパク質変換酵素の認識配列である Arg-X-X-Arg 配列に一致する Argl76-His l77-T rl78-Argl79 (配列番号 3 1 ) であり、 また ADHRでのミスセン ス変異がこの 176番目又は 179番目の Arg残基の置換変異であることが知られて いる。 そこで我々は、 タンパク質変換酵素による切断に耐性を示し、 尚かつ ADHR に見られる変異 FGF-23のモデルとして C末端に Hi s6-tagを有し、 176番目と 179 番目の Arg残基を Gin残基に置換した変異 FGF- 23タンパク質 (以下 FGF- 23RQH という) を実験的に作製するためのベクター構築を行った。  The FGF-23 protein was found to be susceptible to protein cleavage between the Arg residue at position 179 and the Ser residue at position 180. The N-terminal amino acid sequence at this cleavage site is Argl76-Hisl77-Trl78-Argl79 (SEQ ID NO: 31) corresponding to the Arg-XX-Arg sequence, which is a recognition sequence of the protein converting enzyme. Is known to be a substitution mutation at the 176th or 179th Arg residue. Therefore, we developed a model for the mutant FGF-23 found in ADHR that has resistance to cleavage by protein converting enzyme, has a His6-tag at the C-terminus, and removes the 176th and 179th Arg residues from the Gin residue. A vector was constructed to experimentally produce a mutant FGF-23 protein (hereinafter referred to as FGF-23RQH) substituted with a group.
作製方法は、 Argを Ginに置換するための塩基置換配列を含む順方向プライマ 一である RQF (配列番号 3 2 ) と逆方向プライマーである RQR (配列番号' 3 3 ) を 合成した。  For the preparation method, RQF (SEQ ID NO: 32), which is a forward primer containing a base substitution sequence for substituting Arg with Gin, and RQR (SEQ ID NO: 33), a reverse primer, were synthesized.
さらに、この塩基置換プライマーと組み合わせて変異導入部位の 5'側と 3'側の Furthermore, in combination with this base substitution primer, the 5 'and 3'
FGF- 23配列を増幅するためのプライマーである ME1 (配列番号 3 4 ) と HNt (配 列番号 3 5 ) を作製した。 ME1 は FGF- 23 cDNAがコードする開始コドンを含む部 分の順方向のプライマーであり EcoRI制限酵素配列を有する、 HNtは FGF- 23 cDNA がコードする終始コドンの前に His6- tag配列をコードするコドン配列を挿入で き、 Notl制限酵素配列を付加することができる逆方向プライマーである。 ME1 (SEQ ID NO: 34) and HNt (SEQ ID NO: 35), which are primers for amplifying the FGF-23 sequence, were prepared. ME1 contains the start codon encoded by the FGF-23 cDNA HNt can insert a His6-tag sequence-encoding codon sequence before the stop codon encoded by FGF-23 cDNA, and add a Notl restriction enzyme sequence. The reverse primer can be.
RQF: ATACCACGGCAGCACACCCAGAGCGCCGAG (配列番号 3 2 )  RQF: ATACCACGGCAGCACACCCAGAGCGCCGAG (SEQ ID NO: 32)
RQR: CTCGGCGCTCTGGGTGTGCTGCCGTGGTAT (配列番号 3 3 )  RQR: CTCGGCGCTCTGGGTGTGCTGCCGTGGTAT (SEQ ID NO: 33)
ME1: ATGAATTCCACCATGTTGGGGGCCCGCCTCAGG (配列番号 34)  ME1: ATGAATTCCACCATGTTGGGGGCCCGCCTCAGG (SEQ ID NO: 34)
HNt: ATGCGGCCGCCTAATGATGATGATGATGATGGATGAACTTGGCGAAGGG (配列番号 3 5 ) HNt: ATGCGGCCGCCTAATGATGATGATGATGATGGATGAACTTGGCGAAGGG (SEQ ID NO: 35)
10 ngの pGAGGS/FGF-23を铸型として RQFと HNtの組み合わせと ME1 と RQRの 組み合わせのプライマー (各 200nM) で PCR 反応を実施した。 反応は pfu DNA polymerase (米国、 Promega社)を用い、 94°Cで 1分間保温した後、 94°Cで 30秒、 55°Cで 30秒、 72°Cで 1分からなる工程を 1サイクルとした 25サイクル実施した。 この様にして得た 2種類の反応液を 10倍に希釈し、 そのうちの 1/21ずつを混和 して铸型とし、 これに ME1と HNtが終濃度 200nMとなるように添加した 50 1の PCR反応液を調製し、 94°Cで 1分間保温した後、 94°Cで 30秒、 55°Cで 30秒、 72°C で 1分からなる工程を 1サイクルとした 25サイクルの PCR反応を実施した。ここ では LA Ta 丽 A polymerase (日本、 宝酒造社) を使用した。 得られた約 800bp の増幅産物を、 EcoRIと Notlで消化したのちに精製してィンサート DNAを取得し た。 これを、 発現ベクターである PEAKS (米国、 Edge Biosystem社) に分子内リ ポゾームエントリー配列 (IRES) と増強型緑色蛍光タンパク質 (EGFf) を連結し た PEAK8/IRES/EGFPベクターの EcoRIと Notl制限酵素部位に挿入してクローニン グした。 取得したプラスミドの塩基.配列を決定し、 目的の 176番目と 179番目の Argが Ginに変換され、 C末端に His6- tag配列が付加された変異 FGF-23タンパク 質 を コ ー ド し て い る こ と を 確 認 し た 。 こ の ベ ク タ ー を PEAK8/IRES/EGFP/FGF-23RQHと称す。 Using 10 ng of pGAGGS / FGF-23 as type II, PCR reaction was performed with primers (200 nM each) of the combination of RQF and HNt and the combination of ME1 and RQR. The reaction was carried out using pfu DNA polymerase (Promega, USA) at 94 ° C for 1 minute, followed by one cycle consisting of 94 ° C for 30 seconds, 55 ° C for 30 seconds, and 72 ° C for 1 minute. 25 cycles were performed. The two kinds of reaction solutions thus obtained were diluted 10-fold, and 1/21 of each was mixed to form a 铸, and ME1 and HNt were added to a final concentration of 200 nM. Prepare a PCR reaction solution, incubate at 94 ° C for 1 minute, and perform 25 cycles of PCR reaction consisting of a process consisting of 30 seconds at 94 ° C, 30 seconds at 55 ° C, and 1 minute at 72 ° C. Carried out. Here, LA Ta 丽 A polymerase (Takara Shuzo, Japan) was used. The obtained amplification product of about 800 bp was digested with EcoRI and Notl, and then purified to obtain an insert DNA. The EcoRI and Notl restriction of the PEAK8 / IRES / EGFP vector, in which an intramolecular liposome entry sequence (IRES) and enhanced green fluorescent protein (EGFf) are linked to the expression vector PEAKS (Edge Biosystem, USA). It was inserted into the enzyme site and cloned. The nucleotide sequence of the obtained plasmid, whose sequence is determined and encodes the mutant FGF-23 protein with the desired Arg at positions 176 and 179 converted to Gin and a His6-tag sequence added to the C-terminus I was sure that . This vector is called PEAK8 / IRES / EGFP / FGF-23RQH.
15- (4) FGF- 23H発現細胞、 FGF- 23発現細胞、 および FGF- 23RQH発現細胞の取得、  15- (4) Obtaining FGF-23H-expressing cells, FGF-23-expressing cells, and FGF-23RQH-expressing cells,
(i) FGF- 23H発現細胞の取得 (i) Obtaining FGF-23H-expressing cells
約 20 g の pcDNAFGF- 23H をベクター中のアンピシリン耐性遺伝子内にある About 20 g of pcDNAFGF-23H in the ampicillin resistance gene in the vector
Fspl制限酵素部位で切断して直鎖化し、精製した。これを 10 1の純水に溶解し、It was cut at the Fspl restriction enzyme site, linearized, and purified. Dissolve this in 10 1 pure water,
1ズ107個の(^01^5 (10116-1細胞( Shirahata, S. , et al. Biosci Biotech Biochem, 59: 345-347, 1995 ) と混和して Gene Pulser II (米国、 Bio Rad社) を用いて 電気穿孔法にて細胞への遺伝子導入を行った。 この細胞 10% FCSを含む MEM ο;培 養液 (米国、 Gibco BRL社) で 24時間培養したのち、 終濃度 0.5mg/ml となるよ うに Zeocin (米国、 Invitrogen社) を添加して 1週間培養した。 接着し増殖した 細胞をトリプシンで遊離して、 終濃度 0.3mg/mlの Zeocin存在下で限界希釈法に よるクローニングを行い、 35 種のクローン化細胞を得た。 これらの細胞の中でOne cell contains 107 (^ 01 ^ 5 (10116-1 cells (Shirahata, S., et al. Biosci Biotech Biochem, 59: 345-347, 1995), and the gene was introduced into cells by electroporation using Gene Pulser II (Bio Rad, USA). After culturing the cells for 24 hours in a MEM ο; culture medium (Gibco BRL, USA) containing 10% FCS, add Zeocin (Invitrogen, USA) to a final concentration of 0.5 mg / ml for 1 week Cultured. The cells that adhered and proliferated were released with trypsin and cloned by the limiting dilution method in the presence of Zeocin at a final concentration of 0.3 mg / ml to obtain 35 cloned cells. In these cells
FGF-23H 夕ンパク質を最もよく発現する細胞を以下に示すウェスタンプロッティ ングにて同定した。 各クローン化細胞の培養上清を採取して、 SDS-ポリアクリフレ アミド電気泳動を行ったのち、 PVDF膜 (米国、 Millipore社) にタンパク質を転 写し、 抗 His- tag (C末) 抗体 (米国、 Invitrogen社) と ECL発光システム (米 国、 Amersham Pharmacia Biotech社) を用いて約 32kDa付近の FGF- 23Hタンパク 質に由来するシグナルを検出した。 その結果、 #20 と称すクローンにおいて最も 高い発現が認められ、 これを CH0-0ST311Hと命名して 2000年 8月 1 1日付で、独 立行政法人産業技術総合研究所、 特許生物寄託センター (日本国茨城県つくば巿 東 1丁目 1番地 1 中央第 6) に寄託した (受託番号: FERM BP- 7273)。 本明細書 では、 CH0- 0ST311Hを CHO-FGF23Hと称する。 Cells that best expressed FGF-23H protein were identified by Western blotting as described below. After collecting the culture supernatant of each cloned cell and performing SDS-polyacrylamide electrophoresis, the protein was transferred to a PVDF membrane (Millipore, USA), and an anti-His-tag (C-terminal) antibody (US, Invitrogen) and an ECL luminescence system (Amersham Pharmacia Biotech, USA) were used to detect a signal derived from the FGF-23H protein of about 32 kDa. As a result, the highest expression was observed in the clone named # 20, which was named CH0-0ST311H and was filed on August 11, 2000 by the National Institute of Advanced Industrial Science and Technology, the Patent Organism Depositary Center (Japan). Deposited at Tsukuba, Higashi 1-chome, 1-1, Chuo No. 6), Ibaraki, Japan (Accession number: FERM BP-7273). In this specification, CH0-0ST311H is referred to as CHO-FGF23H.
(ii) FGF-23および FGF-23RQH発現細胞の取得  (ii) Obtaining FGF-23 and FGF-23RQH expressing cells
PEAK8/I ES/EGFP/FGF-23 と EAK8/IRES/EGFP/FGF-23RQH ベクターの CH0 Ras clone- 1細胞への導入は膜融合脂質を用いた遺伝子導入法により行った。 CHORas clone-1細胞を 6- well plateに底面の約 60%を細胞が覆う程度に培養する。そし て培養液を除去し、 血清を含まない ΜΕΜα培養液を lml添加する。 導入するべク 夕一 2.5 gと IO の Transfectam (登録商標) (米国、 Promega社) をそれぞれ The PEAK8 / IES / EGFP / FGF-23 and EAK8 / IRES / EGFP / FGF-23RQH vectors were introduced into CH0 Ras clone-1 cells by the gene transfer method using membrane fusion lipid. Culture CHORas clone-1 cells in a 6-well plate so that the cells cover about 60% of the bottom surface. Then remove the culture medium and add 1 ml of serum-free ΜΕΜα culture medium. Introduce 2.5 g Yuichi and IO Transfectam (registered trademark) (Promega, USA)
50^1の血清を含まない ΜΕΜα培養液と混和して、 両者を混合して 10分間静置し たのち、 両者を混合して準備しておいた 6- well plateに添加した。 2時間培養し た後、 この DNAを含む培養液を除去して 10%の FCSを含む培養液に置換して終夜 培養した。 翌日、 終濃度が S^g/ml となるように Puromycin (米国、 Sigma社) を 添加して、 薬剤耐性細胞を選択した。 この様にして得られた薬剤耐性細胞は前述 の FGF-23H発現細胞の取得と同様に限界希釈法にてクローン化を行った。 さらに ウエスタンプロッティングにより目的のタンパク質を最もよく発現する細胞株を 取得した。 これらの細胞をそれぞれ、 CH0 - FGF23、 CH0-FGF23RQと称す。 (i i i) 組換え体タンパク質の精製 The mixture was mixed with 50 ^ 1 serum-free ΜΕΜα culture medium, mixed, and allowed to stand for 10 minutes. Then, both were mixed and added to a prepared 6-well plate. After culturing for 2 hours, the culture solution containing the DNA was removed, replaced with a culture solution containing 10% FCS, and cultured overnight. The next day, drug-resistant cells were selected by adding Puromycin (Sigma, USA) to a final concentration of S ^ g / ml. The drug-resistant cells thus obtained were cloned by the limiting dilution method in the same manner as in the above-mentioned acquisition of FGF-23H-expressing cells. Furthermore, a cell line that best expresses the target protein was obtained by western blotting. These cells are called CH0-FGF23 and CH0-FGF23RQ, respectively. (iii) Purification of recombinant protein
