WO2001061004A1 - Nouvelle proteine comportant une sequence signal - Google Patents
Nouvelle proteine comportant une sequence signal Download PDFInfo
- Publication number
- WO2001061004A1 WO2001061004A1 PCT/JP2001/001083 JP0101083W WO0161004A1 WO 2001061004 A1 WO2001061004 A1 WO 2001061004A1 JP 0101083 W JP0101083 W JP 0101083W WO 0161004 A1 WO0161004 A1 WO 0161004A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- dna
- present
- cells
- smap
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/02—Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
Definitions
- the present invention relates to novel proteins derived from human fetal hepatocytes and having a signal sequence, their genes, and their production and use.
- proteins synthesized by cells are localized in various organelles in the cell, such as nucleus, mitochondria, cytoplasm, and proteins that function by binding to cell membranes, such as receptors and channel molecules. They can be classified into those that function by being secreted extracellularly, such as growth factors and cytokines. Among them, secretory proteins play important biological functions such as control of cell proliferation, differentiation and apoptosis, inflammatory response, and cell-cell interaction. For these reasons, secretory proteins have become suitable target molecules for diagnostic and therapeutic agents for various diseases.
- fetal tissues are more dynamic than adult tissues in which many cells are in the telogen phase, because of the expression of various genes, such as cell proliferation, regeneration, differentiation, and cell death. Change is happening.
- fetal liver is an essential place for hematopoiesis, and it is expected that many important genes related to life support will be expressed. For this reason, fetal liver plays an important role in the body; ! Molecules that are suitable targets for isolating genes and that are highly expressed in fetal liver are expected to play an important role in the main body. Disclosure of the invention The present invention provides a novel protein derived from human fetal hepatocytes and having a signal sequence, a gene thereof, a molecule functionally equivalent thereto, and production and use thereof.
- the present inventors screened a cDNA library derived from human fetal hepatocytes using a proprietary TMT method (International Publication No. W099 / 60113) to obtain a signal sequence.
- the present inventors have succeeded in isolating a cDNA encoding a novel protein having the following sequence (this clone was named "SMAP-1").
- SMAP-1 may be a factor that controls the proliferation and differentiation of hematopoietic cells.
- marked expression in liver and kidney suggests that it may be a molecule involved in liver and kidney fibrosis.
- SAP-1 has homology to yeast mannosyltransferase. Therefore, it is suggested that SMAP-1 may have a function as a glycosyltransferase.
- SMAP-1 may have a function as a glycosyltransferase.
- changes in the sugar chains of glycoproteins expressed on the cell surface are associated with metastasis of cancer and enhancement of malignancy.
- Mucl mucin a cell surface molecule containing a large number of 0-linked sugar chains, has been suggested as a tumor marker because of its association with enhanced malignancy in colorectal cancer and kidney cancer. Therefore, SMAP-1 may be involved in cancer cell invasion, metastasis, angiogenesis, etc.
- SAP-1 may be a molecule that regulates the mutual recognition and traffic control between immune cells such as lymphocytes and leukocytes and vascular epithelium. is there. This suggests the development of cancer metastasis inhibitors and immunomodulators targeting SMAP-1.
- AML-1 is an important transcription factor that controls the proliferation and differentiation of hematopoietic cells during the early stage of hematopoiesis. It is known that abnormalities due to chromosomal translocation are deeply involved in the development of leukemia. The presence of the AML-1 binding motif in the promoter region of SMAP-1 indicates that SMAP-1 functions in adult hematopoiesis (the process by which hematopoietic stem cells differentiate and proliferate to all blood cell lines in fetal liver). Suggests a possible molecule. It also suggests that abnormal AML-1 induces abnormal expression of SMAP-1 and may lead to the development of acute leukemia. Therefore, development of a therapeutic agent for leukemia targeting SMAP-1 is considered.
- GATA-1 is a transcription factor having an important role in the terminal differentiation of pro-erythroblasts to mature erythrocytes, the formation of platelets from megakaryocytes, and the process of formation of connective tissue-type cells. This suggests that SMAP-1 may regulate the proliferation, differentiation and apoptosis of red blood cells, megakaryocytes and connective tissue mast cells.
- GATA-2 is a transcription factor that plays important roles in the maintenance and proliferation of blood stem cells, differentiation of mast cells, and formation of the urinary system.
- SMAP-1 is a molecule that functions in the maintenance of blood cells, the differentiation of proliferating mast cells, and the formation of the urinary system.
- GATA-3 is an essential transcription factor for T cell differentiation and Th2 cell formation. This suggests that SMAP-1 is a molecule that functions in T cell differentiation and CD4 + T cell Th2 cell formation (promotes Th2 cell differentiation or suppresses Thl cell differentiation). I have.
- the presence of the GATA-1, 2, and 3 binding motifs in SMAP-1 is due to the development of drugs that promote hematopoietic stem cells and individual blood cells that target SMAP-1, as well as antiallergic drugs and immunomodulators. It implies that development is possible.
- AP-1 and NF- ⁇ are transcription factors that induce the production of inflammatory cytokines such as IL-1, TNF, and IL-6.
- IL-1 IL-1
- TNF inflammatory cytokines
- IL-6 IL-6 binding motifs
- SMAP-1 may be an inflammatory site whose expression is enhanced by AP-1 and NF-B during inflammation . Therefore, the development of anti-inflammatory drugs targeting SMAP-1 is also conceivable.
- SMAP-1 may regulate lymphocyte proliferation, differentiation and apoptosis. Therefore, development of an immunomodulator targeting SMAP-1 is considered.
- SMAP-1 may function during sperm differentiation and maturation.
- HSF heat shock factor
- SMAP-1 is supposed to play an important role in the subject, and is particularly useful as a target for drug development.
- the present invention relates to a novel protein "SMAP-1" which is derived from human fetal hepatocytes and has a signal sequence, its gene, and a molecule which is functionally equivalent thereto, and their production and use. , More specifically,
- a polynucleotide comprising at least 15 nucleotides complementary to a DNA consisting of the nucleotide sequence of SEQ ID NO: 1 or a complementary strand thereof,
- the present invention provides a gene “SMAP-1” encoding a novel protein having a signal sequence.
- SMAP-1 The nucleotide sequence of human-derived SMAP-1 cDNA is shown in SEQ ID NO: 1, and the amino acid sequence of the protein encoded by this cDNA is shown in ffi Column No. 2. As shown in column ⁇ : 1, human SMAP-1 cDNA has 0RF encoding a protein of 221 amino acids.
- the SMAP-1 protein of the present invention is a molecule isolated by the TMT method, and the presence of a region that can function as a transmembrane site or a signal sequence in its N-terminal region from analysis using analysis software S0SUI. Was predicted. In addition, analysis using soft P SORT for estimating the localization site predicted that the N-terminal region was a signal sequence. Therefore, it is considered that the SMAP-1 protein functions as a secretory protein having a signal sequence in the N-terminal region. As described above, SMAP-U is considered to be a molecule that plays an important function in living organisms based on its expression characteristics and structural characteristics, and is a useful target for drug development.
- the present invention also includes a protein functionally equivalent to the human SMAP-1 protein (SEQ ID NO: 2).
- Such proteins include, for example, variants of the human SMAP-1 protein, homologs of non-human organisms, and the like.
- “functionally equivalent” means that the target protein has a function as a protein having a signal sequence, like the SMAP-1 protein.
- a function may be, for example, a function described in International Publication No. WO 00/00610 or a function as a secretory protein. Whether or not the target protein is a secretory protein can be determined by the following method.
- a human SMAP-1 fusion gene with a gene encoding a commercially available peptide (eg, His-tag or FLAG) linked to the 3 'end of the human SMAP-1 gene is prepared. You.
- the fusion gene is introduced into animal cells (eg, COS cells) using an expression vector for animal cells (eg, pcDNA3 or pCOS-1), and human SMAP-1 is converted to a peptide. It is expressed as a fusion protein. Whether or not the human SMAP-1 fusion protein is secreted into the culture supernatant is evaluated by ELISA, Western blotting or immunoprecipitation using an antibody against the peptide.
- Secreted proteins have various industrial advantages. For example, when it is desired to obtain a certain recombinant protein, if the protein is expressed in a cell as a fusion protein with a secretory protein or a partial peptide capable of secreting the same, the fusion protein is extracellularly distributed. '; There is an advantage that the purification of the recombinant protein is easy. In addition, since many secreted proteins are useful drugs, they can be applied as drugs themselves.
- a method of introducing a mutation into a protein is known.
- those skilled in the art can use a site-directed mutagenesis method (Hashimoto-Gotoh, T. et al. (1995) Gene 152, 271-275, Zoller, MJ, and Smith, M. (1983) Methods Enzymol. 100).
- a protein functionally equivalent to the human SAP-1 protein (SEQ ID NO: 2) can be prepared by appropriately introducing a mutation into the amino acid. Amino acid mutations can also occur in nature. As described above, a protein having an amino acid sequence in which one or more amino acids are altered in the amino acid sequence of the human SMAP-1 protein (SEQ ID NO: 2) and which is functionally equivalent to the protein is also described in the present invention. It is contained in proteins. The number of amino acids to be mutated in such a mutant is usually within 50 amino acids, preferably within 30 amino acids, and more preferably within 10 amino acids (for example, within 5 amino acids). .
