WO2007086342A1 - METHOD OF DETECTING CANCER USING SPLICING VARIANT OF c-myc GENE TRANSCRIPTIONAL REGULATOR FIR OR FOUR-BASE REPETITIVE SEQUENCE IN INTRON 2 - Google Patents

METHOD OF DETECTING CANCER USING SPLICING VARIANT OF c-myc GENE TRANSCRIPTIONAL REGULATOR FIR OR FOUR-BASE REPETITIVE SEQUENCE IN INTRON 2 Download PDF

Info

Publication number
WO2007086342A1
WO2007086342A1 PCT/JP2007/050915 JP2007050915W WO2007086342A1 WO 2007086342 A1 WO2007086342 A1 WO 2007086342A1 JP 2007050915 W JP2007050915 W JP 2007050915W WO 2007086342 A1 WO2007086342 A1 WO 2007086342A1
Authority
WO
WIPO (PCT)
Prior art keywords
fir
cancer
protein
dna
seq
Prior art date
Application number
PCT/JP2007/050915
Other languages
French (fr)
Japanese (ja)
Inventor
Hideaki Shimada
Kazuyuki Matsushita
Takeshi Tomonaga
Fumio Nomura
Takenori Ochiai
Original Assignee
National University Corporation Chiba University
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 National University Corporation Chiba University filed Critical National University Corporation Chiba University
Priority to JP2007513551A priority Critical patent/JP4806776B2/en
Publication of WO2007086342A1 publication Critical patent/WO2007086342A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present invention relates to a splicing variant of a c-myc gene transcription repressor FIR or a cDNA encoding the same or a mRNA corresponding thereto, a DNA containing a 4-base repeat sequence in intron 2 of the FIR gene, and the presence thereof
  • the present invention relates to a cancer detection method to be detected and a measurement kit to be used for the method.
  • c-Myc protein is extremely important for cell life activity and is a transcription factor that controls cell proliferation, differentiation and cell death (apoptosis). It is strictly controlled by transcription factors.
  • the c-myc gene senses physical and mechanical changes in double-stranded DNA generated by transcription, not through protein synthesis, in order to respond quickly to intracellular and external stimuli and maintain homeostasis. There is an unknown mechanism, which is thought to keep the transcription level constant. So far, any site force upstream of the c-myc gene 3 ⁇ 4-myc gene has been examined for its influence on transcription, and only a few tens of bases 1.5 kb upstream of the transcription start site of the c-myc gene.
  • FUSE Fluor Upstream Element
  • This FBP protein has a strong transcriptional activity (2_4), and FIR was identified by the Yeast Two-Hybrid System as a protein that binds to this FBP (5).
  • FIR FBP Interacting Repressor
  • c-Myc protein is thought to suppress c-myc gene transcription by suppressing the helicase activity of TFIIH (6).
  • Patent Document 1 Western blot and RT-PCR using anti-FIR antibodies have been carried out. Describes that FIR expression in cancer tissues is increased.
  • Patent Document 1 International Publication No. WO2004Z018518 Pamphlet
  • Patent Document 2 International Publication No. WO2004Z018679 Pamphlet
  • An object of the present invention is to provide more effective and reliable detection / diagnosis of cancer, particularly digestive organ cancer represented by colorectal cancer, and to provide a genetic diagnosis system and gene for such cancer.
  • Treatment The purpose is to provide various biomolecules used in the development of clinical research systems.
  • amino acid sequence shown in SEQ ID NO: 1 the amino acid sequence in which one or several amino acids are deleted, substituted or added, or homology with the amino acid sequence shown in SEQ ID NO: 1 is 95% or more
  • DNA comprising a continuous base sequence including a base sequence indicating a gene polymorphism in a 4-base repeat sequence in intron 2 of the FIR gene.
  • Aspect 8 The protein according to Aspect 1 or 2, or a fragment peptide thereof, the cDNA according to Aspect 3 or 4, or the mRNA corresponding thereto, or an oligonucleotide comprising a part thereof, or any one of Aspects 5-7 A method for detecting cancer, comprising measuring the presence of DNA comprising a continuous base sequence comprising the 4-base repeat sequence described in the section.
  • Aspect 9 The detection method according to Aspect 8, using an antigen-antibody reaction with an antibody specific for FIR or a splicing variant thereof.