CH0-FGF23Hの培養上清中の組換え体を C末端の His6- tag配列に対する抗体を 用いてウエスタンブロッテイングにて検出すると、 図 1 0 A に示すように 32kDa 付近のバンドと l OkDa付近のバンドが認められた。 この二つのバンドをゲルから 切り出し N末端側のアミノ酸配列を決定したところ、 分子量の大きい方のバンド は配列番号 3 6の 25番目からのアミノ酸配列が検出され、 FGF- 23タンパク質か ら分泌過程でシグナル配列が除去されたものと考えられた。 一方、 分子量の小さ いバンドからは配列番号 3 6の 180番目からのアミノ酸配列が確認され、 179番 目と 180番目の間での切断により生じた断片であることが判明した。また、 FGF - 23 の N末側を認識するポリクロ一ナル抗体を用いて検出することで、 179番目まで の配列を持つと考えられるペプチドの存在も認められた。  When the recombinant in the culture supernatant of CH0-FGF23H was detected by Western blotting using an antibody against the His6-tag sequence at the C-terminus, a band around 32 kDa and a band near lOkDa were observed as shown in Figure 10A. A band was observed. When these two bands were cut out of the gel and the amino acid sequence at the N-terminal side was determined, the band with the larger molecular weight detected the amino acid sequence from the 25th position in SEQ ID NO: 36, and was secreted from the FGF-23 protein during secretion. It was considered that the signal sequence had been removed. On the other hand, the band with a small molecular weight confirmed the amino acid sequence from the 180th position of SEQ ID NO: 36, and was found to be a fragment generated by cleavage between the 179th position and the 180th position. In addition, by using a polyclonal antibody recognizing the N-terminal side of FGF-23, the presence of a peptide thought to have a sequence up to the 179th position was also confirmed.
CHO- FGF23H細胞の培養上清 1000mlを 200gで 15分間、 4°Cで遠心分離し、 浮遊細胞を除去した後、 上清を内径 30 腿 X長さ 200 丽 のカラムに充填した SP- sepharose FF (登録商標) (米国、 Amersham Pharmacia Biot ech 社) に通すこ とで、配列番号 3 6の 180から 251番目に相当するペプチドに Hi s6- t agが付加し たペプチドが素通りし、 25〜251番目のペプチド (以下、 全長 FGF- 23タンパク質 ということがある)に His6- tag配列を付加したものはカラムに吸着した。 この力 ラムに 50 mM リン酸ナトリウム緩衝液 (pH6. 7) 中で 0から 0. 7 Mまでの NaC l濃 度勾配で吸着物質を溶出させたところ、 約 0. 3M の NaC l で溶出される画分に His6- tagが付加した全長 FGF- 23タンパク質が認められ、約 0. 4Mで溶出される画 分に配列番号 3 6の 179番目より N末側の配列を持つと考えられるペプチドが確 認された。 このように SP- Sepharoseカラムで分離された画分は次に、金属ァフィ 二ティカラム、 Tal on Superilow (登録商標) (米国、 Clontech社) にかけること でさらに分離する事が出来た。 179番目より N末側の配列についても金属カラム との親和性を有しており、精製に有効であった。 さらに SP-Sepharoseカラムにて 精製を行い、 CBB染色で単一のバンドとして全長の FGF- 23Hを取得する'ことがで きた。 この結果を図 1 0 Bに示す。  After centrifuging 1000 ml of the culture supernatant of CHO-FGF23H cells at 200 g for 15 minutes at 4 ° C to remove the suspended cells, the supernatant was packed into a column of 30 t ID x 200 mm length SP- sepharose FF (Registered trademark) (Amersham Pharmacia Biotech, U.S.A.), a peptide having His6-tag added to the peptide corresponding to positions 180 to 251 of SEQ ID NO: 36 passed through, and passed through 25 to 251. The sixth peptide (hereinafter, sometimes referred to as full-length FGF-23 protein) with a His6-tag sequence added was adsorbed to the column. When the adsorbed material was eluted with 50 mM sodium phosphate buffer (pH 6.7) with a NaCl concentration gradient of 0 to 0.7 M, the eluate was eluted with approximately 0.3 M NaCl. In this fraction, the full-length FGF-23 protein with His6-tag added was observed, and in the fraction eluted at about 0.4M, a peptide thought to have an N-terminal sequence from the 179th position of SEQ ID NO: 36 was found. confirmed. The fraction thus separated on the SP-Sepharose column was then further separated by applying to a metal affinity column, Tal on Superilow (registered trademark) (Clontech, USA). The N-terminal sequence from the 179th position also had an affinity for the metal column and was effective for purification. Furthermore, purification was performed using an SP-Sepharose column, and it was possible to obtain full-length FGF-23H as a single band by CBB staining. The result is shown in FIG. 10B.
FGF - 23タンパク質も同様の方法で精製することができる。 CH0- FGF23の培養上 清をポアサイズが 0. 2 mのメンブレンである SuperCap (登録商標) (米国、 Pal l The FGF-23 protein can be purified in a similar manner. The culture supernatant of CH0-FGF23 was purified using SuperCap (registered trademark), a membrane having a pore size of 0.2 m (Pall, USA).
Ge lman Laboratory社) でろ過し、 ろ過された溶液を SP- Sepharose FF (米国、 Amersham Pharmacia Biotech社) に通した。 カラムとの親和性が弱い物質を 50ιηΜ のリン酸ナトリウム緩衝液, ρΗ6.7で洗浄、 溶出させた。 カラム保持されたタンパ ク質を 0から 0.7Μまでの NaCl濃度勾配で溶出させたところ、 約 0.3Mの NaClで 溶出される画分に全長 FGF-23タンパク質が認められた。これを金属ァフィ二ティ カラムである Talon Superilow (登録商標) (米国、 Clontech社) に吸着させたの ち、 50mMのリン酸ナトリウム緩衝液, pH6.7で洗浄したのち、 Imidazoleの濃度を 変化させて添加しタンパク質を溶出精製した。 さらに、 目的のタンパク質を含む 画分を SP Sep arose FF カラムに吸着、 溶出させて精製した。 同様な方法で CHO-FGF23RQ上清より全長 FGF- 23RQ夕ンパク質を精製した。 Gel-man Laboratory), and the filtered solution is SP- Sepharose FF (US, Amersham Pharmacia Biotech). Substances with low affinity for the column were washed and eluted with 50ιηΜ sodium phosphate buffer, ρΗ6.7. When the protein retained on the column was eluted with a gradient of NaCl from 0 to 0.7Μ, full-length FGF-23 protein was observed in the fraction eluted with about 0.3 M NaCl. This was adsorbed to Talon Superilow (registered trademark) (Clontech, USA), a metal affinity column, washed with 50 mM sodium phosphate buffer, pH 6.7, and then the concentration of Imidazole was changed. And the protein was eluted and purified. Further, the fraction containing the target protein was adsorbed and eluted on a SP Separose FF column and purified. In the same manner, full-length FGF-23RQ protein was purified from the CHO-FGF23RQ supernatant.
〔実施例 16〕 ヒト FGF- 23に対するヒトモノクローナル抗体産生ハイプリドー マの取得  [Example 16] Obtaining human monoclonal antibody-producing hybridoma against human FGF-23
本実施例におけるモノクローナル抗体の作製は、 単クローン抗体実験操作入門 (安東民衛ら著作、講談社発行 1991)等に記載されるような一般的方法に従って 調製した。 被免疫動物は、 BALB/cマウスを用い、 ヒト FGF- 23の免疫は免疫原の 違いにより以下の 2種類の方法で行なった。  The preparation of the monoclonal antibody in this example was prepared according to a general method as described in "Introduction to Monoclonal Antibody Experimental Procedures" (written by Tamoe Ando et al., Published by Kodansha 1991). BALB / c mice were used as immunized animals, and human FGF-23 was immunized by the following two methods depending on the difference in immunogen.
16- (1) ベクタ一投与と組換え体夕ンパク質投与の組み合わせによる免疫 16- (1) Immunization by a combination of single vector administration and recombinant protein administration
BALB/cマウスに、 実施例 15- (1)で作製した INPEP4/FGF-23ベクター (10またはThe INPEP4 / FGF-23 vector prepared in Example 15- (1) (10 or
50 g/EE) を Trans IT (登録商標) In Vivo Gene Del ivery System試薬 (日本、 宝酒造社) を静脈より導入して初回免疫を実施した。 初回免疫から同ベクターを50 g / EE) was transvenously introduced with Trans IT (registered trademark) In Vivo Gene Delivery System reagent (Takara Shuzo Co., Ltd., Japan) to perform the first immunization. The same vector from the first immunization
1週間後に 1回導入し追加免疫した。さらに、実施例 15- (4)で作製した FGF- 23RQH タンパク質(20〜30 g/匹)をスクアレン、 Tween80、 Monophosp oryl lipid A および Trehalose dimycolateを含む RIBIアジュバント (米国、 Corixa社) に懸 濁しェマルジヨンを調製した後、 腹腔内注射により 4回または 5回追加免疫し、 さらに以下に述べる脾臓細胞の取得 4日前に実施例 15- (4)で作製した FGF-23H夕 ンパク質(18 g/匹)を尾静脈内注射により免疫した。 One week later, it was introduced once and boosted. Further, the FGF-23RQH protein (20 to 30 g / animal) prepared in Example 15- (4) was suspended in RIBI adjuvant (Corixa, USA) containing squalene, Tween80, Monophosporyl lipid A and Trehalose dimycolate, and emulsion was added. Was prepared, and boosted four or five times by intraperitoneal injection, and the FGF-23H protein (18 g / animal) prepared in Example 15- (4) four days before the acquisition of the following spleen cells ) Was immunized by tail vein injection.
16- (2) ヒト FGF- 23を用いた免疫  16- (2) Immunization using human FGF-23
BALB/cマウスに、 実施例 15-(4)で作製した?6?-23 (22 /匹)を上述の 81 アジュバントに懸濁して腹腔内注射により初回免疫した。 さらに、 同タンパク質 を腹腔内注射により 1週間毎に 1回、 4週間にわたり追加免疫し、 以下に述べる 脾臓細胞の取得 3日前に FGF - 23(10 z g/匹)を尾静脈内注射により免疫した。 16- (3) ハイプリドーマの作製と選別 Was it prepared in Example 15- (4) for BALB / c mice? 6? -23 (22 / animal) were suspended in the above-mentioned 81 adjuvant and primed by intraperitoneal injection. In addition, the same protein was boosted once a week by intraperitoneal injection for 4 weeks, and FGF-23 (10 zg / animal) was immunized by tail vein injection 3 days before the acquisition of spleen cells described below. . 16- (3) Preparation and selection of high prioma
上述のように免疫したマウスから脾臓を摘出し、 それより回収した脾臓細胞を マウスミエローマ SP2/0 (ATCC: CRL 1581) と 5 : 1で混合し、 融合剤としてポリ エチレングリコール 1500 (日本、 ロッシュダイァグノステイクス社) を用いて細 胞融合させ、 ハイプリドーマを作製した。 ハイプリドーマの選択は、 10%のゥシ胎 児血清 (Fet al Cal f SermiK FCS) とヒポキサンチン(H)、 アミノプテリン(A)、 チ ミジン(T)を含有する HAT含有雇 EM培地 (米国、 Gibco BRL社) 中で培養するこ とにより行った。 さらに、 HT含有 DMEM培地を用いて限界希釈法によりクロー二 ングを実施した。 単一細胞に由来するクローン化ハイプリドーマを取得した。 16- (4) 抗 FGF-23抗体を産生するクローン化したハイプリドーマの選別  The spleen was excised from the mouse immunized as described above, and the spleen cells collected from the spleen were mixed with mouse myeloma SP2 / 0 (ATCC: CRL 1581) at a ratio of 5: 1, and polyethylene glycol 1500 (Roche, Japan) was used as a fusion agent. The cells were fused using Diagnostics, Inc. to produce hybridomas. The selection of hybridomas was carried out using a HAT medium containing HAT containing 10% fetal calf serum (Fetal Calf SermiK FCS) and hypoxanthine (H), aminopterin (A) and thymidine (T) (US , Gibco BRL). Furthermore, cloning was performed by a limiting dilution method using HT-containing DMEM medium. A cloned hybridoma derived from a single cell was obtained. 16- (4) Selection of cloned hybridoma producing anti-FGF-23 antibody
ハイプリドーマが産生する抗体と FGF- 23 タンパク質との結合性を調べること で、 FGF- 23タンパク質を特異的に認識する抗体を産生するハイプリドーマを選別 した。 上述の第一の方法で免疫して得たハイプリドーマの選別は以下のように実 施した。 50mM NaHC03の溶液に 1 z g/mlの濃度に希釈した FGF- 23Hタンパク質溶液 を、 ELISA用 96穴マイクロプレート (Maxisorp (登録商標)、 米国、 Nunc社) の 各ゥエルに 50 1ずつ加え、 37°Cで 30分または 4 °Cで 12時間ィンキュベートし、By examining the binding property between the antibody produced by the hybridoma and the FGF-23 protein, the hybridoma producing an antibody that specifically recognizes the FGF-23 protein was selected. The selection of hybridomas obtained by immunization according to the first method described above was carried out as follows. Diluted to a concentration of 1 zg / ml in a solution of 50 mM NaHCO 3 FGF the 23H protein solution, a 96-well microplate for ELISA (Maxisorp (TM), USA, Nunc, Inc.) was added in 50 1 in each Ueru of 37 Incubate for 30 minutes at ° C or 12 hours at 4 ° C,