- the amino acid residue to be mutated is desirably mutated to another amino acid that preserves the properties of the amino acid side chain.
- the properties of amino acid side chains include hydrophobic amino acids (A, I, L, M, F, P, W, Y, V) and hydrophilic amino acids (R, D, N, C, E, Q, G, H, K ⁇ S, T), an amino acid having an aliphatic side chain (G, V, Shi P), an amino acid having a hydroxyl group-containing side chain (S, ⁇ , ⁇ ), and having a sulfur atom-containing side chain
- Amino acids ( ⁇ ) carboxylic acids and amino acids with amide-containing side chains (D, N ⁇ E, Q), amino-containing amino groups with side chains (R, K, ⁇ ), and aromatic-containing side chains
- Amino acids ( ⁇ , F, Y, W) can be mentioned (all brackets indicate one letter of amino acids).
- a protein in which a plurality of amino acid residues are added to the amino acid sequence of human SMAP-1 protein includes a fusion protein containing human SAP-1 protein.
- the fusion protein is a fusion of the human SMAP-1 protein and another peptide or protein, and is included in the present invention.
- the fusion protein is prepared by ligating the DNA encoding the human SMAP-1 protein (SEQ ID NO: 2) and the DNA encoding the other peptide or protein so that the frames match, and then ligating this to an expression vector. What is necessary is just to introduce
- Other peptides or proteins to be fused with the protein of the present invention are not particularly limited.
- peptides to be fused to the protein of the present invention include [FLAG (Ho ⁇ , ⁇ . ⁇ . Et al., BioTechnology (1988) 6, 1204-1210); 6 x His, lO x His, influenza agglutinin (HA), human c-myc fragment, VSV-GP fragment, P18HIV fragment, T7-tag, HSV- Known peptides such as tag, E-tag, fragment of SV40T antigen, lck tag, fragment of human tubulin, B-tag, fragment of Protein in C and the like can be used.
- proteins to be fused with the protein of the present invention include, for example, GST (Glutathione-S-transferase), HA (Influenza agglutinin), Immunoglobulin constant Region, / 3-galactosidase, MBP (maltose binding protein) and the like.
- a fusion protein is prepared by fusing a DNA encoding the peptide or protein, which has been prepared, with a DNA encoding the protein of the present invention, and homologating the fusion DNA prepared thereby. Can be.
- the present invention includes a protein which encodes a DNA encoding a human SMAP-1 protein and a DNA which hybridizes with a hyphen, and is functionally equivalent to the human SMAP-1 protein.
- proteins include, for example, homologs of humans and other mammals (eg, proteins encoded by mice, rats, rabbits, magpies, etc.).
- Hybridization conditions for isolating a DNA encoding a protein functionally equivalent to the human SMAP-1 protein can be appropriately selected by those skilled in the art.
- conditions for hybridization include, for example, 'low stringency conditions.
- Low stringent conditions are, for example, 42 in washing after hybridization.
- C 2 SSC, 0.1% SDS, preferably 50 ° (: 2 ⁇ SSC, 0.1% SDS. More preferred hybridization is high stringency.
- High stringent conditions include, for example, conditions of 65 ° C., O.lx SSC, and 0.1% SDS Under these conditions, the higher the temperature, the higher the homology c
- DNA having sex can Tomomachi be obtained efficiently, factors affecting the scan Bok Rinjienshi of High Priestess die See Chillon considered elements of the number 3 ⁇ 4 such temperature and salt concentration, the art 3 ⁇ 4 If so, it is possible to realize the same stringency by appropriately selecting these elements.
- a gene amplification method using primers synthesized based on the sequence information of DNA (SEQ ID NO: 1) encoding human SMAP-1 protein for example, a polymerase chain reaction (PCR) method is used. It is also possible to use and isolate.
- the proteins functionally equivalent to the human SMAP-1 protein encoded by the DNA isolated by the hybridization and gene amplification techniques are usually human SMAP-1 protein (SEQ ID NO: 2) and amino acid sequence. Have high homology.
- the protein of the present invention also includes a protein that is functionally equivalent to the human SMAP-1 protein and has high homology to the amino acid sequence shown in SEQ ID NO: 2. High homology usually means at least 65% identity, preferably 7%, at the amino acid level.
- More than 5% identity more preferably more than 85% identity, more preferably more than 95% identity.
- the algorithm described in the literature can be used. Good.
- the protein of the present invention may differ in amino acid sequence, molecular weight, isoelectric point, presence / absence and form of sugar chains, etc., depending on the cell, host, or purification method for producing the protein described below. However, as long as the obtained protein has a function equivalent to that of the human SMAP-1 protein, it is included in the present invention.
- the protein of the present invention when expressed in a prokaryotic cell, for example, Escherichia coli, a methionine residue is added to the N-terminal of the amino acid sequence of the original protein.
- the proteins of the present invention also include such proteins.
- the protein of the present invention can be prepared as a recombinant protein or a natural protein by methods known to those skilled in the art.
- a DNA encoding the protein of the present invention for example, a DNA having the nucleotide sequence of SEQ ID NO: 1
- an appropriate expression vector for example, a DNA having the nucleotide sequence of SEQ ID NO: 1
- the obtained transformant is recovered, and an extract is obtained.
- chromatography such as ion exchange, reverse phase, gel filtration, etc., or affinity chromatography in which an antibody against the protein of the present invention is immobilized on a column is used. Or by combining a plurality of these columns.
- the protein of the present invention When the protein of the present invention is expressed in a host cell (for example, an animal cell or Escherichia coli) as a fusion protein with glutathione S-transferase protein or as a recombinant protein to which a plurality of histidines are added,
- the expressed recombinant protein can be purified using a glutathione column or a nickel column. After purifying the fusion protein, if necessary, a region other than the target protein in the fusion protein can be cleaved with thrombin or Factor-I Xa and removed.
- the protein is a natural protein
- a method known to those skilled in the art for example, affinitino binding to an extract of a tissue or cell expressing the protein of the present invention, which is bound to an antibody that binds to the protein of the present invention described below, may be used.
- the protein can be isolated by purification using ram.
- the antibody may be a polyclonal antibody or a monoclonal antibody.
- the present invention also includes partial peptides of the protein of the present invention.
- the partial peptide of the present invention comprises an amino beta sequence of at least 7 amino acids or more, preferably 8 amino acids or more, and more preferably 9 amino acids or more.
- the partial peptide can be used, for example, for the production of antibodies against the protein of the present invention, for screening compounds that bind to g-white of the present invention, and for the screening of promoters and inhibitors of the protein of the Byon invention. obtain. In addition, it may be an antagonist or a competitive inhibitor of the protein of the present invention.
- the partial peptide of the present invention RJ] can be produced by a genetic engineering technique, a known peptide synthesis method, or by cleaving the protein of the present invention with an appropriate peptidase.
- the peptide may be synthesized by, for example, either a solid phase synthesis method or a liquid phase synthesis method.
- the DNA encoding the protein of the present invention is used for the primary production of the protein of the present invention in vivo and in riiro as described above, and is also caused by, for example, an abnormality in the gene encoding the protein of the present invention. It is also conceivable to apply the gene therapy to diseases that can be treated or diseases that can be treated with the protein of the present invention.
- the DNA of the present invention may be in any form as long as it can encode the protein of the present invention. That is, it does not matter whether it is cDNA synthesized from mRNA, genomic DNA, or chemically synthesized DNA. Also, as long as the protein of the present invention can be encoded, DA having an arbitrary nucleotide sequence based on the efficiency of the genetic code is included.
- the DNA of the present invention can be prepared by a method known to those skilled in the art. For example, it is prepared by preparing a cDNA library from cells expressing the protein of the present invention, and performing hybridization using a part of the sequence of the DNA of the present invention (for example,, 3 ⁇ 4, SEQ ID NO: 1) as a probe. it can.
- a part of the sequence of the DNA of the present invention for example, 3 ⁇ 4, SEQ ID NO: 1
- One of the cDNA libraries is, for example, the literature (Sam brook, J. et al., Molecular Clonings Cold Spring Harbor Laboratory Press
- RNA is prepared from cells expressing the protein of the present invention, cDNA is synthesized by reverse transcriptase, and oligo DNA is synthesized based on the sequence of the DiNA of the present invention (for example, SEQ ID NO: 1). It can also be prepared by performing a PCR reaction using this as a primer and amplifying the cDNA encoding the protein of the present invention.
- the translation region encoded by the cDNA can be determined, and the amino acid sequence of the protein of the present invention can be obtained. Further, by screening the genomic DNA library using the obtained cDNA as a probe, the genomic MA can be separated 1 ⁇ 1.
- mRNA is isolated from cells, tissues, and fluk organs (eg, tissues such as liver and kidney) that express the protein of the present invention.