  • Aspect 12 The detection method according to Aspect 11, which is RT—PCR.
  • Aspect 13 The detection method according to Aspect 11, which is real-time quantitative PCR.
  • a detection method which utilizes PCR using a primer set for amplifying a nucleic acid molecule comprising the DNA according to any one of Aspects 5-7.
  • Aspect 15 The detection method according to any one of Aspects 8 to 14, wherein the cancer is a cancer of the extinct organ.
  • Aspect 16 The detection method according to any one of Aspects 8 to 14, wherein the cancer is colorectal cancer.
  • the measurement kit according to embodiment 17, comprising a primer set for amplifying the nucleic acid molecule according to any one of -7 as an element.
  • the splicing variant protein lacking FIR exon 2 which is a transcriptional repressor of the c myc gene of the present invention is specifically expressed only in cancer thread and tissue, and encodes an external protein and the protein.
  • m RNA (cDNA) can be significantly detected at level.
  • cancer detection 'diagnosis can be performed based on the gene polymorphism in the 4-base repeat sequence in intron 2 of the FIR gene.
  • FIG. 1 FIR splicing variant (FIR ⁇ exon2) lacking exon 2, which is a transcriptional repression site of c-myc gene, is specifically expressed in cancer tissue (colon) cancer tissue!
  • RT PCR results (2.5% agarose gel electrophoresis photograph) showing that.
  • FIR A exon2 is data from various experiments showing that, unlike FIR, it increases the expression of endogenous c-Myc.
  • FIG. 4 FIR A exon2 is data of various experiments showing that apoptosis cannot be induced.
  • FIG. 4 is an image diagram showing that the cause of carcinogenesis.
  • FIR intron 2 (2400 bp) has a CCCG (/ CCTG) repeat sequence of 9 times.
  • FIG. 7 shows the number of FCG intron 2 CCCG (/ CCT G) repeat sequences for which significant differences were observed between healthy subjects and cancer patients.
  • FIG. 8 shows the detailed structure of the repeating sequence of FIR intron 2 (CCCG / CCTG) in healthy subjects and cancer patients shown in FIG.
  • FIR FBP Interacting Repressor
  • the first to eighth “MATATIAL” in SEQ ID NO: 1 is exon 1, followed by exon 2 consisting of 29 amino acids “QVNGQQGGGSEPAAAAAWAAGDK WKPPQ” is missing in SEQ ID NO: 1.
  • the alanine continuous sequence “AA AAAWAA” in Exon 2 is a characteristic sequence for transcription repressor.
  • the protein of the present invention includes an amino acid sequence in which one or several amino acids are deleted, substituted, or added in the amino acid sequence shown in SEQ ID NO: 1, or the amino acid sequence shown in SEQ ID NO: 1. And a protein having an amino acid sequence of 95% or more, preferably 98% or more, and having substantially the same activity as the polypeptide having the amino acid sequence shown in SEQ ID NO: 1.
  • homology means each amino acid residue comprising the chain between two chains in a polypeptide sequence (or amino acid sequence) or polynucleotide sequence (or base sequence). This means the amount (number) of things that can be determined to be the same in the mutual relationship between each other or each base, and the degree of sequence correlation between two polypeptide sequences or two polynucleotide sequences. That means. Homology can be easily calculated.
  • BLASTP BLASTN ⁇ Forces
  • FASTA Altschul, SF et al., J. Molec. Biol, 215: 403 (1990) and the like can be used methods known in the art without being limited thereto.
  • substantially the same activity as the polypeptide having the amino acid sequence represented by SEQ ID NO: 1 refers to various protein activities such as physiological activity, biological activity, and physicochemical activity. It means that the activity is substantially equivalent or homogeneous. Specific examples of such activity include, for example, a polypeptide having the amino acid sequence shown in SEQ ID NO: 1, that is, the same antigenicity as a splicing variant lacking exon 2 of FIR (an antibody that reacts with the splicing variant). As well as functions such as increased expression of c-myc gene, inhibition of apoptosis induction, cell proliferation and canceration.
  • SEQ ID NO: 2 is shown as a specific example of the base sequence (coding region) of a cDNA encoding a splicing noreant lacking exon 2 of FIR of the present invention.