FGF-23H タンパク質をマイクロプレートに吸着させた。 次にこの溶液を除去し、 各ゥエルにブロッキング試薬 (SuperBIock (登録商標) Blocking Buf fer,米国、The FGF-23H protein was adsorbed on the microplate. The solution was then removed and each well was filled with a blocking reagent (SuperBIock® Blocking Buf fer, USA,
PIERCE社) を加え室温で 30 分間インキュベートしたのち、 各ゥエルを 0. 1¾の(PIERCE) and incubate at room temperature for 30 minutes.
Tween20 を含有する Tri s- buf fered sal ine (500mM NaC l 含有 TRIZMA pre - se t crys t al s (登録商標)米国、 Sigma社)(T- TBS)で 2回洗浄した。このように FGF- 23H タンパク質をコーティングしたマイクロプレートの各ゥエルに、 各々のハイブリ ドーマの培養上清を 50 1加え、 30分間反応させた後、 各ゥエルを、 T- TBSで 2 回洗浄した。次いで、各ゥエルに 50 1ずつ 3000倍希釈した過酸化酵素標識ャギ 抗マウス IgG抗体 (米国、 Zymed l aboratori es社) を加え、 室温下で 30分間ィ ンキュペートした。 これを T- TBSで 3回洗浄後、 テトラメチルベンジジン (デン マーク、 DAK0社) を含む基質緩衝液を各ゥエルに 50 1ずつ加え、 室温下で 15 分間インキュベートした。 次いで、 0. 5M硫酸を各ゥエルに 50 1ずつ加え、 反応 を止めた。 参照波長を 570nmとして波長 450nmでの吸光度をマイクロプレートリ ーダー (MTP - 300、 日本、 コロナ電気社) で測定した。 ここで、 明確な吸光度の上 昇を示したハイプリドーマを選別し、 さらに、 FGF- 23タンパク質を用いて同様の 実験を実施して FGF-23との結合が再確認されたクローンを選別した。 これより、 FGF-23 タンパク質を認識する抗体を産生するハイプリ ドーマとして 9種のクロ ーンを得た。 この中には後に示す 1C3H、 1D6A、 2A2B、 2C3B、 2C5Lが含まれていた。 上述の第二の方法で免疫して得たハイプリ ドーマの選別は、 以下のように実施し た。 50mM NaHC03の溶液に l /i g/ml の濃度に希釈した FGF- 23タンパク質溶液を、The plate was washed twice with Tris-buf fered saline containing Tween20 (TRIZMA pre-setcrys tals (registered trademark, Sigma, USA) containing 500 mM NaCl) (T-TBS). To each well of the microplate coated with the FGF-23H protein in this manner, 50 1 of the culture supernatant of each hybridoma was added and reacted for 30 minutes. Then, each well was washed twice with T-TBS. Then, a peroxidase-labeled goat anti-mouse IgG antibody (Zymed Laboratories, USA) diluted 3000-fold to 50: 1 was added to each well, and incubated at room temperature for 30 minutes. After washing the plate three times with T-TBS, 50 1 of a substrate buffer containing tetramethylbenzidine (Denmark, DAK0) was added to each well, and the mixture was incubated at room temperature for 15 minutes. Then, 0.5 M sulfuric acid was added to each well at a rate of 50 1 to stop the reaction. The absorbance at a wavelength of 450 nm was measured using a microplate reader (MTP-300, Corona Electric, Japan) with a reference wavelength of 570 nm. Where the clear absorbance The hybridoma showing the rise was selected, and a similar experiment was performed using FGF-23 protein to select a clone in which the binding to FGF-23 was reconfirmed. As a result, nine clones were obtained as hybridomas producing antibodies recognizing the FGF-23 protein. These included 1C3H, 1D6A, 2A2B, 2C3B, and 2C5L, which will be described later. The selection of hybridomas obtained by immunization according to the second method described above was carried out as follows. Diluted in a solution of 50 mM NaHCO 3 to a concentration of l / ig / ml FGF- 23 protein solution,
ELI SA用 96穴マイクロプレート (Max i sorp (登録商標)、 米国、 Nunc社) の各ゥ エルに 50 i lずつ加え、 4 °Cで 10時間ィンキュベートして FGF- 23タンパク質を マイクロプレートに吸着させた。 次にこの'溶液を除去し、 各ゥエルにブロッキン グ試薬 (SuperBI ock (登録商標) Bl ocking Buf fer, 米国、 P IERCE 社) を加え 室温で 30分間ィンキュベートしたのち、 各ゥエルを 0. 1%の Tween20を含有する Tr i s-buf fered sal ine (T-TBS) で 2回洗浄した。 このように FGF- 23タンパク質 をコーティングしたマイクロプレー卜の各ゥエルに、 各々のハイブリドーマの培 養上清を 50 1加え、 30分間反応させた後、 各ゥエルを、 0. 1%の Tween20を含有 する Tr i s-bui iered sal ine (T-TBS) で 2回洗浄した。 次いで、 各ゥエルに ずつ 3000 倍希釈した過酸化酵素標識ャギ抗マウス IgG 抗体 (米国、 Zymed l aborat or ies社) を加え、 室温下で 30分間インキュベートした。 これを T- TBS で 3回洗浄後、 テトラメチルベンジジン (デンマーク、 DAK0社) を含む基質緩衝 液を各ゥエルに 50 ずつ加え、 室温下で 15分間インキュベートした。 次いで、 0. 5M硫酸を各ゥエルに ずつ加え、 反応を止めた。 参照波長を 570nmとして 波長 450 ηιηでの吸光度をマイクロプレートリーダ— (MTP- 300、 日本、 コロナ電 気社) で測定した。 ここで、 明確な吸光度の上昇を示したハイプリドーマを選別 した。 これより、 FGF- 23タンパク質を認識する抗体を産生するハイプリ ドーマと して新たに 4種のクローンを得た。 この中に 3C 1Eが含まれていた。 Add 50 il to each well of a 96-well microplate for ELI SA (Maxisorp (registered trademark), Nunc, USA) and incubate at 4 ° C for 10 hours to allow the FGF-23 protein to adsorb to the microplate. Was. Next, the solution was removed, a blocking reagent (SuperBIock (registered trademark) Blocking Buf fer, Pierce, USA) was added to each well, and the mixture was incubated at room temperature for 30 minutes. Was washed twice with Tris-buf fered saline (T-TBS) containing Tween20. To each well of the microplate coated with FGF-23 protein in this manner, add 50 1 of the culture supernatant of each hybridoma and react for 30 minutes.After that, each well contains 0.1% Tween20. Washed twice with Tris-bui iered saline (T-TBS). Next, peroxidase-labeled goat anti-mouse IgG antibody (Zymed Laboratories, USA) diluted 3000-fold was added to each well, and incubated at room temperature for 30 minutes. After washing the plate three times with T-TBS, 50 ml of a substrate buffer containing tetramethylbenzidine (DAK0, Denmark) was added to each well, and the mixture was incubated at room temperature for 15 minutes. Next, 0.5 M sulfuric acid was added to each well to stop the reaction. The absorbance at a wavelength of 450 ηιη was measured using a microplate reader (MTP-300, Corona Electric, Japan) with a reference wavelength of 570 nm. Here, hybridomas showing a clear increase in absorbance were selected. Thus, four new clones were obtained as hybridomas producing antibodies recognizing the FGF-23 protein. This included 3C 1E.
このように取得した FGF- 23 タンパク質を特異的に認識する抗体のサブクラス を I so St rip マウスモノクローナル抗体アイソタイピングキット (米国、 Roche 社) を用いて同定した。 結果は表 1に示した。 抗ヒト FGF-23抗体
Figure imgf000048_0001
The subclass of the antibody specifically recognizing the FGF-23 protein obtained in this manner was identified using an Isotrip mouse monoclonal antibody isotyping kit (Roche, USA). The results are shown in Table 1. Anti-human FGF-23 antibody
Figure imgf000048_0001
1D6A IgG1(/c ) 3.21  1D6A IgG1 (/ c) 3.21
2A2B IgG1(/c ) 2.67  2A2B IgG1 (/ c) 2.67
2C3B IgG1(/c ). 1.21  2C3B IgG1 (/ c). 1.21
3C1E IgG1(/ ) 3.5以上  3C1E IgG1 (/) 3.5 or more
2C5L IgG1(/ ) 1.38 上記ハイプリドーマクローンのうち、 3つのハイプリドーマクローン 2C3B、3C 1 E、 1D6Aを、 2001年 12月 26日付で、独立行政法人 産業技術総合研究所 特許生物寄 託センター (茨城県つくば市東 1丁目 1番地 1中央第 6 ) にブダペスト条約に基 づき国際寄託した。 また、 ハイブリ ド一マクローン 2C5Lを、 2003年 1月 6 日付 で、 独立行政法人 産業技術総合研究所 特許生物寄託センター (茨城県つくば市 東 1丁目 1番地 1中央第 6 ) にブダペスト条約に基づき国際寄託した。 受託番号 は以下の通りである。  2C5L IgG1 (/) 1.38 Of the above hybridoma clones, three hybridoma clones, 2C3B, 3C1E, and 1D6A, were obtained on December 26, 2001 by the National Institute of Advanced Industrial Science and Technology, Patent Organism Depositary Center (Ibaraki, Japan). Based on the Budapest Treaty, an international deposit was made at Higashi 1-chome, 1-chome, 1-Chuo 6). Based on the Budapest Treaty, the Hybrid Maclone 2C5L was registered on January 6, 2003 with the National Institute of Advanced Industrial Science and Technology (AIST) at the Patent Organism Depositary Center (1-1 1-1 Higashi 1-Chuo 6th, Tsukuba City, Ibaraki Prefecture). Deposited. The accession numbers are as follows.
2C3B : FERM BP- 7838  2C3B: FERM BP-7838
3C 1E : FERM BP-7839  3C 1E: FERM BP-7839
1D6A : FERM BP- 7840  1D6A: FERM BP-7840
2C5L : FERM BP-8268  2C5L: FERM BP-8268
〔実施例 1 7〕 モノクローナル抗体の調製  [Example 17] Preparation of monoclonal antibody
17- (1) 抗 FGF-23抗体を含む培養上清の調製 17- (1) Preparation of culture supernatant containing anti-FGF-23 antibody
抗 FGF- 23抗体産生ハイプリドーマを 10 g/mlのゥシインシュリン(米国、 S igma 社)、 5. 5 g/ml のヒトトランスフェリン (米国、 Sigma社)、 0. O lmMのェタノ一 ルァミン (米国、 Sigma社)、 5ng/mlの亜セレン酸ナトリウム (米国、 Sigma社) を含有する eRDF培地 (日本、 極東製薬社) に馴化した。 抗体を調製するための八 イブリドーマの培養はスピナ—フラスコで実施した。培養液を孔径が 0. 2 X mのフ ィルター (米国、 Pal l Gelman Laboratory社) を通すことでハイブリ ドーマ等の 雑排物を除去し、 抗体を含む培養上清を回収した。  The anti-FGF-23 antibody-producing hybridoma was ligated with 10 g / ml of periinsulin (Sigma, USA), 5.5 g / ml of human transferrin (Sigma, USA), and 0.1 lmM of ethanolamine ( Adapted to eRDF medium (Farto Pharmaceuticals, Japan) containing 5 ng / ml sodium selenite (Sigma, USA). Culture of the eight hybridomas to prepare antibodies was performed in spinner flasks. The culture solution was passed through a filter having a pore size of 0.2 Xm (Pall Gelman Laboratory, USA) to remove foreign substances such as hybridomas, and the antibody-containing culture supernatant was recovered.
17- (2) プロティン Gを用いたモノクローナル抗体の精製 17- (2) Purification of monoclonal antibody using protein G
抗 FGF- 23抗体を含む培養上清をプロテイン G Sepharose 4 FFカラム (米国、 The culture supernatant containing the anti-FGF-23 antibody was applied to a protein G Sepharose 4 FF column (US,
Amers ani Pharmac i a Biotech社) に通して抗体をカラムに吸着させ、 0. 1M グリ シン緩衝液 (PH2. 8) を用いて溶出させた。 溶出画分は 1 M Tri s_HCl を添加して pH7. 2に調整した。 このように調製した抗体溶液を分画分子量 10000カツトの透 析膜(米国、 Spect rum Laborator i es社) を用いて PBS (-)で透析置換し、孔径 0. 22 mのメンブランフィルタ一 MILLEX-GV (米国、 Mi l l ipore社) でろ過滅菌し、 精 製抗 FGF- 23抗体を得た。 精製抗体の濃度は 280nmの吸光度を測定し、 lmg/ml を 1. 350Dとして算出した。 The antibody was adsorbed onto the column through Amersani Pharmacia Biotech) and eluted with 0.1 M glycine buffer (PH2.8). The eluted fraction is added with 1 M Tris_HCl. The pH was adjusted to 7.2. The antibody solution thus prepared was subjected to dialysis substitution with PBS (-) using a membrane with a cut-off molecular weight of 10,000 cuts (Spectrum Laboratories, U.S.A.), and a 0.22 m membrane filter MILLEX- The solution was sterilized by filtration with GV (Millipore, USA) to obtain a purified anti-FGF-23 antibody. The concentration of the purified antibody was calculated by measuring absorbance at 280 nm and setting lmg / ml to 1.350D.
17- (3) プロティン Aを用いたモノクローナル抗体の精製  17- (3) Purification of monoclonal antibody using protein A
プロテイン A担体カラム (日本、 免疫生物研究所社) を用い、 吸着緩衝液とし てグリシン緩衝液(pH8. 9)、 溶出緩衝液としてクェン酸緩衝液 (pH 4. 0) を用いて 抗 FGF- 23抗体を含む培養上清から抗体をァフィ二ティ一精製した。この抗体を含 む溶出画分に 1M Tris-HClを添加して pH7. 2付近に調整した。 次に透析膜を用い てこの抗体を含む溶液を PBS (-)に置換し、 さらに孔径 0. 22 mのメンブランフィ ルターでろ過滅菌し、精製抗 FGF- 23抗体を得た。精製抗体の濃度は 280mnの吸光 度を測定し、 lmg/mlを 1. 350Dとして算出した。  Using a protein A carrier column (Immune Biological Laboratories, Japan), glycine buffer (pH 8.9) as the adsorption buffer, and citrate buffer (pH 4.0) as the elution buffer, anti-FGF- The antibody was affinity purified from the culture supernatant containing the 23 antibody. 1M Tris-HCl was added to the eluted fraction containing this antibody to adjust the pH to around 7.2. Next, the solution containing the antibody was replaced with PBS (-) using a dialysis membrane, and further sterilized by filtration with a membrane filter having a pore size of 0.22 m to obtain a purified anti-FGF-23 antibody. The concentration of the purified antibody was calculated by measuring the absorbance at 280 mn and setting lmg / ml to 1.350D.