- mRNA isolation can be performed by known methods, for example, guanidine ultracentrifugation (Chirgwin, JM et al., Biochemistry (1979) 18, 5294-5299), and AGPC method (Chomczynski, P. and Sacchi, N., Ana. l. Biochem. (1987) 162, 156-159), etc., prepare total RNA, and purify mRNA from total RNA using mRNA Purification Kit (Pharmacia). Alternatively, mRNA can be directly prepared by using the QuickPrep mRNA Purification Kit (Pharmacia).
- CDNA is synthesized from the obtained mRNA using reverse transcriptase.
- cDNA can also be synthesized using AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Corporation).
- AMV Reverse Transcriptase First-strand cDNA Synthesis Kit (Seikagaku Corporation).
- 5-Ampli FINDER RACE Kit manufactured by Clontech
- PCR polymerase chain reaction
- RACE method Frohman, Acad. Sci. USA (1988) 85, 8998-9002; Belyavsky, A. et al., Nucleic A According to cids Res. (1989) 17, 2919-2932
- cDNA can be synthesized and amplified.
- a target DNA fragment is prepared from the obtained PCR product and ligated to a vector DNA. Further, a recombinant vector is prepared from this, introduced into E. coli, etc., and a colony is selected to prepare a desired recombinant vector.
- the nucleotide sequence of the target DNA can be confirmed by a known method, for example, by the dideoxynucleotide chain minification method.
- the DNA of the present invention can be modified by a commercially available kit or a known method. Modifications include, for example, digestion with restriction enzymes, ⁇ insertion of adult lignonucleotides or appropriate DNA fragments, addition of a linker, start codon (ATG) and / or stop codon (TAA, TGA, or TAG). And the like.
- the DNA of the present invention specifically includes D consisting of base A at position 27 to base c at position 689 in the base sequence of SEQ ID NO: ⁇ : 1.
- the DNA of the present invention is a DNA that hybridizes with a DNA consisting of the nucleotide sequence shown in SEQ ID NO: 1 and that encodes a protein that is effectively equivalent to the protein of the present invention. Including.
- the conditions in the hybridization can be appropriately selected by those skilled in the art, and specifically, the conditions described above can be used. In these conditions, DA it c above hybridizing capable of obtaining a DNA ⁇ high 3 ⁇ 4 sex enough to raise the temperature is preferably naturally occurring DNA, for example cDNA or chromosomal DNA.
- the present invention also provides a vector into which the DNA of the present invention has been inserted.
- the vector of the present invention is useful for retaining the DNA of the present invention in a host cell or expressing the protein of the present invention.
- a vector for example, when E. coli is used as a host, the vector is amplified in E. coli (for example, JM109.DH5H, HB10K XLlBlue), etc.
- E. coli for example, JM109.DH5H, HB10K XLlBlue
- vectors examples include M13-based vectors, pUC-based vectors, pBR322, pBluescript, pCR-Script, etc.
- vectors for the purpose of subcloning and excision of cDNA, in addition to the above vectors, for example, pGEM-T, pDIRECT, pT7, etc.
- the present vector is useful.
- Escherichia coli in addition to the above-mentioned characteristics that the vector is amplified in Escherichia coli, in addition to the above-mentioned characteristics that the vector is amplified in Escherichia coli, in addition to the above-mentioned characteristics that the vector is amplified in Escherichia coli,
- a promoter that can be efficiently expressed in Escherichia coli such as the lacZ promoter (Ward et al., Nature (1989) 341, 544-546; FASEB J.
- the vector may include a signal sequence for polypeptide secretion.
- a signal sequence for protein secretion the pelB signal sequence (Lei, SP et al J. Bacteriol. (1987) 169, 4379) may be used when produced in E. coli periplasm.
- the introduction of the vector into the host cell can be carried out using the calcium chloride method or the electroporation method in general.
- vectors for producing the protein of the present invention include mammalian expression vectors (for example, pcDNA3 (manufactured by Invitrogen) and pEGF-B0S (cellulic Acids. Res.
- promoters necessary for expression in cells for example, the SV40 promoter (Muligan et al., Nature (1979) 277, 108), MMLV-LTR promoter, Yuichi ⁇ HIPROMO (Mizushima et al., Nucleic Acids Res. (1990) 18, 5322), CMV promoter
- a gene for selection of cell transformation for example, a drug-sensitive gene that can be identified by a drug (eg, neomycin, G418, etc.)
- a vector having the properties as c two for example, p Yuzururu pDR2, pBK-RSW pBK-CMV ⁇ pOPRSV ⁇ p0P13 like are Ru mentioned.
- a DHFR gene that complements ffl in a nucleic acid synthesis pathway-deficient CH0 cell is used.
- a vector for example, pCHOI
- MTX methotrexate
- transient expression of a gene use SV40 T antigen.
- An example is a method in which a COS cell having a gene to be expressed on a chromosome is used to transform a single vector (eg, pcD) having an SV40 origin of replication.
- expression vectors are selected as one of the selectable markers, such as aminoglycoside transferase (APH) gene, thymidine kinase (TK) gene, Escherichia coli xanthinguanine phosphoribosyltransferase (Ecogpt) gene, It can contain the dihydrofolate reductase (dhfr) gene and the like.
- APH aminoglycoside transferase
- TK thymidine kinase
- dhfr dihydrofolate reductase
- the DNA of the present invention is incorporated into an appropriate vector, for example, a retrovirus method, a liposomal method, a cationic liposome method, an adenovirus method.
- an appropriate vector for example, a retrovirus method, a liposomal method, a cationic liposome method, an adenovirus method.
- the method of introduction into a living body by a method is exemplified.
- the vector used include, but are not limited to, an adenovirus vector (for example, pAdexlcw) and a retrovirus vector (for example, pZIPneo), such as insertion of the DNA of the present invention into a vector.
- General gene manipulation can be performed according to a conventional method (Molecular Cloning, 5.61-5.63). Is also good.
- the present invention also provides a host cell into which the vector of the present invention has been introduced.
- the host cell into which the vector of the present invention is introduced is not particularly limited, and for example, Escherichia coli and various animal cells can be used.
- the host cell of the present invention can be used as a production system for production and expression of the protein of the present invention.
- Production systems for protein production include in and //? TO production systems. Examples of in vitro production systems include production systems using eukaryotic cells and production systems using prokaryotic cells.
- Kei cells include mammalian cells, for example, CHO (J. Exp. Med. (1995) 108, 945), COS ⁇ 3T3, myeloma, BHK (baby hamster kidney), HeLa, Vero, amphibian cells, for example, Africa Olive cells of the crab frog (Val le, et al., Nature (1981) 291, 358-340) or insect cells such as Sf9 and Sf2K Tn5 are known. Have been.
- CHO cells in particular, DHFR gene-deficient CH0 cells such as dhfr-CHO (Proc. Natl.
- the vector can be introduced into a host cell by, for example, the calcium phosphate method, DEAE dextran; a method using force ribosome D0TAP (manufactured by Boehringer Mannheim), an electroporation method, or a lipofection method. It is.
- a cell derived from Nicotima jujuta tapacam ( ⁇ '/ coi / a / w tabacua) is known as a protein production system, which can be callus cultured.
- Fungal cells include yeasts, for example, the genus Saccharomyces), for example, Saccharomyces I, Saccharomyces cerevisiae), filamentous fungi, tandem; ti; llus niger) is known.
- prokaryotic cells When prokaryotic cells are used, there are production systems using bacterial cells.
- the bacterial cells include E. coli, (for example, JM109, DH5, HB101, etc., of which ⁇ and Bacillus subtilis are known.
- the protein is obtained by transforming these cells with the desired DNA and culturing the transformed cells in iro.
- the culturing can be performed according to a known method.
- a culture solution of animal cells for example, DMEM MEM, RPI 1640, IMDM can be used.
- serum supplement such as fetal calf serum (FCS) can be used together, or serum-free culture may be performed.
- FCS fetal calf serum
- the pH during the culturing is preferably about 6 to 8.c
- the culturing is usually performed at about 30 to 40 ° C for about 15 to 200 hours, and the medium may be replaced, aerated, and agitated as necessary. Add.
- a production system using animals and a production system using plants can be mentioned. Intended for these animals or plants
- the DNA is introduced and the protein is produced and recovered in the body of the animal or plant.
- the “host” in the present invention includes these animals and plants.
- mice When using animals, there are production systems using mammals and insects. As mammals, goats, bushes, hidges, mice, and mice can be used (Vicki Glasser, SPECTRUM Biotechnology Applications, 1993). When a mammal is used, a transgenic animal can be used.
- the target DNA is prepared as a fusion gene with a gene encoding a protein that is specifically produced in milk, such as goat casein.
- the DNA fragment containing the fusion gene is introduced into the goat's R, and the second embryo is transferred to the female goat.
- the target protein can be obtained from milk produced by the transgenic goat born from the goat that has received the embryo or its progeny. Hormones may be used in transgenic goats as appropriate to increase the amount of milk containing proteins produced by transgenic goats (Ebert, KM et al., Bio / Technology (1994) 12, 699-7 02).
- silkworms can be used as insects, for example.
- the target protein can be obtained from the body fluid of the silkworm by infecting the silkworm with a baculovirus into which DNA encoding the target protein has been inserted (Sus drawing, M. et.al. , Nature (1985) 315, 592-594).