  • cctctctgcg tga An example of the base sequence encoding exon 2 (SEQ ID NO: 4: 87 bp) is as follows: AGTGGTGGCAGCGGGAGACAAATGGAAACCTCCACAG 0
  • a DNA encoding the protein (or polypeptide) of the present invention can be easily prepared by those skilled in the art based on the description in the present specification and known techniques in the technical field. For example, RT-PCR using mRNA prepared from cancer tissue force as a template, and other NASBA (Nucleic acid sequence base amplification) methods and TMA (Transcription-mediated) for appropriate cDNA libraries. Amplification), SDA (Strand Displacement Amplification) and ICAN (isothermal and chimeric primer-initiated amplification of nucleic acids) methods, etc. It is possible.
  • the human FIR gene containing the exon 2 part is registered under NCBI GenBank as registration number NM_014281. Therefore, the base sequence of a primer used for RT-PCR or the like can be appropriately designed and selected based on the registered base sequence and the information disclosed in Patent Document 1 or Patent Document 2.
  • the above DNA can be obtained by known methods (for example, Carruthers (1982) Cold Spring Harbor Symp. Quant. Biol. 47: 411—418; Adams (1983) J. Am. Chem. Soc. 105: 661 ; Belousov (19 97) Nucleic Acid Res. 25: 3440-3444; Frenkel (1995) Free Radic. Biol. Med. 19: 373—380; Blommers (1994) Biochemistry 33: 7886—7896; Narang (1979) Meth. Enzymol. 68: 90; Brown (1979) Meth. Enzymol. 68: 109; Beaucage (1981) Tetra. Lett. 22: 1859; US Pat. No. 4,458,066).
  • stringent conditions are conditions that enable selective and detectable specific binding between the polynucleotide or oligonucleotide and genomic DNA.
  • Stringent conditions are defined by a suitable combination of salt concentration, organic solvent (eg, formamide), temperature, and other known conditions. That is, stringency increases by decreasing salt concentration, increasing organic solvent concentration, or increasing hybridization temperature.
  • the washing conditions after the noisyization also affect the stringency. This wash condition is also defined by salt concentration and temperature, and the stringency of the wash increases with decreasing salt concentration and increasing temperature.
  • the “stringent condition” means the degree of homology between each base sequence, for example, about 80% or more, preferably about 90% or more, more preferably about 95% or more on average in the whole. This means that the hybrid is specifically formed only between the base sequences having high homology.
  • the conditions include a sodium concentration of 150 to 900 mM, preferably 600 to 900 mM, and a pH of 6 to 8 at a temperature of 60 ° C to 68 ° C.
  • Specific examples of stringent conditions include 5 X SSC (750 mM NaCl, 75 mM trisodium citrate), 1% SDS, 5 X Denhardt's solution 50% formaldehyde, and 42 ° C. The sample is washed and washed under the conditions of 0.1 X SSC (15 mM NaCl, 1.5 mM trisodium citrate), 0.1% SDS, and 55 ° C.
  • Hybridization is described, for example, in Current Protocol in Molecular Biology (.edited by Frederick M. Ausubel et al, 1987). It can be carried out according to a method known in the art such as the method described, or a method analogous thereto, and when a commercially available library is used, it can be carried out according to the method described in the attached instruction manual.
  • the thus obtained DNA encoding the protein of the present invention is inserted into an appropriate recombination DNA such as a plasmid vector, a fuzzy vector, and various hybrid vectors, and the expression vector thus obtained is obtained.
  • an appropriate recombination DNA such as a plasmid vector, a fuzzy vector, and various hybrid vectors
  • the protein of the present invention can be produced by transforming various cells using, and culturing the transformant under appropriate culture conditions. This recombination DNA should be handled using conventional recombinant DNA techniques known to those skilled in the art. Can be any vector.
  • host cells transformed with a vector containing a DNA encoding a protein are not particularly limited, but human-derived cells such as HeLa, mammalian cells such as COS7 and CHO cells, insect cells, and yeasts.
  • human-derived cells such as HeLa
  • mammalian cells such as COS7 and CHO cells
  • insect cells such as E. coli and Bacillus subtilis
  • prokaryotic cells containing various bacteria such as E. coli and Bacillus subtilis can be used.
  • the protein of the present invention can be produced using an appropriate in vitro translation system known to those skilled in the art, such as a rabbit reticulocyte lysate or a wheat germ extract.