このようにして得られた各精製モノクローナル抗体を、 産生するハイプリドー マの名前を用いて示すことにする。例えばハイプリドーマ 1D6Aが産生する抗体に 関しては 1D6A抗体と記載することにする。  Each of the purified monoclonal antibodies thus obtained will be indicated using the name of the produced hybridoma. For example, an antibody produced by hybridoma 1D6A will be referred to as 1D6A antibody.
〔実施例 1 8〕 EGR 1プロモータ ·ルシフェラーゼを用いた klothoタンパク質 発現 PEAK細胞でのアツセィ系での抗 FGF- 23抗体の影響  [Example 18] Effect of anti-FGF-23 antibody in Atsushi system on PEAK cells expressing klotho protein using EGR1 promoter-luciferase
抗 FGF-23 抗体の中和活性能が実施例 9のアツセィ系で測定できるかどうかを 調べる為に、 実施例 1 5で作製した抗 FGF-23抗体 (2C3B) を用いて検討した。 方 法は実施例 9に従って行ない、 FGF- 23添加直前に 2C3B抗体を最終濃度 2 . 5 I g /mlになるように添加した。その結果、 2C3B抗体は FGF- 23によるルシフェラー ゼの上昇を有意に抑制した (図 1 1 )。 このことから、 EGR 1プロモー夕 ·ルシフ エラーゼを用いた klotho夕ンパク質発現細胞でのアツセィ系は FGF- 23と klotho タンパク質のシグナル伝達を抑制するような物質の探索に有効であることが示さ れた。  In order to examine whether the neutralizing activity of the anti-FGF-23 antibody could be measured by the Atsushi system of Example 9, the anti-FGF-23 antibody (2C3B) prepared in Example 15 was examined. The method was performed according to Example 9, and the 2C3B antibody was added to a final concentration of 2.5 Ig / ml immediately before the addition of FGF-23. As a result, the 2C3B antibody significantly suppressed the increase in luciferase caused by FGF-23 (FIG. 11). This indicates that the Atssey system in klotho protein-expressing cells using EGR-1 Promoter-Luciferase is effective in searching for substances that suppress signal transduction of FGF-23 and klotho protein. Was.
〔実施例 1 9〕 FGF-23と可溶化型 klothoタンパク質による CHOras c lone l細 胞の ERKリン酸化亢進のアツセィ系への抗 FGF- 23抗体の影響  [Example 19] Effect of anti-FGF-23 antibody on Atsuyi system in enhancing ERK phosphorylation of CHO ras clone cells by FGF-23 and soluble klotho protein
抗 FGF- 23抗体の中和?舌性能が実施例 8のアツセィ系で測定できるかどうかを調 ベる為に、 実施例 1 5で作製した抗 FGF- 23抗体 (2C3B) を用いて検討した。 方法は実施例 8にしたがって行ない、 FGF- 23と 2 C 3 B抗体をあらかじめ混合し た状態で細胞へ添加した。その結果、 図 1 2に示すように 2C3B抗体濃度依存的に FGF-23と可溶化型 kl 0 tlioタンパク質による ERKリン酸化亢進を抑制した。 この ことから、 FGF-23と kl othoタンパク質を用いた ERKリン酸化亢進検出のアツセ ィ系は FGF- 23と kl'o thoタンパク質シグナル伝達を抑制するような物質の探索に 有効であることが示された。 Neutralization of anti-FGF-23 antibody? To determine whether tongue performance could be measured with the Atsushi system of Example 8, an investigation was conducted using the anti-FGF-23 antibody (2C3B) prepared in Example 15. The method was carried out according to Example 8, and FGF-23 and 2C3B antibodies were added to the cells in a premixed state. As a result, as shown in FIG. 12, the enhancement of ERK phosphorylation by FGF-23 and the soluble k10 tlio protein was suppressed in a 2C3B antibody concentration-dependent manner. This suggests that the assay system for detecting ERK phosphorylation enhancement using FGF-23 and klotho protein is effective in searching for substances that suppress FGF-23 and kl'otho protein signaling. Was done.
〔実施例 2 0〕 抗 FGF- 23抗体" 2C3B抗体"のマウス投与における効果 抗ヒト FGF-23モノクローナル抗体の正常マウスへの影響を検討するため、以下の実験を 実施した。正常マウス (BALBA;、 雄、 12週齢) を 4匹ずつからなる 5つの群に無 作為に分別し、 図 1 3に示すように、 第 1群には媒体として PBS を、 第 には 0. 67mg/mlの抗ヒト FGF- 23モノクローナル抗体 (2C3B) を、 また第 3群には対照 として 0. 67mg/mlの抗 TP0モノクローナル抗体を、それぞれ 0. 15mlずつ尾静脈内 に単回投与した。投与から 24時間後に、 エーテル麻酔下、 心採血を実施し、 マイ クロティナ(米国 べクトンディッキンソン社)を用いて血清を分離した。得られ た血清中のリン酸濃度をリン-テストヮコー(日本国 和光純薬社)、 血清 1, 25D濃 度を 1, 25 (OH) 2D RIAキット TFB (米国 ティエフビー社)をそれぞれ用いて添付の 文書に従い測定した。投与から採血までの間、各群毎にプラスチックケージで飼育 し、 リンおよびカルシウムを ずつ含む固形食 CE- 2 (日本国 日本クレア社)およ び水道水を自由摂取させた。  [Example 20] Effect of administration of anti-FGF-23 antibody "2C3B antibody" on mice In order to examine the effect of anti-human FGF-23 monoclonal antibody on normal mice, the following experiment was performed. Normal mice (BALBA; male, 12 weeks old) were randomly sorted into 5 groups of 4 animals each, and as shown in Fig. 13, PBS was used as a vehicle in group 1 and 0 in group 1 A single dose of 0.15 ml each of 67 mg / ml anti-human FGF-23 monoclonal antibody (2C3B) and 0.67 mg / ml anti-TP0 monoclonal antibody as a control was injected into the tail vein. . Twenty-four hours after the administration, cardiac blood was collected under ether anesthesia, and serum was separated using Mycrotina (Becton Dickinson, USA). Phosphoric acid concentration in the obtained serum was measured using Phosphorus-Test Co., Ltd. (Wako Pure Chemical Industries, Japan). Serum 1.25 D concentration was measured using a 1,25 (OH) 2D RIA kit TFB (Tifb, USA). Measured according to documentation. Each group was bred in a plastic cage from administration to blood collection, and fed a solid diet containing phosphorus and calcium CE-2 (CLEA Japan) and tap water freely.
得られた結果を図 1 3に示す。測定値は各群における平均値 +/_標準偏差で表さ れている。 *を付与した群は、 s tudent-t により有意差検定を実施した結果、 媒 体 (PBS) 投与群および抗 TP0抗体投与群の双方に対し、 pく 0. 01を示した群を表 す。  The results obtained are shown in FIG. Measured values are expressed as mean + / _ standard deviation in each group. The group marked with * indicates a group that showed a p <0.01 with respect to both the vehicle (PBS) administration group and the anti-TP0 antibody administration group as a result of a significant difference test using student-t. .
〔実施例 2 1〕 可溶化 kl o thoタンパク質の精製  [Example 21] Purification of solubilized klo tho protein
実施例 5で作製したマウス可溶化 kl o tho タンパク質安定発現細胞をローラー ボトル (米国ファルコン社) にて SFMI I培地 (米国 インビトロジェン社) にて 5日間培養を行なった。培養上清をミリスタック DE60フィルター(米国 ミリポ ァ社) にて浮遊細胞を除去した後、 上清を SART0C0N Casse t e Hyd rosar t Cu t o f f 30000 (ドイツ国 ザルトリウス社)により約 1 0倍濃縮を行なった。濃縮培養上清 を NaOHにより pH7.95に調整後、 Q SepharoseTM FF (米国 アマシャムバイオサイ エンス社)に吸着させた。非特異的吸着物を洗浄バッファ一(20mMTrisHCl (pH8.0) . 150mM NaCl) にて洗浄後、 溶出バッファー (20mM TrisHCl (pH8.0) , 150mM NaCl から 20mMTrisHCl (pH8.0) , 500mM NaClのグラジェント) にて溶出。 次に Phenyl SepharoseTM HP (米国 アマシャムバイオサイエンス社)にサンプルアプライ後、 洗浄バッファー (700mM (NH4) 2S04) にて洗浄を行い、 700〜師 (NH4) 2S04のダラ ジェントにて溶出。 次に Con A SepharoseTM 4B (米国 アマシャムバイオサイエ ンス社)を用いバッファー A(PBS)、バッファー B(0.5MMethyl-alp a-D-Glucoside, 0.5M NaCl , PBS) のステップワイズにて溶出。 さらに Wheat Germ Lectin SepharoseTM 6MB (米国 アマシャムバイオサイエンス社)にてバッファー A (PBS), バッファ一 B (0.5M N-acety卜!) -Glucosamine, 0.5M NaCl , PBS) のステップワイ ズにて溶出を行なった。 その後 DEAE- SepharoseTM FF (米国 アマシャムバイオサ ィエンス社)を用い、 バッファー A (20mM TrisHCl ρΗ8·0)、 バッファー Β (20mM TrisHCl pH8.0 , 0.5M NaCl) のグラジェントにて溶出。 Q SepharoseTM HP を用 ぃバッファー A (lOinM リン酸バッファ一 ρΗ7· 5)、バッファー B (10mM リン酸バッ ファー PH7.5 , 0.5M NaCl) のグラジェントにて溶出。 図 14は精製物の銀染色 および実施例 1 0で作製した抗 klotho 抗体によるウエスタンブロッテイングの 結果である。 The mouse solubilized klo tho protein stably expressing cells prepared in Example 5 were cultured in a roller bottle (Falcon, USA) in SFMI I medium (Invitrogen, USA) for 5 days. After removing the suspended cells from the culture supernatant with a Millistack DE60 filter (Millipore, USA), the supernatant was concentrated approximately 10-fold with a SART0C0N Cassete Hydrosart Cu toff 30000 (Sartorius, Germany). . Concentrated culture supernatant Was adjusted to pH 7.95 with NaOH, and then adsorbed to Q Sepharose ™ FF (Amersham Biosciences, USA). After washing the non-specific adsorbate with a washing buffer (20 mM Tris HCl (pH 8.0) .150 mM NaCl), the elution buffer (20 mM Tris HCl (pH 8.0), 150 mM NaCl to 20 mM Tris HCl (pH 8.0), 500 mM NaCl Eluted). Then Phenyl Sepharose ™ HP (US Amersham Biosciences) after a loading, washed with wash buffer (700mM (NH 4) 2 S0 4), at 700 Rev. (NH 4) 2 S0 4 Dara stringent Elution. Next, using Con A Sepharose ™ 4B (Amersham Biosciences, USA), buffer A (PBS) and buffer B (0.5 MMethyl-alpaD-Glucoside, 0.5 M NaCl, PBS) were eluted stepwise. Elution was performed with Wheat Germ Lectin SepharoseTM 6MB (Amersham Biosciences, USA) using buffer A (PBS) and buffer B (0.5M N-acety to!) -Glucosamine, 0.5M NaCl, PBS in a stepwise manner. Done. Then, using DEAE-Sepharose ™ FF (Amersham Biosciences, USA), elution was performed with a buffer A (20 mM TrisHCl ρΗ80 ·) and a buffer Β (20 mM TrisHCl pH 8.0, 0.5 M NaCl) gradient. Using Q SepharoseTM HP 溶出 Eluted with a gradient of buffer A (10OmM phosphate buffer pH 7.5) and buffer B (10mM phosphate buffer PH7.5, 0.5M NaCl). FIG. 14 shows the results of silver staining of the purified product and the results of Western blotting using the anti-klotho antibody prepared in Example 10.
〔実施例 22〕 精製可溶化 klotho夕ンパク質を抗原に用いた抗 klothoポリク ローナル抗体の作製  [Example 22] Production of anti-klotho polyclonal antibody using purified solubilized klotho protein as antigen
22- (1) 抗マウス可溶化 klothoゥサギポリクローナル抗体の調整  22- (1) Preparation of anti-mouse solubilized klotho ゥ sagi polyclonal antibody
実施例 2 1に記載した方法で精製されたマウス可溶化 klotho タンパク質を抗 原として免疫に供した。'免疫は 2羽に行った。 初回免疫は、 ゥサギ 1羽あたり 50 tig の抗原をフロイント完全アジュバントでェマルジヨンとし、 ゥサギの皮内ま たは皮下に投与した。 初回免疫の 1週間後に の抗原をフロイント不完全ァ ジュバントでェマルジヨンとしたものを同様に投与した。 これと同じ投与を 2週 間間隔でさらに 6回実施し、 最終投与の 1週間後に全採血を行ない、 抗血清を調 製した。  The mouse solubilized klotho protein purified by the method described in Example 21 was used for immunization as an antigen. 'Immunization went to two birds. For the first immunization, 50 tig of an antigen per rabbit was emulsified with Freund's complete adjuvant and administered intradermally or subcutaneously to the rabbit. One week after the first immunization, an antigen prepared by emulsifying with Freund's incomplete adjuvant was administered in the same manner. The same administration was performed six more times at two-week intervals, and one week after the final administration, whole blood was collected to prepare antisera.