- tobacco when using a plant, for example, tobacco can be used.
- the DNA encoding the protein of interest is inserted into a plant expression vector, for example, PM0N530, and the second vector is introduced into a bacterium such as Agrobacterium tumefaciens.
- the bacteria are infected with tobacco, for example, Nicotiana tabacum, and the desired polypeptide can be obtained from the leaves of this tobacco (Julian .-C. A et a 1., Eur. J. Immunol. 1994) 24, 131-138).
- the protein of the present invention thus obtained can be isolated from the inside or outside of the host cell (such as a medium) and purified as a substantially pure and homogeneous protein.
- the separation and purification of the protein can be performed by any of the separation and purification methods used in ordinary protein purification, and is not limited in any way. For example, chromatographic columns, filters, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric focusing, dialysis, recrystallization, etc. When combined, proteins can be separated and purified.
- chromatography examples include affinity chromatography, ion-exchange chromatography, hydrophobic chromatography, gel chromatography, reversed-phase chromatography, and adsorption chromatography.
- chromatographs can be performed using liquid phase chromatography, for example, liquid phase chromatography such as HPLC and FPLC.
- the present invention also encompasses highly purified proteins using these purification methods.
- the protein can be arbitrarily modified or partially removed by applying an appropriate protein modifying enzyme before or after purification of the protein.
- an appropriate protein modifying enzyme for example, trypsin, chymotrypsin, lysylendopeptidase, protein kinase, glucosidase and the like are used.
- the present invention also provides an antibody that binds to the protein of the present invention.
- the form of the antibody of the present invention is not limited, and includes not only polyclonal antibodies but also monoclonal antibodies. Also included are antisera obtained by immunizing immunized animals such as rabbits with the protein of the present invention, polyclonal antibodies and monoclonal antibodies of all classes, and human antibodies and humanized antibodies obtained by genetic recombination. .
- the protein of the present invention used as a sensitizing antigen for obtaining an antibody is not limited to the animal species from which it is derived, but is preferably a protein derived from a mammal, for example, a human, mouse or rat. Particularly, human-derived proteins are particularly preferable.
- a human-derived protein can be obtained using the gene sequence or amino acid sequence disclosed herein.
- the protein used as the sensitizing antigen may be a complete protein or a partial peptide of the protein.
- the partial peptide of the protein include an amino group ( ⁇ ) terminal fragment and a carboxy (C) terminal fragment of the protein.
- antibody refers to an antibody that reacts with the full length or fragment of a protein.
- a gene encoding the protein of the present invention or a fragment thereof is inserted into a known expression vector system, and the host cell described in the present specification is transformed with the vector. It may be obtained by a known method and these may be used as a sensitizing antigen. Further, a cell expressing the protein, a lysate thereof, or a chemically synthesized protein of the present invention may be used as the sensitizing antigen. It is preferable that the short peptide is appropriately bound to a carrier protein such as keyhole lysine hepatocyanin, pepsin albumin, ovalbumin, etc. to form an antigen.
- a carrier protein such as keyhole lysine hepatocyanin, pepsin albumin, ovalbumin, etc.
- the mammal to be immunized with the sensitizing antigen is not particularly limited, but is preferably selected in consideration of compatibility with the parent cell used for cell fusion. Eyes, egrets, and primates are used.
- mice, rats, hamsters, and the like are used in many cases.
- a heronoid animal for example, a heron is used.
- monkeys are used as primates.
- monkeys of the lower nose for example, cynomolgus monkeys, rhesus monkeys, baboons, chimpanzees, etc. are used.
- Immunization of an animal with a sensitizing antigen is performed according to a known method.
- a sensitizing antigen is injected intraperitoneally or subcutaneously into a mammal.
- the sensitizing antigen is diluted to an appropriate amount with PBS (Phosphate-Buffered Sine) or physiological saline, and then turbidized.
- PBS Phosphate-Buffered Sine
- physiological saline physiological saline
- a normal adjuvant such as Freund's complete buffer
- -Mix the appropriate amount of 22-juvant, emulsify and administer to mammals. Thereafter, it is preferable to administer the sensitizing antigen mixed with an appropriate amount of Freund's incomplete adjuvant several times every 4 to 21 days.
- an appropriate carrier can be used at the time of immunizing a seizure antigen. Immunization is performed in this manner, and an increase in the desired antibody level in the serum is confirmed by a conventional method.
- the blood of a mammal sensitized with the antigen is removed after confirming that the desired antibody level in the serum has increased.
- the serum is separated from the second blood by a known method.
- blood containing a polyclonal antibody; 3 ⁇ 4 ; may be used, or if necessary, a fraction containing the polyclonal antibody may be further isolated from this serum and used.
- a fraction that recognizes only the protein of the present invention is obtained using an affinity column to which the protein of the present invention is coupled, and the fraction is further purified using a protein A or protein G column.
- immunoglobulin G or M can be prepared.
- the immune cells may be removed from the mammal and subjected to cell fusion.
- preferred immune cells used for cell fusion include spleen cells.
- the other parent cell to be fused with the immune cell is preferably a mammalian myeloma cell, and more preferably a myeloma cell that has acquired the properties for selecting fused cells by a drug.
- the cell fusion between the immune cells and myeloma cells is basically performed according to a known method, for example, the method of Milstein et al. (Galfre, G. and Milstein, C, Methods Enzymol. (1981) 73, 3-46). Can be done.
- the hybridoma obtained by cell fusion is selected by culturing it in a normal selective culture medium, for example, a HAT culture medium (a culture medium containing hypoxanthine, aminopterin and thymidine). Cultivation in the HAT culture solution is the desired hybrid. It is performed for a period of time sufficient to kill non-fused cells (non-fused cells), usually for several days to several weeks. Next, a conventional limiting dilution method is performed to screen and clone a hybridoma producing the desired antibody.
- a normal selective culture medium for example, a HAT culture medium (a culture medium containing hypoxanthine, aminopterin and thymidine). Cultivation in the HAT culture solution is the desired hybrid. It is performed for a period of time sufficient to kill non-fused cells (non-fused cells), usually for several days to several weeks. Next, a conventional limiting dilution method is performed to screen and clone a hybridoma producing
- human lymphocytes for example, human lymphocytes infected with EB virus, are sensitized with proteins or protein-expressing cells or lysates thereof in vitro. And sensitized lymphocytes are fused with human-derived myeloma cells capable of permanent division, such as U266, to obtain a hybridoma that produces the desired human antibody having protein binding activity.
- c human lymphocytes
- the obtained hybridoma is transplanted into the peritoneal cavity of a mouse, ascites is collected from the mouse, and the obtained monoclonal antibody is subjected to, for example, ammonium sulfate precipitation, protein A, protein G column, DEAE ion exchange chromatography, and the present invention. It can be prepared by purifying the above protein using a coupled affinity column or the like.
- the antibody of the present invention is used for purification and detection of the protein of the present invention, and is also a candidate for an agonist and an angonilist of the protein of the present invention. It is also conceivable to apply this antibody to antibody therapy for diseases involving the protein of the present invention.
- a human antibody or a human antibody is preferable in order to reduce immunogenicity.
- a transgenic animal having a human antibody gene repertoire is immunized with a protein serving as an antigen, a protein-expressing cell, or a lysate thereof to obtain antibody-producing cells, which are fused with myeloma cells.
- a human antibody to the protein can be obtained using a dormer (see International Publication Nos. W092--03918, 093-227, 094-02602, W094-25585, W096-33735 and W096-34096).
- cells in which immune cells such as sensitized lymphocytes producing antibodies have been immortalized with oncogenes may be used.
- the monoclonal antibody thus obtained can also be obtained as a recombinant antibody produced using a gene recombination technique (for example, Borrebaeck, AK and Larrick, JW, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the Unit ed Kingdom by MACMILLAN PUBLISHERS LTD, 1990).
- Recombinant antibodies are produced by cloning the DNA encoding them from hybridomas or immunized cells such as sensitized lymphocytes producing the antibodies, incorporating the DNA into an appropriate vector, and introducing it into a host.
- the present invention includes this recombinant antibody.
- the antibody of the present invention may be a modified antibody fragment thereof as long as it binds to the protein of the present invention.
- Fab, F (ab ') 2, Fv or a single chain Fv (scFv) in which an Fv of an H chain and an L chain are linked by an appropriate linker Huston, JS et al., Proc. Natl. Acad. Sci. USA (1988) 85, 5879-5883.
- the antibody is treated with an enzyme such as papain or pepsin to generate an antibody fragment, or a gene encoding these antibody fragment is constructed and introduced into an expression vector.
- Co MS et al., J. Immunol.
- modified antibody an antibody bound to various molecules such as polyethylene glycol (PEG) can be used.
- PEG polyethylene glycol
- the “antibody” of the present invention also includes these modified antibodies.
- Such a modified antibody can be obtained by subjecting the obtained antibody to chemical modification. These methods are already established in this field.
- the antibody of the present invention can be prepared using known techniques such as a CDR derived from a quinula antibody or a CDR derived from a non-human antibody comprising a variable region derived from a non-human antibody and a constant region derived from a human antibody. Constant region), a human antibody-derived FR (framework region), and a constant region.