  • the protein of the present invention is produced as a fusion protein with any tag known to those skilled in the art such as HA tag, His tag, FLAG tag, and myc tag for convenience of purification operation and detection. It is also possible to make it live. Furthermore, for convenience of protein measurement and detection, it can also be produced as a fusion protein with enzymes such as GST and various fluorescent proteins.
  • the above expression vectors include various promoters such as constitutive expression promoters or various inducible expression promoters known to those skilled in the art, depending on the expression system used, host cells, and the like. And various regulatory elements such as silencers, ribosome binding sites, signal sequences, replication origins, cloning sites, translation initiation sequences, partial positions of terminators, and poly A addition sites, as well as other foreign or endogenous proteins. It can appropriately contain genes to be encoded, various drug resistance genes, genes that complement auxotrophy, and the like.
  • DNA encoding the protein of the present invention can be appropriately selected depending on the host cell into which these vectors are to be introduced.
  • vectors known to those skilled in the art such as pCGNM2 vector plasmid (6), pKAl, pCDM8, and pSVK3 can be mentioned.
  • Such a vector can be introduced into a host cell to transiently express the protein of the present invention, or the entire or part of the host cell genome can be located in one or more places in the genome. Can be threaded.
  • various commercially available vectors known to those skilled in the art can be used.
  • the transformant of the present invention can be obtained by using an appropriate DNA fragment itself containing a gene encoding the protein of the present invention obtained by PCR amplification or the like instead of the expression vector. Both are possible.
  • the composition can be used for transformation as a composition such as a solution optionally containing an appropriate buffer and other auxiliaries in addition to a strong DNA fragment.
  • the expression vector (DNA for recombination) containing the DNA of the present invention or the DNA fragment itself such as, for example, the method, elctroporation, Lipofectamine Plus reagents (Gibco BRL input [repulsive norme method, protoplast-PEG method, It can be introduced into a host cell by any known method such as Ti plasmid method, particle gun method, nanovirus method, etc. Further, a transformant can be prepared. It is also possible by the Yong method.
  • a transformant that expresses the protein of the present invention can be produced by culturing the protein under conditions favorable for production of the protein, expressing the protein, and recovering the host cell and Z or medium power.
  • the medium used for host cell culture is appropriately selected from any medium known to those skilled in the art, depending on the configuration of the expression vector used (type of promoter, etc.) and the type of host. be able to.
  • the protein of the present invention produced by a host cell is used in an appropriate combination of any means known to those skilled in the art, for example, separation of a medium and cells by centrifugation or filtration, and an ammonium sulfate-like protein. It can be recovered from the medium by precipitation of the protein components of the medium with the appropriate salt and subsequent use of hydrophobic chromatography, ion exchange chromatography, affinity chromatography, or other chromatography. Alternatively, the protein of the present invention can be produced by a chemical synthesis method.
  • the DNA (oligonucleotide) containing a base sequence showing a gene polymorphism in the 4-base repeat sequence in intron 2 of the FIR gene of the present invention is, for example, 1 to 2100 (for example, about 2058), preferably 1 to: L It consists of a continuous base sequence of 100 (for example, about 1032), more preferably 1 to 600, and still more preferably 40 to 600 (for example, about 513).
  • SEQ ID NO: 3 genomic DNA containing exon 2 and intron 2 of the FIR gene shown below can be mentioned.
  • SEQ ID NO: 3 the first sequence shown in capital letters is exon 2 (87 bp), and the sequence shown in lower case following is the sequence of intron 2 (2399 bp). Furthermore, it is a sequence showing a gene polymorphism in a partial force base repeat (9 times) sequence surrounded by parentheses.
  • FIR is expressed not only in cancer tissues but also in normal tissues, whereas in some cases, splicing variants lacking exon 2 of FIR are observed.
  • the protein of the present invention is expressed only in cancer tissue cells of cancer patients and not expressed in normal tissue cells or blood cells adjacent to the cancer tissue. Therefore, such a protein and its encoding cDNA or corresponding mRNA, or an oligonucleotide that partially relies on them, serve as a very specific cancer detection marker in cancer detection and diagnosis. It is a substance that has utility.
  • the proportion of 9 homo repeats (including hetero in the base sequence type of repeat units) in the 4-base repeat sequence in intron 2 of the FIR gene is compared with that in normal tissues. Therefore, the above DNA containing a nucleotide sequence showing such a gene polymorphism is also a useful substance as a cancer detection marker in cancer detection and diagnosis.