抗マゥス可溶化 kl 0 tho抗体を含む抗血清をプロテイン G Sepharose 4 FFカラム (米国、 Amersham Pharmaci a Bi otech社) に供して抗体をカラムに吸着させ、 0. 1M グリシン緩衝液(PH2. 8) を用いて溶出させた。溶出画分は l M Tri s- HC 1 (pH9. 0) を添加して pH7. 2に調整した。 このように調製した抗体溶液を NAP10カラム (米 国、 Amersham Pharmac ia Biotech社) を用いて PBS (-)に置換した後、 孔径 0. 22 mのメンブランフィルター MILLEX-GV (米国、 Mi l l ipore社) でろ過滅菌し、 精 製抗 kl otho抗体を得た。 精製抗体の濃度は 280nmの吸光度を測定し、 lmg/ml を 1. 350Dとして算出した。免疫したゥサギは 2羽存在したため 2種類の抗体が取得 された。それぞれを区別する目的で、 これらの精製抗体を以後、 msKl抗体- 1、 msKl 抗体 - 2と記載することにする。 Antiserum containing anti-mouse-solubilized KL 0 tho antibody was transferred to Protein G Sepharose 4 FF column. (Amersham Pharmacia Biotech, USA) to adsorb the antibody to the column and elute it with 0.1 M glycine buffer (PH2.8). The eluted fraction was adjusted to pH 7.2 by adding 1 M Tris-HCl (pH 9.0). After replacing the antibody solution thus prepared with PBS (-) using a NAP10 column (Amersham Pharmacia Biotech, USA), a 0.22 m pore size membrane filter MILLEX-GV (Millipore, USA) ) To obtain a purified anti-klotho antibody. The concentration of the purified antibody was calculated by measuring absorbance at 280 nm and setting lmg / ml to 1.350D. Since two puppies were immunized, two types of antibodies were obtained. For the purpose of distinguishing between them, these purified antibodies are hereinafter referred to as msKl antibody-1 and msKl antibody-2.
22- (2) 抗マウス klotho タンパク質ゥサギポリクロ一ナル抗体によるマウス klo thoタンパク質の認識  22- (2) Recognition of mouse klotho protein by anti-mouse klotho protein and heron polyclonal antibody
上述の方法により取得した msKl抗体- 1および msKl抗体- 2を用いて、実施例 2 1によって作製したマウス可溶化 klotho タンパク質に対するウエスタンブロッ ティング法解析を行った。 図 1 5に示すように、 いずれの抗体でも 130 kDa付近 の可溶化マウス klothoタンパク質が検出された。 したがって、 これら 4種類の抗 体はマウス可溶化 klothoタンパク質や同様な配列を有するマウス klothoタンパ ク質を認識し得る抗体であることが判明した。  Using the msKl antibody-1 and the msKl antibody-2 obtained by the above method, Western blotting analysis was performed on the mouse solubilized klotho protein prepared in Example 21. As shown in Fig. 15, solubilized mouse klotho protein at around 130 kDa was detected for all antibodies. Therefore, these four types of antibodies were found to be antibodies capable of recognizing mouse solubilized klotho protein and mouse klotho protein having a similar sequence.
〔実施例 2 3〕 精製可溶化 klothoタンパク質を抗原に用いた抗 klothoモノク 口一ナル抗体の作製  [Example 23] Preparation of anti-klotho monoclonal antibody using purified solubilized klotho protein as antigen
23 -(1) ハイブリド一マの作製と選別  23-(1) Production and selection of hybrids
本実施例におけるモノクローナル抗体の作製は、 単ク口一ン抗体実験操作入門 (安東民衛ら著作、講談社発行 1991)等に記載されるような一般的方法に従って 調製した。 被免疫動物は、 ラットを用い以下の方法で行なった。  Preparation of the monoclonal antibody in this example was prepared according to a general method as described in Monoclonal Antibody Experiment Procedure Introduction (written by Tamto Ando et al., Published by Kodansha 1991). The animals to be immunized were performed in the following manner using rats.
抗原として実施例 2 1で作製したマウス可溶化 klothoタンパク質を用いた。初 回免疫として、 ラット 1匹あたり の抗原をフロイント完全アジュバントで ェマルジヨンとしラットの皮内または皮下に投与した。 初回免疫の 1週間後から 週 1回の投与を合計 3回、 50 Sの抗原をフロイント不完全アジュバントでエマ ルジョンとしたものを同様に投与した。 最終投与の 2週間後に 50 _i gを尾静脈内 投与し最終免疫とした。 最終免疫後 4日目に以下に記載する方法で、 ハイブリド 一マを作製した。 The mouse solubilized klotho protein prepared in Example 21 was used as an antigen. For the first immunization, the antigen per rat was emulsified with Freund's complete adjuvant and administered intradermally or subcutaneously to the rats. One week after the first immunization, once weekly administration was performed a total of three times, and a 50 S antigen prepared by emulsification with Freund's incomplete adjuvant was similarly administered. Two weeks after the final administration, 50 ig was administered into the tail vein for final immunization. Four days after the final immunization, hybrid I made one.
上述のように免疫したラッ卜から脾臓を摘出し、 それより回収した脾臓細胞を ミエローマ X - 63 Ag8. 6. 5. 3 (ATCC CRL 1580) と混合し、 融合剤としてポリェチレ ングリコール溶液 (日本、 免疫生物研究所社) を用いて細胞融合させ、 ハイプリ ドーマを作製した。 ハイプリ ドーマの選択は、 10%のゥシ胎児血清 (Fe t al Cal f Serum, FCS) とヒポキサンチン(H)、 アミノプテリン(A)、 チミジン(T)を含有する HAT含有 TIL Med ia l (日本国、 免疫生物研究所社) 中で培養することによりおこ なった。 さらに、 限界希釈法によりクロ一ニングを実施し、 単一細胞に由来する クローン化ハイブリドーマを取得した。  The spleen was excised from the immunized rat as described above, and the spleen cells recovered from the spleen were mixed with myeloma X-63 Ag 8.6.5.3 (ATCC CRL 1580) and used as a fusion agent in polyethylene glycol solution (Japan). , Immunological Biology Laboratories) to produce hybridomas. The selection of Hypridoma was performed using TIL Med ial containing HAT containing 10% fetal calf serum (Fetal Calf Serum, FCS) and hypoxanthine (H), aminopterin (A), and thymidine (T). (Immune Biological Laboratory, Japan). Cloning was performed by the limiting dilution method to obtain a cloned hybridoma derived from a single cell.
23- (2) 抗 klotho抗体を産生するクローン化したハイプリ ドーマの選別  23- (2) Screening of cloned hybridoma producing anti-klotho antibody
ハイプリドーマが産生する抗体とマウス kl otho 発現細胞との結合性を調べる ことで、 klotho蛋白質を特異的に認識する抗体を産生するハイプリドーマを選別 した。後述の実施例 3 1で記載した kl otho発現 HEK293細胞を ELISA用 96穴マイ クロプレート (米国、 グライナ一社) の各ゥエルに 1 X 105 細胞加え、 プレート に結合させた。次に培地を除去し、各ゥエルを 2%の FCSを含有する PBS (-)で洗浄 した後、ハイブリドーマの上清を加え 4°Cで 30分インキュベートした後洗浄した。 2000倍希釈した過酸化酵素標識抗ラット IgGャギ I gG Fab' (米国、 SouthernB io tech 社) を加え、 4°Cで 30分間インキュベートした。 洗浄後、 テトラメチルベンジジ ン (デンマーク国、 DAK0社) を含む基質緩衝液を各ゥエルに加え、.室温下で 8分 間インキュベートした。 次いで、 1 M硫酸を各ゥエルに加え、 反応を止めた。 波 長 450 nmでの吸光度をマイクロプレートリーダー(Emax、米国、 Mol ecu l ar Devi ces 社)で測定した。ここで、明確な吸光度の上昇を示したハイプリドーマを選別し、 klotho 蛋白質を認識する抗体を産生するハイプリドーマとして 6 種のクローン (3GK 4A1、 9A1、 12B K 67G4 および 70E2)を得た。 このように取得した klotho 蛋白質を特異的に認識する抗体のサブクラスを Rat MonoAB ID/SP KIT (米国、 ZYMED LABORATORIES社) を用いて同定した。 結果は表 2に示した。 表 2
Figure imgf000054_0001
By examining the binding property between the antibody produced by the hybridoma and cells expressing mouse klotho, hybridomas producing an antibody that specifically recognizes the klotho protein were selected. 1 × 10 5 cells of the klotho-expressing HEK293 cells described in Example 31 described later were added to each well of a 96-well ELISA microplate (Grainer Inc., USA) and allowed to bind to the plate. Next, the medium was removed, and each well was washed with PBS (-) containing 2% FCS, and then the supernatant of the hybridoma was added, followed by incubation at 4 ° C for 30 minutes, followed by washing. Peroxidase-labeled anti-rat IgG goat IgG Fab '(Southern Biotech, USA) diluted 2000-fold was added and incubated at 4 ° C for 30 minutes. After washing, a substrate buffer containing tetramethylbenzidine (DAK0, Denmark) was added to each well and incubated at room temperature for 8 minutes. Then, 1 M sulfuric acid was added to each well to stop the reaction. The absorbance at a wavelength of 450 nm was measured with a microplate reader (Emax, Molecular Devices, USA). Here, hybridomas showing a clear increase in absorbance were selected, and six clones (3GK 4A1, 9A1, 12BK 67G4 and 70E2) were obtained as hybridomas producing antibodies recognizing the klotho protein. The subclass of the antibody specifically recognizing the klotho protein thus obtained was identified using Rat MonoAB ID / SP KIT (ZYMED LABORATORIES, USA). The results are shown in Table 2. Table 2
Figure imgf000054_0001
67G4 IgG1 (κ)  67G4 IgG1 (κ)
70E2 IgG1 (κ) 上記ハイブリドーマクローンのうち、 3種のハイブリ ドーマクローン 4Α1、9Α1、 12B1を、 2004年 3月 30日付で、独立行政法人産業技術総合研究所 特許生物寄託 センター (茨城県つくば市東 1丁目 1番地 1 中央第 6) にブダペスト条約に基 づき国際寄託した。 受託番号は以下の通りである。 ski A1 : FERM BP- 08678  70E2 IgG1 (κ) Of the above hybridoma clones, three hybridoma clones 4Α1, 9Α1, and 12B1 were purchased by the National Institute of Advanced Industrial Science and Technology, National Institute of Advanced Industrial Science and Technology, Patent Organism Depositary on March 30, 2004. An international deposit was made at Chome No. 1 1 Central No. 6) based on the Budapest Treaty. The accession numbers are as follows. ski A1: FERM BP- 08678
ski 9A1 : FERM BP- 08679 ski 9A1: FERM BP- 08679
ski 12A1 : FERM BP- 08680 ski 12A1: FERM BP- 08680
23- (3) 抗 klothoモノクローナル抗体を含む培養上清の調製 23- (3) Preparation of culture supernatant containing anti-klotho monoclonal antibody
抗 klotho抗体産生ハイブリドーマを 10 g/mlのゥシィンシユリン(米国、 Sigma 社)、 5.51 g/ml のヒトトランスフェリン (米国、 Sigma社)、 0. OlmMのエタノー ルァミン (米国、 Sigma社)、 5 ng/mlの亜セレン酸ナトリウム (米国、 Sigma社) および 1%の低 IgGFCS (米国、 Hyclone社) を含有する ERDF培地 (日本、 極東製 薬社) に馴化した。 抗体を調製するためにハイプリドーマの培養を行い、 培養液 を孔径が 0. のフィルター (米国、 Pall Gelman Laboratory社) を通すこと でハイプリドーマ等の雑排物を除去し、 抗体を含む培養上清を回収した。  Anti-klotho antibody-producing hybridoma was prepared by adding 10 g / ml of cincinulin (Sigma, USA), 5.51 g / ml of human transferrin (Sigma, USA), 0. OlmM ethanolamine (Sigma, USA), 5 ng / ml ERDF medium (Farto Seiyaku, Japan) containing 1% sodium selenite (Sigma, USA) and 1% low IgGFCS (Hyclone, USA). The hybridoma is cultured to prepare the antibody, and the culture solution is passed through a filter with a pore size of 0 (Pall Gelman Laboratory, USA) to remove foreign substances such as the hybridoma, and the culture containing the antibody is removed. Qing was recovered.
23- (4) プロティン Gを用いたモノク口一ナル抗体の精製 23- (4) Purification of Monoclonal Monoclonal Antibody Using Protein G
抗 klotho抗体を含む培養上清をプロテイン G Sepharose4FFカラム (米国、 The culture supernatant containing the anti-klotho antibody was applied to a protein G Sepharose4FF column (US,
Amersham Pharmacia Biotech社) に供し、 抗体をカラムに吸着させ、 0.1 M グリ シン緩衝液 (PH2.8) を用いて溶出させた。 溶出画分は 1 M Tris- HC1 を添加してAmersham Pharmacia Biotech), the antibody was adsorbed on the column, and eluted with 0.1 M glycine buffer (PH2.8). The eluted fraction is added with 1 M Tris-HC1
PH7.2に調整した。 このように調製した抗体溶液をゲルろ過カラム NAP25 (米国、Adjusted to PH7.2. The antibody solution thus prepared is applied to a gel filtration column NAP25 (US,
Amersham Pharmacia Biotech社) を用いて PBS (-)に置換し、 孔径 0.22 III のメ Replace with PBS (-) using Amersham Pharmacia Biotech, and use a 0.22
'一 MILLEX - GV (米国、 Millipore社)でろ過滅菌し、精製抗 FGF - 23 抗体を得た。 精製抗体の濃度は 280 ηπι の吸光度を測定し、 1 mg/ml を 1.35 0D として算出した。 'I Filtrated and sterilized with Millex-GV (Millipore, USA) and purified anti-FGF-23 Antibodies were obtained. The concentration of the purified antibody was calculated by measuring absorbance at 280 ηπι and setting 1 mg / ml to 1.350 D.