- the antibody obtained as described above can be purified to homogeneity.
- the separation and purification of the antibody used in the present invention may be performed by the separation and purification methods used for ordinary proteins. If appropriate, chromatographic columns such as affinity chromatography, filters, ultrafiltration, salting-out, dialysis, SDS polyacrylamide gel electrophoresis, isoelectric focusing, etc. can be selected and combined as appropriate. It can be separated and purified (Antibodies: A Laboratory Manual. Ed Harlow and David Lane, Cold Spring Harbor Laboratory, 1988), but is not limited thereto.
- the concentration of the antibody obtained above can be measured by measuring the absorbance or by enzyme-linked immunosorbent assay (EUSA).
- Columns used for affinity chromatography include protein A column and protein G column.
- columns using a protein A column include Hyper D, POROS, Sepharose F.F. (Pharmacia), and the like.
- chromatographs other than affine chromatographs include, for example, ion exchange chromatography, hydrophobic 'I' main chromatograph, gel ⁇ I, reverse phase chromatograph, and adsorption chromatograph. (Strategies for Protein Purification and Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., Cold Spring Harbor Laboratory Press, 1996) c
- These chromatography methods are liquid phase chromatography such as HPLC and FPLC. This can be done using one.
- Methods for measuring the antigen-binding activity of the antibody of the present invention include, for example, measurement of absorbance, enzyme-linked immunosorbent assay (ELISA), EIA (enzyme-linked immunosorbent assay), and IA (enzyme-linked immunosorbent assay). Radioimmunity '; j! Ij standard method) or a fluorescent antibody method.
- ELISA enzyme-linked immunosorbent assay
- EIA enzyme-linked immunosorbent assay
- IA enzyme-linked immunosorbent assay
- Radioimmunity '; j! Ij standard method or a fluorescent antibody method.
- a secondary antibody that recognizes an antibody labeled with an enzyme, for example, alkaline phosphatase, and incubate the plate.After washing, add an enzyme substrate such as P-ditrophenyl phosphate and measure the absorbance. Can be used to evaluate the antigen binding activity.
- an enzyme substrate such as P-ditrophenyl phosphate and measure the absorbance.
- the protein a protein fragment, for example, a fragment consisting of its C-terminus may be used.
- BIA core Pharmacia
- the antibody of the present invention is brought into contact with the sample contained in the sample which is expected to contain the protein of the present invention, and an immune complex of the antibody and the protein is detected or measured.
- the method for detecting or measuring the protein of the present invention can be carried out.
- INDUSTRIAL APPLICABILITY The method for detecting or measuring a protein of the present invention can specifically detect or measure a protein, and thus is useful for various experiments and the like using proteins. It is also useful for the examination of Alzheimer's disease or cancer described below.
- the present invention also provides a polynucleotide comprising at least 15 nucleotides complementary to DNA encoding human SMAP-1 protein (SEQ ID NO: 1) or its complementary strand.
- complementary strand refers to one strand of a double-stranded nucleic acid composed of A: T (U for RNA) and G: C base pairs with respect to the other strand.
- the term “complementary” is not limited to:] when the sequence is completely complementary to at least 15 contiguous nucleotide regions, but is at least 70%, preferably at least 80, more preferably 90, and even more preferably 95 It suffices to have at least %% homology on the base sequence. Algorithms for determining homology may be used those described in the present ⁇ document c
- nucleic acids include probes and primers used for detection and amplification of DNA encoding the protein of the present invention, probes and primers for detecting expression of the DNA, and control of the expression of the protein of the present invention.
- Nucleotides or nucleotide derivatives e.g., antisense oligonucleotides or ribozymes, or MA etc.
- Such a nucleic acid can also be used for producing a DNA chip.
- the 3 ′ region can be complementary, and a restriction enzyme recognition sequence, a tag, or the like can be added to the 5 ′ region.
- the antisense oligonucleotide includes, for example, an antisense oligonucleotide that hybridizes at any position in the nucleotide sequence of SEQ ID NO: 1.
- the antisense oligonucleotide is preferably an antisense oligonucleotide for at least 15 or more consecutive nucleotides in the base sequence of SEQ ID NO: 1. More preferably, it is an antisense oligonucleotide in which at least 15 or more consecutive nucleotides contain a translation initiation codon.
- derivatives and modifications thereof can be used as the antisense oligonucleotide.
- modified product examples include a modified lower alkylphosphonate such as a methylphosphonate type or an ethylphosphonate type, a phosphorothioate modified product, a phosphoroamidate modified product, and the like.
- Antisense oligonucleotides include not only those whose nucleotides corresponding to nucleotides constituting a predetermined region of DNA or mRNA are all complementary sequences, but also those having nucleotides represented by SEQ ID NO: 1 in which DNA or mRNA and an oligo nucleotide are represented by SEQ ID NO: 1. As long as it can hybridize specifically to the sequence, it includes those having one or more nucleotide mismatches.
- the antisense oligonucleotide derivative of the present invention acts on cells producing the protein of the present invention to bind to DNA or mRNA encoding the protein, thereby inhibiting the transcription or translation of the protein or degrading the mRNA. Or by suppressing the expression of the protein of the present invention, thereby effectively suppressing the action of the protein of the present invention.
- the antisense oriconucleotide derivative of the present invention can be mixed with a suitable base material which is inactive against the derivative to form an external preparation such as a coating agent or a knipping agent.
- a suitable base material which is inactive against the derivative to form an external preparation
- excipients, isotonic agents, solubilizing agents, stabilizers, preservatives, soothing agents, etc. may be added to tablets, splinters, granules, capsules, ribosome capsules, It can be a lyophilized agent such as a propellant, a liquid, a nasal drop and the like. These can be prepared according to a conventional method.
- the antisense oligonucleotide derivative of the present invention is applied directly to an affected area of a patient, or is applied to a patient so as to be able to reach the affected area as a result of intravenous administration or the like.
- an antisense-encapsulated material that enhances durability and membrane permeability can be used.
- the dosage of the antisense oligonucleotide derivative of the present invention can be appropriately adjusted according to the condition of the patient, and a preferred amount can be used. For example, it can be administered in the range of 0.1 to 100 mg / kg, preferably 0.1 to 50 mg / kg.
- the antisense oligonucleotide of the present invention inhibits the expression of the protein of the present invention and is therefore useful in suppressing the biological activity of the protein of the present invention. Moreover, an expression inhibitor containing the antisense oligonucleotide of the present invention is useful in that it can suppress the biological activity of the protein of the present invention.
- the protein of the present invention is useful for screening for a compound that binds to the protein. That is, the protein of the present invention is brought into contact with a test sample expected to contain a compound that binds to the protein, and a compound having an activity of binding to the protein of the present invention is selected. It is used in a method of screening for a compound that binds to the protein of the present invention.
- the protein of the present invention used for the screening may be a recombinant protein or a protein of natural origin. It may be a partial peptide. It may also be a form expressed on the cell surface or a form as a membrane fraction.
- the test sample is not particularly limited, and includes, for example, cell extract, cell culture supernatant, fermented microorganism product, marine organism extract, plant extract, purified or crude protein, peptide, non-peptide ⁇
- the protein of the present invention can be used as a fusion protein having a recognition site of a monoclonal antibody.
- a commercially available epitope-antibody system can be used (Experimental Medicine! ⁇ , 85-90 (1995)).
- vectors that can express fusion proteins with o-galactosidase, maltose-binding protein, glutathione S-transferase, and green fluorescent protein t (GFP) have been marketed. I have.
- polyhistidine His-tag
- influenza agglutinin HA human c-my FLAG
- VSV-GP Vesicular stomatitis virus glycoprotein
- T7 genelO protein T7-tag
- human simple herpes Epitopes such as viral glycoproteins (HSV-tags) and E-tags (epitopes on monoclonal fuji) and monoclonal antibodies recognizing them can be used to screen for proteins that bind to the protein of the invention. It can be used as an epitope antibody system (Experimental Medicine 1 ⁇ , 85-90 (1995)).
- these antibodies are added to a cell lysate prepared using an appropriate surfactant to form an immunocompetent antibody.
- This immunoconjugate comprises the protein of the present invention, a protein capable of binding thereto, and an antibody.
- immunoprecipitation can also be performed using antibodies against the protein of the present invention.
- Antibodies against the protein of the present invention can be obtained, for example, by introducing a gene encoding the protein of the present invention into an appropriate Escherichia coli expression vector, expressing it in Escherichia coli, purifying the protein expressed, , Goat, It can be prepared by immunizing a chicken or the like. Alternatively, it can be prepared by immunizing the above animal with the synthesized partial peptide of the protein of the present invention.
- the immune complex can be precipitated using Protein A Sepharose or Protein G Sepharose.
- the protein of the present invention is prepared as a fusion protein with an epitope such as GST, it can be prepared by using a substance that specifically binds to these epitopes such as glutathione-Sepharose 4B.
- An immunoconjugate can be formed in the same manner as in the case of using the antibody of the protein ⁇ .