  • the splicing variant protein or its fragment peptide lacking exon 2 of the above FIR, the cDNA encoding them or the mRNA corresponding thereto, or a partial oligonucleotide thereof, or in the intron 2 of the FIR gene Cancer can be detected or diagnosed significantly by measuring the presence of DNA (genomic DNA) consisting of a continuous base sequence containing a base sequence indicating a gene polymorphism in a 4-base repeat sequence.
  • DNA genomic DNA
  • cancer is detected by measuring the expression level of the above-mentioned splicing variant protein, mRNA (cDNA) or oligonucleotide in cancer thread and tissue, and using this as a normal tissue or normal person derived from the same cancer patient. This can be done by measuring the expression level in the tissues and comparing these values. Such measurement of the expression level can be quantitative, semi-quantitative, or qualitative depending on the principle of the measurement method.
  • the splicing variant protein lacking exon 2 of the FIR of the present invention or the mRNA (cDNA) encoding the same is expressed specifically only in the cancer thread and tissue, so that cancer can be conveniently and highly sensitive (highly sensitive). (Specificity) can be detected.
  • the expression level of the splicing variant protein can be measured by any method known to those skilled in the art. Examples include Western plotting using appropriate antibodies, immunostaining and methods using various immunological specific reactions such as EIA, amino acid sequence analysis of gas phase sequencer etc. peptides using Edman's method, Furthermore, it can be detected by mass spectrometry represented by MALDI-TOF / MS and ESI Q-TOFZMS methods.
  • a test method is preferred in which the expression level of the white matter is measured by an antigen antibody reaction with an antibody specific for FIR or a splicing variant thereof.
  • antibodies include, for example, antibodies that recognize both FIR and a splicing variant lacking exon 2 of FIR as antigens.
  • the above-mentioned antibody is a splicing variant that lacks FIR or exon 2 of FIR.
  • a suitable partial polypeptide (peptide fragment) thereof or various derivatives or complexes thereof as antigenic substances or immunogens can be prepared by an appropriate method known to those skilled in the art.
  • a polyclonal antibody it can be administered to an appropriate animal such as mouse, rat, rabbit, goat, or chicken, and its antiserum can be prepared.
  • known methods described in monoclonal antibody production methods (“monoclonal antibodies”, Nagamune Kamei, Hiroshi Terada, Yodogawa Shoten, 1990; “Monoclonal Antibody” James W. Goding, third edition, Academic Press, 1996) It is also possible to prepare a monoclonal antibody by a method using cell fusion. For example, it can be prepared by the method described in Patent Document 1 or Patent Document 2.
  • Such an antibody is derived from a complete antibody such as Fab, F (ab '), Fv fragment, etc. by genetic engineering (DNA recombination technology) as long as the original antibody activity is not lost.
  • Such antibodies and the like include those labeled with various labeling substances known to those skilled in the art, such as enzymes, radioisotope fluorescent dyes, and metal atoms.
  • an antigen-antibody reaction with an anti-FIR antibody for example, when measured by Western plotting, a splicing variant protein (513 amino acids) lacking FIR exon 2 and FIR (542 Amino acid) can be specifically detected by the difference in molecular weight.
  • an antibody that specifically reacts only with a splicing variant protein lacking exon 2 of FIR for example, it can be measured by an enzyme immunoassay such as EIA.
  • Nucleic acid molecules containing DNA consisting of a contiguous base sequence that includes a base sequence that indicates a gene polymorphism in the sequence include various PCR methods such as RT-PCR and real-time quantitative PCR, and various microarray (DNA chip) methods. Can be amplified by methods known to those skilled in the art.
  • nucleic acid molecule containing each of the above DNAs is amplified by an RT-PCR method and electrophoresed
  • the nucleic acid molecule can be detected and identified from the amount of the molecules measured in combination.
  • the base sequence of mRNA corresponding to the splicing variant lacking FIR etason 2 (for example, FIR exon 1 and exon 3)
  • FIR etason 2 for example, FIR exon 1 and exon 3
  • a primer that specifically amplifies a nucleic acid molecule containing only a contiguous nucleotide sequence encoding an oligonucleotide or a partial oligonucleotide thereof. It becomes possible. It is also possible to use a method (sequence method) for directly determining the base sequence of the amplified DNA.