このようにして得られた各精製モノクローナル抗体を、 産生するハイプリ ドー マの名前を用いて示すことにする。 例えばハイプリドーマ 4A1が産生する抗体に 関しては 4A1抗体と記載することにする。  Each of the purified monoclonal antibodies thus obtained will be indicated using the name of the produced hybridoma. For example, an antibody produced by hybridoma 4A1 will be referred to as a 4A1 antibody.
23- (5) 抗マウス可溶化 klothoラットモノクローナル抗体のマウス klothoへの結 合の確認  23- (5) Confirmation of binding of anti-mouse solubilized klotho rat monoclonal antibody to mouse klotho
抗マウス可溶化 klothoラットモノクローナル抗体がマウス可溶化 klothoと結 合することを以下の実験によって示した。実施例 8と同様な方法で取得されたマ ウス可溶化 klothoタンパク質を含む培養上清を用いて、免疫沈降を行った。マウ ス可溶化 klothoタンパク質を含む培養上清 200 Lに実施例 23—(1)から 23—(4) で記載された方法で作製及び精製された抗 klotho マウスモノクローナル抗体 6 種 (3G1抗体、 4A1抗体、 9A1抗体、 12B1抗体、 67G4抗体、 70E2抗体) およびコ ントロールとして klothoを認識しないラッ卜 IgGl抗体およびラット IgG2b抗体 をそれぞれ 加え、 4°Cにおいて 1.5時間転倒混和することにより、 抗体と klothoの複合体をそれぞれ形成させた。これらの複合体にプロテイン GSepharose The following experiments showed that the anti-mouse solubilized klotho rat monoclonal antibody binds to the mouse solubilized klotho. Immunoprecipitation was performed using a culture supernatant containing mouse-solubilized klotho protein obtained in the same manner as in Example 8. Six kinds of anti-klotho mouse monoclonal antibodies (3G1 antibody, 4A1 antibody) prepared and purified by the method described in Examples 23- (1) to 23- (4) in 200 L of culture supernatant containing mouse-solubilized klotho protein Antibody, 9A1 antibody, 12B1 antibody, 67G4 antibody, 70E2 antibody) and rat IgG1 and rat IgG2b antibodies that do not recognize klotho as controls, and mix by inverting at 4 ° C for 1.5 hours. Each complex was formed. These complexes contain protein GSepharose
4FFカラム (米国、 Amersham Pharmacia Biotech社) を 15 L加え 4 °CにおいてAdd 15 L of 4FF column (Amersham Pharmacia Biotech, USA) at 4 ° C
2 時間転倒混和した後、 遠心分離し、 液相(レジン非吸着物)および免疫沈降物を 含むレジンに分けた。レジンを PBS (-)でよく洗浄し、非特異的結合物を除去した。 洗浄済みのレジンに 50 1のサンプルバッファー(50 mM Tris- CI pH6.8,0.4% SDS、After mixing by inversion for 2 hours, the mixture was centrifuged and separated into a liquid phase (non-resin-adsorbed material) and a resin containing immunoprecipitates. The resin was thoroughly washed with PBS (-) to remove non-specifically bound substances. Add 50 1 sample buffer (50 mM Tris-CI pH 6.8, 0.4% SDS,
6%グリセロール、 0.002%ブロムフエノールブルー、 2% )3メルカブトエタノール) を添加し、 95°Cで 5分間加熱したのち、 遠心分離して得た上清 (レジン吸着物) を回収した。 レジン吸着物は実施例 1 0で取得した mklotho- 118抗体を用いて、 ウエスタンブロッテイング法により解析を行った。 その結果を図 1 6に示す。 レ ジン吸着物の抗 klotho抗体を用いたウエスタンブロット法による解析から、 3G1 抗体、 4A1抗体、 9A1抗体、 12B1抗体、 67G4抗体、 70E2抗体を添加したサンプル のおいて可溶型 klothoがレジンに吸着していること、すなわち免疫沈降されてい ることが示された。 この結果から、上述の抗 klothoラットモノクローナル抗体は マウス可溶化 klothoと結合することが確認された。 〔実施例 2 4〕 精製可溶化 kl othoタンパク質を抗原に用いた抗 kl o thoポリク ローナル抗体の EGR 1プロモータ ·ルシフェラ一ゼアツセィ系での中和活性測定 実施例 2 2で作製した抗 kl othoポリクローナル抗体の EGR1プロモータ ·ルシ フェラ一ゼアツセィ系での FGF23刺激への抑制効果を検討した。 After addition of 6% glycerol, 0.002% bromphenol blue, 2%) 3 mercaptoethanol) and heating at 95 ° C for 5 minutes, the supernatant (resin adsorbate) obtained by centrifugation was collected. The resin adsorbate was analyzed by the western blotting method using the mklotho-118 antibody obtained in Example 10. Figure 16 shows the results. Analysis of the resin adsorbate by Western blot analysis using anti-klotho antibody showed that soluble klotho was adsorbed to the resin in the sample to which 3G1, 4A1, 9A1, 12B1, 67G4, and 70E2 antibodies were added. In other words, that is, immunoprecipitation was observed. From these results, it was confirmed that the anti-klotho rat monoclonal antibody described above binds to mouse solubilized klotho. Example 24 Measurement of Neutralizing Activity of Anti-klotho Polyclonal Antibody Using Purified Solubilized klotho Protein as Antigen in EGR 1 Promoter Lucifera Zeatosis System Anti-klotho Polyclonal Prepared in Example 22 The inhibitory effect of the antibody on the FGF23 stimulation in the EGR1 promoter-Lucifera azetus system was examined.
方法は実施例 9に従って行い、 FGF23 による刺激前にあらかじめ細胞培養液に 抗 kl othoポリクロ一ナル抗体を添加した。 FGF23の刺激は最終濃度 10ng/mlで行 なった。その結果、図 1 7に示すようにいずれの抗 kl othoポリクロ一ナル抗体も 濃度依存的に FGF23による刺激を抑制した。 このことから実施例 2 2で作製した 抗 kl otho抗体は FGF23による細胞への刺激を抑制することが示された。  The method was performed according to Example 9, and an anti-klotho polyclonal antibody was added to the cell culture solution before stimulation with FGF23. FGF23 stimulation was performed at a final concentration of 10 ng / ml. As a result, as shown in FIG. 17, all of the anti-klotho polyclonal antibodies suppressed the stimulation by FGF23 in a concentration-dependent manner. This indicated that the anti-klotho antibody prepared in Example 22 suppressed FGF23-induced stimulation of cells.
〔実施例 2 5〕 精製可溶化 kl othoタンパク質を抗原に用いた抗 kl o thoポリク ローナル抗体のマウス投与における効果  [Example 25] Effect of administration of anti-clotho polyclonal antibody to mice using purified solubilized kliotho protein as an antigen
実施例 2 2で作製した抗 kl otho ポリクローナル抗体のマウスへの投与による 効果を検討した。 BALB/c、 雄、 Ί 週齢の正常マウスに実施例 2 2で得られた抗マ ウス kl othoタンパク質ゥサギポリクローナル抗体 mskl抗体- 1、 2を各々、 尾静 脈内より 1匹あたり lmg/0. 2 mLずつ単回投与した。 対照群へは、 klotho免疫前 ゥサギ血清を実施例 2 2と同条件で精製したものをコントロール抗体として、 0. 2 mLずつ尾静脈内より投与した。投与 9時間後にガラス製キヤビラリを用いて眼窩 より採血し、 マイクロティナ (米国、 べクトンディツキソン社) を用いて血清を 分離、 さらに 24時間後に心臓より採血し、 同じくマイクロティナを用いて血清を 分離した。 得られた 9 時間後の血清で 1, 25-ジヒドロキシビタミン D 濃度を 1, 25 (OH) 2D RIAキット 「TFB」 (日本国、 ティエフビー社)を用いて測定した。 ま た 24時間後の血清でリン酸濃度をフォスファ C-テストヮコ一(日本国、和光純薬) を用いて添付文書に従い、決定した。抗 kl othoポリクロ一ナル抗体投与群および 対照群はそれぞれ 5匹のマウスから構成され、水道水および 1. 03 %の無機リン酸 および 1. 18%のカルシウムを含んだ固形食 CE2 (日本国、 日本クレア社) 自由摂 取させた。  The effect of administering the anti-klotho polyclonal antibody prepared in Example 22 to mice was examined. BALB / c, male, Ί week-old normal mouse Anti-mouse klotho protein obtained in Example 22 ゥ Heron polyclonal antibody mskl antibody-1, 2 A single dose of 0.2 mL was administered. To the control group, 0.2 mL of the persimmon serum before klotho immunization purified under the same conditions as in Example 22 was injected into the tail vein in an amount of 0.2 mL. 9 hours after administration, blood was collected from the orbit using a glass capillary, serum was separated using Microtina (Becton Dickson, USA), and blood was collected 24 hours later from the heart, and serum was also collected using Microtina. Was separated. Nine hours after the obtained serum, the concentration of 1,25-dihydroxyvitamin D was measured using a 1,25 (OH) 2D RIA kit “TFB” (Tiefbee, Japan). The phosphate concentration of the serum 24 hours later was determined using Phospha C-Test Co., Ltd. (Wako Pure Chemical Industries, Japan) according to the package insert. The group treated with anti-klotho polyclonal antibody and the control group each consisted of 5 mice, and had a solid diet containing tap water and 1.03% inorganic phosphate and 1.18% calcium CE2 (Japan, (Clear Japan, Inc.)
図 1 8に示すとおり、抗マウス klo thoタンパク質ゥサギポリクローナル抗体の 単回投与によりコントロール抗体投与群と比較して、 mskl抗体 _ 1、 2ともに 9時 間後の血清ジヒドロキシビタミン Dに有意な上昇を(t t es t **pく 0. 01)、 また 24 時間後の血清リン酸に有意な上昇を認めた(t tes t * く 0. 05)。 As shown in Fig. 18, a single dose of anti-mouse clone protein ゥ sagi polyclonal antibody significantly increased serum dihydroxyvitamin D in both mskl antibodies _ 1 and 2 after 9 hours compared to the control antibody-administered group. (Tt es t ** p <0.01) and 24 There was a significant increase in serum phosphate after time (t tes t * 0.05).
〔実施例 2 6〕 精製可溶化 klotho夕ンパク質を抗原に用いた抗 klothoモノク ローナル抗体の EGR 1プロモータ ·ルシフェラーゼアツセィ系での中和活性測定 実施例 2 3で作製した抗 klothoモノクローナル抗体の EGR1プロモータ ·ルシ フェラーゼアツセィ系での FGF23刺激への抑制効果を検討した。 方法は実施例 2 4と同様に行なった。 FGF23の刺激は最終濃度 10ng/mlで行った。 抗 klothoモノ クロ一ナル抗体は skl4Al抗体, skl9Al抗体, ski 12B1抗体を用いコントロールと して IgGl, IgG2bのサブタイプコントロール抗体を用いた。その濃度は lOO ^ g/ml から 0. l g/mlの範囲で 10分の 1ずつ振った。 その結果、 図 1 9に示すように、 skl9Al抗 klothoモノクローナル抗体は FGF23による刺激を用量依存的に抑制し た。 skl4Al抗体、 skl l2Bl抗体には抑制は確認されなかった。 このことから実施 例 2 3で作製した抗 klotho抗体 skl'9Alは FGF23による細胞への刺激を抑制する ことが示された。  (Example 26) Measurement of neutralizing activity of anti-klotho monoclonal antibody using purified purified solubilized klotho protein as antigen as an antigen in EGR 1 promoter / luciferase Atsushi system Example 2 of anti-klotho monoclonal antibody prepared in Example 23 The inhibitory effect of the EGR1 promoter-luciferase atsey system on FGF23 stimulation was examined. The method was the same as in Example 24. FGF23 stimulation was performed at a final concentration of 10 ng / ml. Anti-klotho monoclonal antibodies were skl4Al, skl9Al, and ski12B1 antibodies, and IgGl and IgG2b subtype control antibodies were used as controls. The concentration was shaken in tenths from lOO ^ g / ml to 0.1 g / ml. As a result, as shown in FIG. 19, the skl9Al anti-klotho monoclonal antibody suppressed the stimulation by FGF23 in a dose-dependent manner. No inhibition was confirmed with the skl4Al antibody or the skl12Bl antibody. This indicated that the anti-klotho antibody skl'9Al prepared in Example 23 suppressed the stimulation of cells by FGF23.
〔実施例 2 7〕 精製可溶化 klotho夕ンパク質を抗原に用いた抗 klothoモノク ローナル抗体のマウス投与における効果  [Example 27] Effect of mouse administration of anti-klotho monoclonal antibody using purified solubilized klotho protein as antigen