- SDS-PAGE is generally used to analyze immunoprecipitated proteins, and by using an appropriate concentration of gel, it is possible to analyze proteins bound by the molecular weight of the protein.
- the protein bound to the protein of the present invention is a radioisotope because it is difficult to detect the protein by ordinary staining methods such as Coomassie staining and silver staining.
- 35 Cells can be cultured in a culture solution containing S-methionine or S-cysteine, and 3 ⁇ 4 labeling proteins in the cells and stubbing out the cells can improve the detection sensitivity. Once the molecular weight of the protein is known, the protein of interest can be purified directly from the SDS-polyacrylamide gel and its sequence determined.
- a method for isolating a protein that binds to an S protein using the protein of the present invention is as follows: 'If they are listed, they can be performed using the West Western blotting method (Skolnik, E.Y. et al., Cell (1991) 65, 83-90). That is, a phage vector (human gtll, ZAP, etc.) is used from cells, tissues, and armpits (eg, liver and kidney) that are supposed to express the protein that binds to the protein of the present invention.
- a cDNA library was prepared, expressed on LB-agarose, and expressed on a filter.
- the protein of the present invention which has been immobilized, purified and labeled, is allowed to react with the above-mentioned filter, and the plaque expressing the protein bound to the protein of the present invention is detected by labeling.
- the method for labeling the protein of the present invention include a method utilizing the binding property of biotin and avidin, and a method for binding to the protein of the present invention or a peptide or polypeptide (for example, GST or the like) fused to the protein of the present invention in an allergic manner.
- the method include a method using an antibody, a method using a radioisotope, and a method using fluorescence.
- a 2-hybrid system using cells Yields, S., and Sternglanz,., Trends. Genet. (1994) 10, 286-292, Dalton S. , and T eisman R (1992) Characterization of SAP-1, a protein recruited by serum response factor to the c-fos serum response ele nient.Cell 68, 597-612, ⁇ MATCHMA KER Two-Hybrid System j, r ammalian MATCHMAKER Two-Hybrid Assay Kitj, “MATCHMAKER One-Hybrid Systemj (both made by Clontech), r HybriZAP Two-Hybrid Vector Systeny (manufactured by Straugene)”.
- the protein of the present invention or a partial peptide thereof is fused with an SRF DNA binding region or GAL4 D'A binding region and expressed in a mother cell to express a protein that binds to the protein of the present invention.
- VP16 or GA transcriptional activation region A cDNA library that expresses in a form fused with the region is prepared, introduced into the yeast cells, and the cDNA derived from the library is isolated from the positive clones detected (in the yeast cells, the cDNA of the present invention is isolated).
- the binding of the two activates the repo overnight gene, and a positive clone can be confirmed.
- a protein that binds to the protein of the present invention or a gene thereof can be prepared .
- a reporter gene used in the 2-hybrid system for example, the HIS3 gene Ade2 gene, LacZ gene, CAT gene, reresifnifase aid PAI-1 (Plasminogen activator inhibitor t ypel) gene, but is not limited thereto.
- Screening by the hybrid method can be carried out using mammalian cells in addition to yeast. Screening for a compound that binds to the protein of the present invention can also be performed using affinity chromatography.
- the protein of the present invention is immobilized on a carrier of an affinity column, and a test sample expected to express a protein that binds to the protein of the present invention is applied here.
- a test sample for example, a cell extract, a cell lysate, etc. may be mentioned.
- the column is washed to prepare a protein bound to the protein of the present invention. Can be.
- the obtained protein is analyzed for its amino ⁇ sequence, oligo DA is synthesized based on it, and DNA encoding the protein is obtained by screening cDNA library using 3 ⁇ 4D'A as a probe. Obtainable.
- a biosensor utilizing the surface plasmon resonance phenomenon may be used as a means for detecting or measuring the bound compound.
- a biosensor utilizing the surface plasmon resonance phenomenon can detect in real time the interaction between the protein of the present invention and a test compound as a surface plasmon resonance signal using a trace amount of protein and without labeling.
- BIAcore manufactured by Pharmacia
- Methods for isolating not only proteins but also compounds that bind to the protein of the present invention include, for example, immobilized protein of the present invention, synthetic compounds, and natural compounds.
- a method for screening a molecule that binds to the protein of the present invention by using a product bank or a random phage peptide display library, or a screening method using a high sloop 'solution by combinatorial chemistry technology (Wrighton C; Farrel l FX; Chang R; Kashyap AK; Barbone FP; Mulcahy LS; Johnson DL; Barrett RW; Jol liffe LK; Dower WJ., Sma 11 peptides as potent mimetics of the protein hormone erythropoietin, Sc ience (UNITED STATES) Jul 26 1996, Z73 p458-64, Verdine GL., The combina torial chemistry of nature.Nature (ENGLAND) Nov 7 1996
- the compound that can be isolated by the screening of the present invention is a candidate for a drug for regulating the activity of the protein of the present invention, and is used for the disease caused by abnormal expression or dysfunction of the protein of the present invention and the activity of the protein of the present invention. It can be applied to treatment of diseases that can be treated by control.
- a part of the structure of a compound that can be isolated using the screening method of the present invention is converted by addition, deletion and / or substitution into a compound that binds to the protein of the present invention. .
- the protein of the present invention is used in a human mammal, for example, a mouse, a rat, a guinea pig, a heron, a chicken, a cat, a dog, a hidge, a bush, a monkey, a monkey.
- a human mammal for example, a mouse, a rat, a guinea pig, a heron, a chicken, a cat, a dog, a hidge, a bush, a monkey, a monkey.
- sterile with tablets or capsules, elixirs, microcapsules, orally with water or other pharmaceutically acceptable liquids as needed; , Or S can be used non-permanently in the form of a suspension.
- pharmacologically acceptable carriers or vehicles such as sterile water or saline, vegetable oils,? Appropriately combined with preservatives, suspending agents, surfactants, stabilizers, flavors, excipients, vehicles, preservatives, binders, etc., in the form of unit doses required for accepted pharmaceutical practice. It is conceivable to formulate by mixing. The amount of the active ingredient is intended to ensure that an appropriate dose within the specified range is obtained c
- excipients that can be added include gelatin, corn starch, tragacanth gum, binders such as gum arabic, excipients such as crystalline cellulose, corn starch, Swelling agents such as gelatin, alginic acid, Lubricants such as magnesium stearate, sweeteners such as sucrose, lactose or saccharin, and flavoring agents such as peppermint, cocoa oil or cherry may be used.
- a liquid carrier such as oils and fats can be further contained in the above-mentioned materials.
- Sterile compositions for injection can be formulated according to normal pharmaceutical practice using a vehicle such as distilled water for injection.
- Aqueous solutions for injection include, if clear, physiological & saline, isotonic containing glucose and other adjuvants, such as D-sorbitol, D-mannose, D-mannitol, sodium chloride,
- a solubilizing agent for example, an alcohol, specifically phenol, alcohol, for example, propylene glycol, polyethylene glycol, a nonionic surfactant, for example, polysorbate 80 (TM), HCO-50 Is also good.
- Oily liquids include sesame oil and soybean ash, and may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizer. It may also be used in combination with a buffer, for example, a phosphate buffer, a sodium acetate buffer, a soothing agent, for example, proforce hydrochloride, a stable agent, for example, benzyl alcohol, phenol, or an antioxidant. Good. Prepared injections; 'fill with appropriate ampoules at night.
- the dose and administration method vary depending on the patient's body weight, age, symptoms, etc., but those skilled in the art can appropriately select the dose of the protein of the present invention.
- the normal adult assuming a body weight of 60 kg
- the dose of the compound that binds to the protein of the present invention or the compound that modulates the activity of the protein of the present invention varies depending on the symptoms. It is believed to be about 0.1 to 100 mg per day, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
- the single dose varies depending on the subject of administration, target organ, symptoms and administration method.
- injection usually in adults (with a body weight of 60 kg)
- administration method usually in adults (with a body weight of 60 kg)
- the dose can be administered in terms of the amount converted per 60 kg body weight or the amount converted per body surface area.
- FIG. 1 shows the nucleotide sequence of human SMAP-1 cDNA and the deduced amino acid sequence. The putative signal sequence is indicated by an underline.
- FIG. 2 shows a comparison of the nucleotide sequence of human SMAP-1 arsenic SDF2. The same base is indicated by “*”.
- FIG. 3 is a continuation of FIG.
- FIG. 4 shows the ratio of the amino acid sequences of human SAP-1 and SDF2. The same amino acid is indicated by “*”.
- FIG. 5 is an electrophoretic photograph showing the results of praying the expression of SMAP-1 in various human tissues and 'cell lines by RT-PCR.
- Ml 100bp ladder marker
- 1 brain
- 2 heart
- 3 liver
- 4 spleen
- 5 kidney
- 6 lymphocyte
- M2 lkbp ladder marker.
- the sample was electrophoresed on a 1.2 agarose gel and stained with ethidium ubiquitide.
- CDNA was synthesized from human fetal liver polyA + mRA (manufactured by Clontech), and incorporated into TMT 3 ⁇ 4Genbe Yuichi pTMT-shPM kappa to prepare a human fetal liver cD'A library.
- the cDNA library was prepared using a cDNA synthesis kit (STRATAGE E cDNA synthesis kit). Basically, it was prepared according to the protocol of STRATAGENE's cDNA synthesis kit, but the following points were improved.
- the reverse transcriptase used was Superscript II manufactured by GIBC0-BRL: Takara's Hindlll-Smal site adapter was used to add the 5 'end of the cDNA.
- the improved oligo dT primer was used as a primer for the first strand synthesis.
- the sequence is shown in SEQ ID NO: 3. That is, by using an oligo dT primer to which a BamHI site was added, the synthesized cDNA was inserted into the Hindlll / BamHI site of the TMT expression vector in the sense direction. In addition, a stop codon was introduced in three frames before the ⁇ ⁇ ' ⁇ ⁇ sequence of the Oligo dT-Bramer to prevent translation even if the cDNA was inserted into the TMT expression vector in the opposite direction. Further, c Kara ⁇ is who was Size sep 400 Spun Coi ⁇ made on Pharmacia Biotech Ne for size fraction
- a specific method for preparing a GDNA library is as follows. First, 5 ⁇ g of mRNA is used as a starter, and the first strand is transcribed from the 3 ′ polyA tail using a reverse transcriptase (Superscript II manufactured by GICO-BRL) using the above primers. Was synthesized. Next, after synthesizing the second strand with DNA polymerase, both ends of the cDNA were blunt-ended, and a Hindi I-SmaI site adapter (Takara) was added. After cutting the 3 'end of the cDNA with BamHI, a size fractionation (Pharmacia Biotech Co., Ltd.
- Size sep 400 Spun Column was performed to eliminate cDNA fragments of 0.4 kb or less in size.
- the recovered cDNA was incorporated into Hindi 11-BamHI site of TMT expression vector shPM kit kappa, and E. coli DH10B (GIBCO-B It was introduced into RL's Elect Max Max DH10B) by the electroporation method to prepare a human fetal liver cDNA library.
- the cDNA library was pooled in 2 ⁇ 10 5 clones per pool, and 5 pools (pool Nos. 1 to 5) were subjected to screening by the TMT method.
- Example 2 Screening of cDNA fetal liver library by TMT method 1) Gene transfer into COS-7 cells-1st time-
- Plasmid DNA prepared from each pool (pool Nos. 1 to 5) was transfused into C0S-7 cells using FuG EXE TM 6 (Boehringer Mannheim) at 4 ⁇ g each.
- DMEM culture solution GIBC0-BRL were incubated overnight at 37 ° 53 ⁇ 4 C0 2 of the conditions at the company made
- a panning dish coated with goat anti-mouse IgG antibody (goat anti-mouse IgG (H + L chain) manufactured by Dainippon Pharmaceutical Co., Ltd.) was placed in a literature (Seed, B. et al., Pro Natl. Sc. USA. (1987) 84, 3365-3369). That is, a goat anti-mouse IgG antibody was added to 50 mM Tris-HCl (pH 9.5) at a concentration of lO ⁇ g / ml, and 3 ml of the antibody solution prepared as described above was added to a 60 thigh diameter dish (Falcon 1007), and incubated for 3 hours at room temperature. After washing three times with a 0.15 M NaCl solution, PBS containing 5 fetal calf serum, imM EDTA, and 0.02% NaN 3 was added, the cells were blocked, and used for the following panning. 3) Panning
- the transfected COS-7 cells were detached with PBS containing ImM EDTA, and washed once with PBS containing 5% fetal bovine serum.
- FACS buffer (2 fetal bovine serum and 0.05% Na (PBS containing PBS). 0. 5 ⁇ G /] 0 6 cel l in addition to soluble IL-6 R the cell turbidity was incubated on ice for 90 minutes. Then, cells were washed twice with FACS buffer and foremost, FACS Roh Nigoshi re 3 ⁇ 4 sofa one, 0. 35 ⁇ G viii 0 ⁇ cel l cells murine anti-IL-6 receptor antibody MT- 18 in ⁇ In addition to the above, the sample was incoupled for 30 minutes on ice.
- the cells were CLEANING twice with FACS buffer one, 2 ml of 5% fetal bovine serum ⁇ '0. 02% NaN:, Nigoshi in PBS containing, catcher formic antimouse 6 antibody co - interference Bruno ring and panning Added to the plate.
- chloramphenicol manufactured by Wako Pure Chemical Industries
- a final concentration of 150 g / ml was added to suppress the growth of E. coli and promote DNA synthesis.
- the cells were collected from 100 ml of the culture solution, and the cells were suspended in 5 ml of ice-aged 20% sucrose / 50 mM Tris-Hcl (PH.8) buffer. 1 ml of 5 rag / mi lysozyme (manufactured by Sigma) was added, and the mixture was placed on ice for 5 minutes.
- the culture solution was removed, and the suspension of the protoblasted Escherichia coli treated as described above was added to a 5 ml dish.
- the dish was placed on a swinging bucket and centrifuged at 2,000 x 10 minutes at room temperature. After aspirating and removing the supernatant, apply 1.5 ml of 50% polyethylene glycol 1500 (Wako Pure Chemical Industries, Ltd.PEG1540) / DMEM solution evenly to the denotch, immediately tilt the dish, and aspirate the excess polyethylene glycol solution.
- the dish was put in the DMEM solution, and the dish was washed.
- - DMEM medium containing 50i / ml Genyumycin (GIBCO-BRL) and 10% fetal bovine serum was placed in a 5ml dish and incubated at 37 ° C for 5 hours. After that, the culture medium was changed, and the cells were further cultured at 37 ° C for 3 days.
- the COS cells thus fused with the protocol were detached with PBS containing ImEDTA, washed once with PBS containing 5 fetal calf serum, and then washed with FACS buffer (2% fetal bovine serum and 0.05% NaN 3 Including PBS)! It became cloudy and was subjected to a second panning. Collect the DNA from the panning plate and transfer it to COS-7 cells for the third time.
- pool No. 5 was selected at random from a plate for Thailand-Yuichi Check, and each was cultured at 37 ° C in 2 ml of LB-ampicillin (100 ⁇ g / ml) liquid medium at 37 ° C. DNA was prepared. Next, the analysis of the fragment sequences using restriction ⁇ the Smal and BamHI, screened clones having the correct insert sequence was performed to determine the nucleotide sequence from 5 'fflij c
- Clone HFL0304h showed high ffl homology at the nucleotide sequence level with the sequence registered in US Pat. No. 5,576,423.
- Clones HFL0304h showed high homology with yeast protein mannosyltransferase at the amino acid level.
- the nucleotide sequence described in U.S. Pat. No. 5,576,423 is the cDNA sequence of SLAM2, which is a splice variant of SLA1, and a part of the nucleotide sequence of clone HFL0304h is 1040 bp downstream from the 5,2 end of SL2. It was identical to the sequence (3, untranslated region of SL 2 cDNA). The nucleotide sequence at the 5 'end of clone HFL0304h was clearly different from SLAM2, and the nucleotide sequence of HFL0304h encoded a very long amino acid sequence (219 amino acid residues). Again, a database search was performed for the latest database.
- SLAM2 was confirmed on chromosome 1, but no site with high homology to clone HFL0304h was found on chromosome 1. It was confirmed on chromosome 22. This suggests that the SLAM2 cDNA disclosed in U.S. Patent No. 5,576,423 may be an artifact created by fusing the original SLAM2 gene on chromosome 1 with the HFL0304h gene on chromosome 22. It was suggested. This is possible when creating a cDNA library.
- clone HFL0304h is not a 3 'untranslated region of SLAM2 cDNA, but a novel gene that actually encodes a protein having homology to human SDF-2 existing on chromosome 22. became.
- the 5 'RACE method was performed using a 5' RACE kit (SMART T "RACE cDNA Amplification kit” manufactured by Clonetech). That is, a 500 ng human fetal liver poiyA + mRM was used to prepare the oligo-dT-Brimer attached to the kit.
- SMART T "RACE cDNA Amplification kit” manufactured by Clonetech. That is, a 500 ng human fetal liver poiyA + mRM was used to prepare the oligo-dT-Brimer attached to the kit.
- the solution was subjected to PCR using a primer specific to clone HFL0304h, followed by PCR using 1.251 first-strand cDNA, 2.5h1 lOxPCR buffer, and 51 Q-solution 0.15 l Taq polymerase, 0.5 ⁇ 1 dNTP mix, 2.5 1 specific primer (with SMART RACE kit), 0.51 10 1M HFL0304h specific primer (HFL030 4HR3, SEQ ID NO: 4) ), and DDW to 12.625 ⁇ 1 added, total 25 ⁇ as 1, the following 2 minutes the PCR reactions in a thermal cycler one c primary modification was performed using ABI2400 is 95 ° C under conditions and 30 cycles of 3-step PCR (30 seconds at 94 ° C, 30 seconds at 60 ° C, 30 seconds at 72 ° C) The PCR was then performed at 72 ° C for 7 minutes to complete the primary PCR, followed by diluting the PCR product 10-fold, nesting it into a gun and using
- a secondary PCR was performed using (HFL0304HR4, SEQ ID NO: 5), ie, a primary PCR solution of 11, a lOxPCR bath sofa of 2.51, a Q-solution of 5 ⁇ 1, 0.125 1 Taq Po Remelase, 0.5 ii dNTP mix, 0.5 ⁇ 1 nested primer (supplied with SMART RACE kit), 0.5 i 10 M HFL0304h specific primer (HFL0304HR4, SEQ ID NO: 5), and 14.875 / 1 DDW PCR reaction was performed using AB 12400 under the same conditions as the primary PCR as 25-1. The band amplified by the secondary PCR was analyzed by 1.2 agarose gel electrophoresis, and stained with ethidium umide.