BALB/c、雄、 8週齢の正常マウスに実施例 27で得られた抗マウス可溶化 klotho ラットモノクローナル抗体 6種 (3G1抗体、 4A1抗体、 9A1抗体、 12B1抗体、 67G4 抗体、 70E2抗体) を各々、 尾静脈内より 1匹あたり lOO g/0. 2 mLずつ単回投与 した。 対照群へは、 klothoを認識しないラット IgGl抗体をコントロール抗体と して、 0. 2 mLずつ尾静脈内より投与した。 投与 9時間後にガラス製キヤビラリを 用いて眼窩より採血し、 マイクロティナ (べクトンディツキソン社、 米国) を用 いて血清を分離、 さらに 24時間後に心臓より採血し、 同じくマイクロティナを用 いて血清を分離した。 得られた 9時間後の血清で 1, 25-ジヒドロキシビタミン D 濃度を 1, 25 (OH) 2D RIAキット 「TFB」 (ティエフビー社、 日本国)を用いて測定し た。 また 24時間後の血清でリン酸濃度をフォスファ C-テストヮコー(和光純薬、 日本国)を用いて添付文書に従い、 決定した。 抗マウス可溶化 klothoラットモノ ク口一ナル抗体投与群および対照群はそれぞれ 5匹のマウスから構成され、 水道 水および 1. 03%の無機リン酸および 1. 18%のカルシウムを含んだ固形食 CE2 (日 本クレア社、 日本国)を自由摂取させた。 図 2 0に示すとおり、抗マウス可溶化 klothoラットモノクローナル抗体の単回 投与のうち、 コントロール抗体投与群と比較して、 3G1抗体、 9A1抗体、 67G4抗 体、 70E2抗体の 4種で 9時間後の血清ジヒドロキシビタミン Dに有意な上昇を(t test * く 0.05、 く 0.01)、 また 24時間後の血清リン酸に有意な上昇を認め た (t test *p く 0.05、 **p < 0.01)。 BALB / c, male, 6-week-old normal mouse, 6 kinds of anti-mouse-solubilized klotho rat monoclonal antibodies obtained in Example 27 (3G1, 4A1, 9A1, 12B1, 67G4, and 70E2 antibodies) Each animal was administered once from the tail vein at a dose of 100 g / 0.2 mL per animal. To the control group, a rat IgG1 antibody that did not recognize klotho was administered as a control antibody by 0.2 mL in a tail vein. Nine hours after administration, blood was collected from the orbit using a glass capillary, serum was separated using Microtina (Becton Dickson, USA), and 24 hours later, blood was collected from the heart, and serum was also used using Microtina. Was isolated. Nine hours after the obtained serum, the concentration of 1,25-dihydroxyvitamin D was measured using a 1,25 (OH) 2D RIA kit “TFB” (TFB, Japan). Phosphoric acid concentration in serum 24 hours later was determined using Phospha C-Test II Co. (Wako Pure Chemical Industries, Japan) according to the package insert. Anti-mouse-solubilized klotho rat Monoclonal antibody-administered group and control group each consisted of 5 mice, each consisting of tap water and a solid diet containing 1.03% inorganic phosphate and 1.18% calcium CE2 (Clea Japan, Japan). As shown in Fig. 20, after a single dose of the anti-mouse solubilized klotho rat monoclonal antibody, 9 hours after treatment with 4 types of 3G1, 9A1, 67G4, and 70E2 antibodies compared to the control antibody-administered group Significantly increased in serum dihydroxyvitamin D (t test * 0.05, 0.01) and serum phosphate 24 hours later (t test * 0.05, ** p <0.01) .
〔実施例 2 8〕 精製可溶化 klothoタンパク質と精製 FGF23のヒ卜およびマウス 培養細胞への同時添加による細胞への刺激の効果  [Example 28] Effect of simultaneous addition of purified solubilized klotho protein and purified FGF23 to human and mouse cultured cells to stimulate cells
実施例 2 1で得た精製可溶化 klothoの活性を確認する為、実施例 8と同様の方 法にて HEK293細胞を用いて可溶化 klothoと FGF23による細胞への刺激の効果を 検討した。 刺激に用いた可溶化 klothoは 4 g/m FGF23は 100ng/mlの濃度に て刺激を行なった。その結果、図 2 1に示すように可溶化 klothoと FGF23による 刺激を行なったときのみ細胞内の ERKのリン酸化亢進が確認された。  In order to confirm the activity of the purified solubilized klotho obtained in Example 21, the effect of stimulating cells with the solubilized klotho and FGF23 was examined using HEK293 cells in the same manner as in Example 8. The solubilized klotho used for the stimulation was stimulated at a concentration of 4 g / m FGF23 at a concentration of 100 ng / ml. As a result, as shown in FIG. 21, only when stimulation with solubilized klotho and FGF23 was performed, the phosphorylation of ERK in cells was enhanced.
また、 マウス骨芽細胞由来細胞 MC3T3- E1細胞に可溶化 klothoは 4 g./mK FGF23は 100ng/mlの濃度にて刺激を行ない、 48時間培養を行なった。その結果、 図 22に示すように可溶化 klothoと FGF23で剌激を行なつたものについては細胞 の形態が bFGF刺激と同様に繊維芽細胞様に変化した。  In addition, mouse osteoblast-derived cells were solubilized in MC3T3-E1 cells. Klotho was stimulated at a concentration of 4 g./mK FGF23 at a concentration of 100 ng / ml, and cultured for 48 hours. As a result, as shown in FIG. 22, the cell morphology of the cells stimulated with solubilized klotho and FGF23 changed to fibroblast-like cells as in the case of bFGF stimulation.
これらのことから、可溶化 klothoと FGF23の同時刺激を行なったときのみに細 胞へ刺激が入ることが示された。  These results indicate that cells were stimulated only when co-stimulation with solubilized klotho and FGF23 was performed.
〔実施例 2 9〕 精製可溶化 klothoタンパク質のマウス投与における効果  [Example 29] Effect of purified solubilized klotho protein on mouse administration
BALB/c、 雄、 7週齢の正常マウスに実施例 2 1で得られたマウス可溶化 klotho タンパク質を 10 g/0.2mL尾静脈より単回投与した。 対照群へは PBS を 0.2 mL ずつ尾静脈内より投与した。 投与 9時間後にガラス製キヤビラリを用いて眼窩よ り採血し、 マイクロティナ (べクトンディツキソン社、 米国) を用いて血清を分 離した。 得られた 9 時間後の血清で 1, 25-ジヒドロキシビタミン D 濃度を BALB / c, male, 7-week-old normal mice were administered a single dose of the mouse-solubilized klotho protein obtained in Example 21 via the tail vein at 10 g / 0.2 mL. To the control group, 0.2 mL of PBS was administered into the tail vein in a dose of 0.2 mL. Nine hours after the administration, blood was collected from the orbit using a glass capillary, and the serum was separated using Microtina (Becton Dickson, USA). Nine hours after the obtained serum, the 1,25-dihydroxyvitamin D
1, 25(0H)2D RIA キット 「TFB」 (ティエフビー社、 日本国)を用いて測定した。 マ ウス可溶化 klothoタンパク質投与群および対照群はそれぞれ 5匹のマウスから構 成され、 水道水および 1.03%の無機リン酸および 1.18%のカルシウムを含んだ固 形食 CE2 (日本クレア社、 日本国)を自由摂取させた。 The measurement was performed using a 1,25 (0H) 2D RIA kit "TFB" (Tifby, Japan). Mouse-solubilized klotho protein-treated and control groups consisted of 5 mice each, and consisted of a solid diet containing tap water and 1.03% inorganic phosphate and 1.18% calcium CE2 (CLEA Japan, Japan) ).
図 2 3に示すとおり、マウス可溶化 klothoタンパク質の単回投与により、対象 群と比較して 9 時間後の血清ジヒドロキシビタミン D に有意な上昇を認めた(t tes t * く 0· 05) As shown in Figure 23, a single dose of mouse solubilized klotho protein A significant increase in serum dihydroxyvitamin D was observed 9 hours later compared to the control group (t tes t * 0 · 05)
〔実施例 3 0〕 抗 FGF23ポリクローナル抗体の作製  [Example 30] Preparation of anti-FGF23 polyclonal antibody
ヒト FGF23に対するポリクローナル抗体を取得する為、 実施例 1 5にて得た精 製ヒト FGF23を用いて、 実施例 2 2と同様の方法にて抗 FGF23全長ポリクローナ ル抗体を得た。  To obtain a polyclonal antibody against human FGF23, an anti-FGF23 full-length polyclonal antibody was obtained in the same manner as in Example 22 using the purified human FGF23 obtained in Example 15.
〔実施例 3 1〕 klotho発現細胞への FGF23の結合  [Example 31] Binding of FGF23 to klotho-expressing cells
細胞に発現させた klothoと FGF23との結合を以下の方法により確かめた。  The binding between klotho expressed in the cells and FGF23 was confirmed by the following method.
31- (1) klotho安定発現 HEK293細胞の作製 31- (1) Preparation of klotho stably expressing HEK293 cells
マウス klotho 全長安定発現 HEK293 細胞を得る為、 実施例 3で得た mklot o/IRES-EGFP-pEAK8 ベクターを用いて、 実施例 5と同様の方法にてマウス klotho全長安定発現 HEK293細胞を得た。  To obtain mouse klotho full-length stable expression HEK293 cells, mouse klotho full-length stable expression HEK293 cells were obtained in the same manner as in Example 5, using the mklot o / IRES-EGFP-pEAK8 vector obtained in Example 3.
31- (2) klothoと FGF23の結合確認 31- (2) Confirmation of binding between klotho and FGF23
31- (1)で作製したマウス klotho全長安定発現 HEK293細胞を用いて細胞に結合 した FGF23 の検出を行なった。 実施例 3 1— ( 1 ) で得られた klotho安定発現 Using the mouse klotho full-length stable expression HEK293 cells prepared in 31- (1), FGF23 bound to the cells was detected. Example 3 Klotho Stable Expression Obtained in 1— (1)
HEK293細胞を細胞乖離バッファ一 (0. 52mM EDTA, PBS) にて培養容器より乖離さ せ、 懸濁バッファー (2% 仔牛血清、 lmM EDTA, 0. 05¾ NaN3) にて懸濁を行なった。 実施例 1 5にて得られたヒト FGF23を l ^ g/mlの濃度で添加し、氷上で 30分静置。 その後懸濁バッファ一にて洗浄した後、 実施例 3 0にて得られた抗 FGF23ポリク ローナル抗体を 20 /2 g/mlの濃度にて添加し、氷上にて 3 0分静置。懸濁バッファ 一にて洗浄後、 抗ゥサギ IgG PE修飾抗体 (米国 サザンバイオテクノロジーァソ シェイツ社) を添加し、 氷上で 30分静置。 洗浄バッファーで洗浄後、 細胞の蛍光 強度を FACS Cal iber装置 (米国 べクトンディッキンソン社) にて検出を行な つた。 結果を図 2 4 Aおよび Bに記す。 図 2 4 Aおよび B中、 上から 4つ目までの グラフは、 それぞれ抗 FGF23抗体、 FGF23 +抗 FGF23抗体、 FGF23 +コントロール 抗体および FGF23を用いた場合の蛍光強度を示し、 一番下のグラフは上 4つのグ ラフを合わせたグラフである。 klotho全長安定発現 HEK293細胞では FGF23と抗HEK293 cells were detached from the culture vessel using a cell detachment buffer (0.52 mM EDTA, PBS), and suspended in a suspension buffer (2% calf serum, lmM EDTA, 0.05 NaN 3 ). The human FGF23 obtained in Example 15 was added at a concentration of l ^ g / ml and left on ice for 30 minutes. After washing with a suspension buffer, the anti-FGF23 polyclonal antibody obtained in Example 30 was added at a concentration of 20/2 g / ml, and the mixture was allowed to stand on ice for 30 minutes. After washing with a suspension buffer, an anti-Egret IgG PE-modified antibody (Southern Biotechnology Asso-Shats, USA) was added, and the mixture was allowed to stand on ice for 30 minutes. After washing with the washing buffer, the fluorescence intensity of the cells was detected using a FACS Cal iber device (Becton Dickinson, USA). The results are shown in FIGS. 24A and B. In Fig. 24 4A and B, the four graphs from the top show the fluorescence intensity when using anti-FGF23 antibody, FGF23 + anti-FGF23 antibody, FGF23 + control antibody and FGF23, respectively, and the bottom graph Is a graph that combines the top four graphs. Klotho full-length stable expression
FGF23を加えたときのみに強い蛍光強度を示し、 HEK293細胞ではそのような結果 は得られなかった。 このことから、細胞に発現した klothoと FGF23が結合するこ とが示された。 本明細書で引用した全ての刊行物、 特許および特許出願をそのまま参考として 本明細書にとり入れるものとする。 産業上の利用の可能性 Only when FGF23 was added, the fluorescence intensity was strong, and such a result was not obtained in HEK293 cells. This indicates that klotho expressed in cells and FGF23 bind. Was shown. All publications, patents and patent applications cited herein are hereby incorporated by reference in their entirety. Industrial potential
実施例に示すように、 klothoタンパク質の投与により血中の 1, 5ジヒドロキ シビタミン D濃度および血清リン濃度が低下または上昇する。 また、 抗 kol otho タンパク質抗体の投与により血中の 1, 25ジヒドロキシビタミン D濃度および血清 リン濃度が上昇する。 また、 klothoタンパク質の投与により体内での FGF23の作 用が増強または阻害され、 抗体 klotho タンパク質抗体の投与により体内での FGF23 の作用が阻害される。 これらの実施例は、 klotho タンパク質および抗 kol othoタンパク質抗体が血中の 1, 25ジヒドロキシビタミン D濃度および血清リ ン濃度を変動させ、 リンおよびノまたはビタミン Dおよび または FGF23が関与 する疾患の治療および Zまたは予防に使用し得ることを示している。  As shown in the Examples, administration of klotho protein reduces or increases the blood 1,5 dihydroxyvitamin D concentration and serum phosphorus concentration. In addition, administration of anti-kolotho protein antibody increases blood 1,25-dihydroxyvitamin D concentration and serum phosphorus concentration. The administration of klotho protein enhances or inhibits the action of FGF23 in the body, and the administration of the antibody klotho protein antibody inhibits the action of FGF23 in the body. These examples demonstrate that klotho protein and anti-kolotho protein antibodies alter blood 1,25-dihydroxyvitamin D and serum phosphorus levels, and are used in the treatment of diseases involving phosphorus and / or vitamin D and / or FGF23. Z or indicates that it can be used for prevention.
配列表フリーテキス卜 Sequence Listing Free Text
配列番号 4〜2 2、 2 8〜3 0、 3 2〜3 5 :合成0  SEQ ID NOs: 4 to 22, 28 to 30 and 32 to 35: Synthesis 0
配列番号 2 4〜2 7、 3 1 :合成ペプチド  SEQ ID NOS: 24 to 27, 31: Synthetic peptide