- the secondary PCR reaction solution was purified by a spin column, followed by direct sequencing with Primer-1 HFL0304HR4, and analysis with an AB 17700 DNA sequencer. By joining the obtained 5 'upstream sequence and HFL0304h, the full-length i3 ⁇ 4 sequence was revealed, and this gene was named SAP-1 (Fig. 1).
- FIG. 1 shows the nucleotide sequence and deduced amino acid sequence of SMAP-1.
- Exon 1 is up to 14 bp, exon 2 from 215 to 411 bp, and exon 3 from 412 to 816 bp.
- a primer was designed to specifically amplify SMAP-1 by PCR (HFL0304HF and HFL0304 dishes, own column numbers: 6, 7), and the current frequency information was obtained by RT-PCR. Analysis was performed.
- PCR was performed using Takasaka's Multiple Choice First-Strand cDNA set (Nippon Gene Osakari). Both the PCR reagents and the reaction conditions are the same as those described in Example 4 (HFL0304h and SDF2 homologs by 5 'RACE method), and are included in ⁇ set CH-1101 above.
- the first-strand cDNA of the 6S (human K, heart, liver side, spleen, kidney, and lymphocyte) tissues was type III and analyzed. As a result, as shown in FIG. It was confirmed that the gene was expressed in the tissue (the amplified gene fragment (394 bp) was confirmed to be free of contaminating other amplified bands such as SDF-2 by the enzyme treatment with BamHI). In particular, strong expression was observed in the liver and kidney vagina.
- SMAP-1iti encoding a novel protein having a signal sequence.
- SMAP-1 may be an important molecule involved in the maintenance of adult and adult survival due to its 3 ⁇ 43 ⁇ 4 characteristics.
- SMAP-1 may have a function as a glycosyltransferase and may be involved in the proliferation and differentiation of blood cells. It is possible that 'is functioning in'.
- the protein encoded by the SMAP-1 gene is considered to be an important molecule in the main body, and thus is useful as a target for drug development.
- compounds that regulate the function of SMAP-1 are useful as drugs. Application is expected.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Biochemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Gastroenterology & Hepatology (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Biophysics (AREA)
- General Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Immunology (AREA)
- Cell Biology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Peptides Or Proteins (AREA)
Description
Claims
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP01904475A EP1260584A4 (en) | 2000-02-15 | 2001-02-15 | PROTEIN WITH SIGNAL SEQUENCE |
US10/204,033 US7067297B2 (en) | 2000-02-15 | 2001-02-15 | Mannosyltransferase polypeptides and polynucleotides encoding them and methods for making and using them |
AU2001232305A AU2001232305A1 (en) | 2000-02-15 | 2001-02-15 | Novel protein having signal sequence |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2000-41459 | 2000-02-15 | ||
JP2000041459 | 2000-02-15 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2001061004A1 true WO2001061004A1 (fr) | 2001-08-23 |
Family
ID=18564696
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2001/001083 WO2001061004A1 (fr) | 2000-02-15 | 2001-02-15 | Nouvelle proteine comportant une sequence signal |
Country Status (4)
Country | Link |
---|---|
US (1) | US7067297B2 (ja) |
EP (1) | EP1260584A4 (ja) |
AU (1) | AU2001232305A1 (ja) |
WO (1) | WO2001061004A1 (ja) |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1996001843A1 (fr) * | 1994-07-08 | 1996-01-25 | Ono Pharmaceutical Co., Ltd. | Polypeptide produit par des cellules du stroma |
WO1998039446A2 (en) * | 1997-03-07 | 1998-09-11 | Human Genome Sciences, Inc. | 70 human secreted proteins |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20030027272A1 (en) * | 1997-09-18 | 2003-02-06 | Genentech, Inc. | Secreted and transmembrane polypeptides and nucleic acids encoding the same |
US7125959B2 (en) * | 1998-05-15 | 2006-10-24 | Genentech, Inc. | PRO4405 polypeptides |
PT1609863E (pt) * | 1999-03-23 | 2009-01-02 | Genentech Inc | Polipéptidos segregados e transmembranares e ácidos nucleicos que os codificam |
-
2001
- 2001-02-15 AU AU2001232305A patent/AU2001232305A1/en not_active Abandoned
- 2001-02-15 EP EP01904475A patent/EP1260584A4/en not_active Ceased
- 2001-02-15 WO PCT/JP2001/001083 patent/WO2001061004A1/ja not_active Application Discontinuation
- 2001-02-15 US US10/204,033 patent/US7067297B2/en not_active Expired - Fee Related
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1996001843A1 (fr) * | 1994-07-08 | 1996-01-25 | Ono Pharmaceutical Co., Ltd. | Polypeptide produit par des cellules du stroma |
WO1998039446A2 (en) * | 1997-03-07 | 1998-09-11 | Human Genome Sciences, Inc. | 70 human secreted proteins |
Non-Patent Citations (2)
Title |
---|
HAMADA T. ET AL.: "Isolation and characterization of a novel secretory protein, stromal cell-derived factor-2(SDF-2) using the sequence trap method", GENE, vol. 176, no. 1-2, 1996, pages 211 - 214, XP004070231 * |
See also references of EP1260584A4 * |
Also Published As
Publication number | Publication date |
---|---|
EP1260584A1 (en) | 2002-11-27 |
AU2001232305A1 (en) | 2001-08-27 |
EP1260584A4 (en) | 2004-05-19 |
US7067297B2 (en) | 2006-06-27 |
US20030138802A1 (en) | 2003-07-24 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP5634674B2 (ja) | 新規ヘモポエチン受容体蛋白質、nr10 | |
WO2001023556A1 (fr) | Nouvelle proteine receptrice d'hemopoietine (nr12) | |
JP5395767B2 (ja) | トランスポーター遺伝子oatp−b、c、d、およびe | |
WO2000023610A1 (fr) | Methode de criblage de composes empechant la transduction de signal de cytokines inflammatoires | |
WO2001009316A1 (fr) | Nouveaux genes codant la proteine kinase / proteine phosphatase | |
WO2002077230A1 (fr) | Variants d'epissage nr10 | |
WO2001009345A1 (fr) | Nouveaux genes codant des proteines kinase et des proteines phosphatase | |
US20090197317A1 (en) | TSG-Like Gene | |
WO2001061004A1 (fr) | Nouvelle proteine comportant une sequence signal | |
WO2002020770A1 (fr) | Methode de criblage d'un agent antitumoral a l'aide d'une interaction entre une proteine arf et une proteine hk33 | |
US20070105143A1 (en) | Novel bHLH type transcription factor genes | |
EP1120426A1 (en) | Novel g protein-coupled receptors | |
JP4590107B2 (ja) | 新規胎児性遺伝子 | |
JP2008099690A (ja) | 新規ヘモポエチン受容体蛋白質、nr12 | |
US7632935B2 (en) | DNA encoding a mast cell-derived membrane protein | |
WO2005030955A1 (ja) | Nk細胞に発現するタンパク質 | |
WO2001012808A1 (fr) | Nouveaux genes 584 de la famille frizzled | |
JP4499926B2 (ja) | 腫瘍抑制遺伝子 | |
JP4441121B2 (ja) | 新規トリプシンファミリーセリンプロテアーゼ | |
WO2001066720A1 (fr) | Genes de souris d'origine adipocyte | |
WO2001027270A1 (fr) | Gene ys68 associe a l'hematopoiese precoce | |
JP4942904B2 (ja) | zincフィンガードメインを有する新規転写因子 | |
WO2001083738A1 (fr) | Facteur de regulation de differenciation et de determination du sexe | |
US7173111B2 (en) | Wt1-interacting protein WTIP | |
JP2002000279A (ja) | 初期造血に関わるys68遺伝子 |
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 BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG |
|
DFPE | Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101) | ||
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
ENP | Entry into the national phase |
Ref country code: JP Ref document number: 2001 560374 Kind code of ref document: A Format of ref document f/p: F |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2001904475 Country of ref document: EP |
|
WWP | Wipo information: published in national office |
Ref document number: 2001904475 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 10204033 Country of ref document: US |
|
REG | Reference to national code |
Ref country code: DE Ref legal event code: 8642 Ref country code: DE Ref legal event code: 8642 |
|
WWR | Wipo information: refused in national office |
Ref document number: 2001904475 Country of ref document: EP |
|
WWW | Wipo information: withdrawn in national office |
Ref document number: 2001904475 Country of ref document: EP |