Claims

請求の範囲 The scope of the claims
1 . klothoタンパク質を有効成分として含む、 血中リン濃度およびノまたは 血中活性型ビタミン D濃度を低下させるための医薬組成物。 1. A pharmaceutical composition for lowering blood phosphorus concentration and blood or active vitamin D concentration, comprising klotho protein as an active ingredient.
2 . klothoタンパク質を有効成分として含む、 血中リン濃度およびノまたは 血中活性型ビタミン D濃度を上昇させるための医薬組成物。  2. A pharmaceutical composition containing klotho protein as an active ingredient for increasing blood phosphorus concentration and blood or active vitamin D concentration.
3 . klothoタンパク質を有効成分として含む、 FGF23の作用を増強するため の医薬組成物。  3. A pharmaceutical composition for enhancing the action of FGF23, comprising a klotho protein as an active ingredient.
4 . klothoタンパク質を有効成分として含む、 FGF23の作用を阻害するため の医薬組成物。  4. A pharmaceutical composition for inhibiting the action of FGF23, comprising a klotho protein as an active ingredient.
5 . klothoタンパク質が可溶化 kl othoタンパク質である、 請求項 1から 4 のいずれか 1項に記載の医薬組成物。  5. The pharmaceutical composition according to any one of claims 1 to 4, wherein the klotho protein is a solubilized klotho protein.
6 . 抗 klothoタンパク質抗体を有効成分として含む、血中リン濃度おょぴ または血中活性型ビタミン D濃度を上昇させるための医薬組成物。  6. A pharmaceutical composition containing an anti-klotho protein antibody as an active ingredient for increasing blood phosphorus level or blood active vitamin D level.
7 . 抗 klothoタンパク質抗体を有効成分として含む、 FGF23の作用を阻害す るための医薬組成物。  7. A pharmaceutical composition for inhibiting the action of FGF23, which comprises an anti-klotho protein antibody as an active ingredient.
8 . 腫瘍性骨軟化症、 ADHIL XLH、 腎性骨異栄養症、 透析骨症、 骨粗鬆症、 低 リン血症、 クル病、 骨軟化症、 尿細管機能障害、 骨減少症、 低カルシウム血症、 高リン血症、 高 1. 25D血症、 副甲状腺機能亢進症、 異所性石灰化、 瘙痒、 骨硬化 症、 パジェット病、 高カルシウム血症、 副甲状腺機能低下症、 骨痛、 筋力低下、 骨格変形、 成長障害および低 1, 25D血症からなる群から選択されるリンおよび またはビタミン Dが関与する疾患の治療およびノまたは予防に使用される請求項 1から 7のいずれか 1項に記載の医薬組成物。  8. Neoplastic osteomalacia, ADHIL XLH, renal osteodystrophy, dialysis osteopathy, osteoporosis, hypophosphatemia, cull disease, osteomalacia, tubular dysfunction, osteopenia, hypocalcemia, Hyperphosphatemia, hyper-1.25D, hyperparathyroidism, ectopic calcification, pruritus, bone sclerosis, Paget's disease, hypercalcemia, hypoparathyroidism, bone pain, muscle weakness, The method according to any one of claims 1 to 7, which is used for treating and / or preventing a disease involving phosphorus and / or vitamin D selected from the group consisting of skeletal deformity, growth disorder, and hypo1,25Demia. Pharmaceutical composition.
9 . 血中リン濃度および //または血中活性型ビタミン D濃度を変動させる物 質をスクリーニングする方法であって、 FGF23 と klothoタンパク質を接触させ、 FGF23と klothoタンパク質との相互作用による細胞内シグナルを検出す'ることに より、 スクリーニングする方法。  9. A method for screening for substances that alter blood phosphorus concentration and / or blood active vitamin D concentration, by contacting FGF23 with klotho protein, and intracellular signals due to the interaction between FGF23 and klotho protein. A screening method by detecting '
PCT/JP2004/006899 2003-05-15 2004-05-14 Klotho protein, anti-klotho protein antibody and use thereof WO2004100976A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2003-137950 2003-05-15
JP2003137950A JP2006240990A (en) 2003-05-15 2003-05-15 Klotho protein and anti-klotho protein antibody and use of them

Publications (1)

Publication Number Publication Date
WO2004100976A1 true WO2004100976A1 (en) 2004-11-25

Family

ID=33447278

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2004/006899 WO2004100976A1 (en) 2003-05-15 2004-05-14 Klotho protein, anti-klotho protein antibody and use thereof

Country Status (2)

Country Link
JP (1) JP2006240990A (en)
WO (1) WO2004100976A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8324160B2 (en) 2009-06-17 2012-12-04 Amgen Inc. Chimeric polypeptides and uses thereof
US8361963B2 (en) 2008-06-04 2013-01-29 Amgen Inc. Uses of FGF21 polypeptides comprising two or more mutations
US8372952B2 (en) 2009-12-02 2013-02-12 Amgen Inc. Binding proteins that bind to human FGFR1C, human β-klotho and both human FGFR1C and human β-klotho
US8795985B2 (en) 2009-05-05 2014-08-05 Amgen Inc. FGF 21 polypeptides comprising two or more mutations and uses thereof
US9279013B2 (en) 2008-10-10 2016-03-08 Amgen Inc. FGF-21 mutants comprising polyethylene glycol and uses thereof
US9284378B2 (en) 2009-12-07 2016-03-15 Shaw-Fen Sylvia Hu Human antigen binding proteins that bind β-Klotho, FGF receptors and complexes thereof
US9493530B2 (en) 2009-05-05 2016-11-15 Amgen Inc. FGF21 mutants comprising a mutation at position 98, 171 and/or 180
US9771567B2 (en) 2012-04-16 2017-09-26 Tel Hashomer Medical Research Infrastructure And Services Ltd. Klotho variant polypeptides
US9987326B2 (en) 2007-05-08 2018-06-05 Tel Hashomer Medical Research Infrastructure And Services Ltd. Klotho protein and related compounds for the treatment and diagnosis of cancer
US10300117B2 (en) 2015-02-06 2019-05-28 The Regents Of The University Of California Methods and compositions for improved cognition
US10555963B2 (en) 2007-05-08 2020-02-11 Tel Hashomer Medical Research Infrastructure And Services Ltd. Klotho protein and related compounds for the treatment and diagnosis of cancer
WO2022243519A1 (en) * 2021-05-21 2022-11-24 Universitat Autònoma De Barcelona Secreted splicing variant of klotho for treating bone disorders

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200936156A (en) * 2008-01-28 2009-09-01 Novartis Ag Methods and compositions using Klotho-FGF fusion polypeptides
JP5927782B2 (en) * 2010-06-15 2016-06-01 国立大学法人広島大学 Use of solubilized Klotho, FGF23 and FGFR complex formation mechanism in calcified tissue
SG11201606018UA (en) 2014-01-24 2016-08-30 Ngm Biopharmaceuticals Inc Binding proteins and methods of use thereof
WO2017019957A2 (en) 2015-07-29 2017-02-02 Ngm Biopharmaceuticals, Inc. Binding proteins and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998029544A1 (en) * 1996-12-26 1998-07-09 Kyowa Hakko Kogyo Co., Ltd. Novel peptide, novel dna, and novel antibody

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998029544A1 (en) * 1996-12-26 1998-07-09 Kyowa Hakko Kogyo Co., Ltd. Novel peptide, novel dna, and novel antibody

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
YAHATA K. ET AL.: "Regulation of stanniocalcin 1 and 2 expression in the kidney by klotho gene", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 310, no. 1, 2003, pages 128 - 134, XP004457194 *
YOSHIDA T. ET AL.: "Mediation of unusually high concentrations of 1,25-dihydroxyvitamin D in homozygous klotho mutant mice by increased expression of renal 1alpha-hydroxylase gene", ENDOCRINOLOGY, vol. 143, no. 2, 2002, pages 683 - 689, XP002981433 *

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9987326B2 (en) 2007-05-08 2018-06-05 Tel Hashomer Medical Research Infrastructure And Services Ltd. Klotho protein and related compounds for the treatment and diagnosis of cancer
US10555963B2 (en) 2007-05-08 2020-02-11 Tel Hashomer Medical Research Infrastructure And Services Ltd. Klotho protein and related compounds for the treatment and diagnosis of cancer
US8361963B2 (en) 2008-06-04 2013-01-29 Amgen Inc. Uses of FGF21 polypeptides comprising two or more mutations
US11840558B2 (en) 2008-06-04 2023-12-12 Amgen Inc. Methods of treating non-alcoholic steatohepatitis using FGF21 mutants
US8410051B2 (en) 2008-06-04 2013-04-02 Amgen Inc. FGF21 mutants and uses thereof
US8642546B2 (en) 2008-06-04 2014-02-04 Amgen Inc. FGF21 mutant fusion polypeptides and uses thereof
US11072640B2 (en) 2008-06-04 2021-07-27 Amgen Inc. Methods of treating non-alcoholic steatohepatitis using FGF21 mutants
US9273106B2 (en) 2008-06-04 2016-03-01 Amgen Inc. FGF mutants with reduced proteolysis and aggregation
US10011642B2 (en) 2008-06-04 2018-07-03 Amgen Inc. Methods of treating of diabetes and obesity using FGF21 mutants
US9279013B2 (en) 2008-10-10 2016-03-08 Amgen Inc. FGF-21 mutants comprising polyethylene glycol and uses thereof
US8795985B2 (en) 2009-05-05 2014-08-05 Amgen Inc. FGF 21 polypeptides comprising two or more mutations and uses thereof
US9493530B2 (en) 2009-05-05 2016-11-15 Amgen Inc. FGF21 mutants comprising a mutation at position 98, 171 and/or 180
US8324160B2 (en) 2009-06-17 2012-12-04 Amgen Inc. Chimeric polypeptides and uses thereof
US8372952B2 (en) 2009-12-02 2013-02-12 Amgen Inc. Binding proteins that bind to human FGFR1C, human β-klotho and both human FGFR1C and human β-klotho
US9493577B2 (en) 2009-12-07 2016-11-15 Amgen Inc. Human antigen binding proteins that bind β-klotho, FGF receptors and complexes thereof
US9284378B2 (en) 2009-12-07 2016-03-15 Shaw-Fen Sylvia Hu Human antigen binding proteins that bind β-Klotho, FGF receptors and complexes thereof
US10570205B2 (en) 2009-12-07 2020-02-25 Amgen, Inc. Human antigen binding proteins that bind β-Klotho, FGF receptors and complexes thereof
US9771567B2 (en) 2012-04-16 2017-09-26 Tel Hashomer Medical Research Infrastructure And Services Ltd. Klotho variant polypeptides
US10300117B2 (en) 2015-02-06 2019-05-28 The Regents Of The University Of California Methods and compositions for improved cognition
US10632180B2 (en) 2015-02-06 2020-04-28 The Regents Of The University Of California Methods and compositions for improved cognition
US10864256B2 (en) 2015-02-06 2020-12-15 The Regents Of The University Of California Methods and compositions for improved cognition
US11738070B2 (en) 2015-02-06 2023-08-29 The Regents Of The University Of California Methods and compositions for improved cognition
WO2022243519A1 (en) * 2021-05-21 2022-11-24 Universitat Autònoma De Barcelona Secreted splicing variant of klotho for treating bone disorders

Also Published As

Publication number Publication date
JP2006240990A (en) 2006-09-14

Similar Documents

Publication Publication Date Title
JP4689781B2 (en) Amino acid transport protein and its gene
WO2004100976A1 (en) Klotho protein, anti-klotho protein antibody and use thereof
CA2471656C (en) Antibody against fibroblast growth factor-23
US20090110677A1 (en) Polypeptide regulating phosphate metabolism, calcium metabolism, calcification and vitamin d metabolism and dnas encoding the same
US20070072236A1 (en) Novel type II Na/Pi cotransporters and type II Na/Pi cotransporter expression regulatory factors
TWI422593B (en) An anti-FGF23 antibody and a pharmaceutical composition containing the same
JPH06503949A (en) Novel insulin-like growth factor binding protein IGFBP-5
EA007985B1 (en) Immunoglobulin-domain containing cell surface recognition molecules
WO2001098493A1 (en) Insulin-like growth factor-binding protein
US20100330070A1 (en) Methods Of Making An Antibody And Compositions Thereof
WO1995017205A1 (en) Recombinant human thymopoietin proteins and uses therefor
JP5205440B2 (en) Amino acid transport protein and its gene
JPH11235186A (en) Nucleic acid coding for sodium channel of nervous tissue
JPH10505508A (en) ICAM-4 Materials and Methods
CA2265458A1 (en) Novel protein, its production and use
JP4346540B2 (en) Extravillous trophoblast cell-specific protein
AU780470B2 (en) Inhibitor of osteoclast precursor formation
JPH06293800A (en) Novel physiologically active substance, epimorphine, gene coding the same and antibody against epimorphine
WO2000037637A1 (en) Novel protein and dna thereof
WO2001005964A1 (en) Inhibitor of osteoclast precursor formation
JP2009031307A (en) Extravillous trophoblast-specific protein
JP2000228989A (en) New protein and its use
WO2000046368A1 (en) Transporter gene octn3
IE990007A1 (en) Cytotoxic Lymphocyte Maturation Factor

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

122 Ep: pct application non-entry in european phase