  • the base sequence of the primer used in the above RT-PCR and the like is based on the base sequence of FIR registered in GenBank as described above and the information disclosed in Patent Document 1 or Patent Document 2. You can choose 'design as appropriate'.
  • the primer it is preferable to have a base number that allows specific binding to the cage, for example, 15-40 bases, more specifically, about 15-25 bases.
  • commercially available primer design software such as 01igo TM (National Bioscience Inc.) can be used.
  • the measurement kit used in the detection method of the present invention can have an appropriate configuration depending on the measurement object or measurement principle.
  • the kit includes, for example, an anti-FIR antibody, an antibody against a splicing noriant protein lacking exon 2 of FIR, various secondary antibodies (labeled antibodies), and the above-mentioned mRNA (cDNA) amplification.
  • Probes for hybridization used in primers, DNA chips, etc. can be included.
  • the above kit contains other elements or components known to those skilled in the art, for example, various reagents, enzymes, buffers, reaction plates (containers), etc., depending on the configuration-purpose of use.
  • Cancer patients and non-cancerous tissues were collected from patients admitted to Chiba University Hospital for the treatment of primary colorectal cancer with written consent prior to surgery and stored at minus 80 ° C.
  • Various human cancer cell lines were obtained from the Cellellology Collection (http://www.atcc.org/3 ⁇ 4earcn and atalogs / ellBiology.c & i). Volunteers for healthy volunteers were collected with the consent of the healthy volunteers working at the Chiba University Medical Research Institute and the Chiba University Hospital.
  • FIR full-length FIR
  • cDNA obtained from cancer tissue and splicing variant cDNA (HA-FIR ⁇ exon2) lacking exon 2 of FIR
  • pCGNM2 vector plasmid 9
  • HA hemagglutinin
  • HA-FIR and HA-FIR A exon2 with a tag attached to its N-terminus were expressed.
  • the human c-Myc expression vector was a commercially available pcDNA3.1-c-myc, ueneStorm Expression—Ready Clones (Invitrogen, o., AL).
  • HeLa cells were cultured on a cover glass, and the above-mentioned vector (150 ftnol) containing HA-FIR and HA-FIR ⁇ exon2 gene was introduced using Lipofectamine Plus reagents (Gibco BRL) to examine the change in expression of c-Myc.
  • the primary antibodies used were mouse monoclonal anti-HA antibody (Santa Cruz Biotechnology, CA and rabbit polyclonal anti-c-My c3 ⁇ 4ri (Upstate Biotechnology, NY), diluted 500-fold and 1,000-fold, respectively.
  • Potato ⁇ cis 7 detected by iiTUNEL assay (Apoptosis Detection system, Fluorescein n Promega, WI, USA). Two-color analysis was performed in 50 ⁇ ⁇ terminal deoxynucleotidyl transferase (TdT) buffer containing FITC-labeled dUTP (MEBSTAIN Ap optosis Kit: Medical & Biological Laboratories, JAPAN) G Finally 250 ⁇ g of DNase -Cells were mixed with 0.5 ml propidium iodide (PI) solution (freshly diluted to 5 ⁇ g / ml in PBS) containing fr ee RNase A.
  • PI propidium iodide
  • lysis buffer (7M urea, 2M thiourea, 2% 3-[(3-Chola midopropyl) dimethylammonio—] 1—propanesulfate (CHAPS), 0.1 M Dithiothreitol (D TT), 2% IPG buffer (Amersham Phrmacia Biothech, Backinghampsnire, UK), 40 mM Tris) Dissolve 500 mg of frozen yarn and weave using Polytron homogenizer (Kinematica, Switzerland) and centrifuge at 100,000 xg for 1 hour at 4 ° C. The supernatant was used.
  • the primary antibodies used for Western blotting are the rabbit anti-FIR antibody (5) and the goat polyclonal anti-13-actin antibody (Santa Cruz, Santa Cruz, CA), which are used at 1,000 and 500 times, respectively.
  • 7 Secondary antibody is 3,000 times diluted with horseradish IgG horseradish peroxidase conjugate (HRP) (Jackson, west Grove, PA), and 500 fold diluted with rabbit horseradish IgG HRP (Cappel, West Chester, PA). used.
  • Antigen was detected with ECL TM detection reagents (Amersham Pharmacia Biothech) and expression levels were quantified by NIH Image.
  • Polyclonal anti-FIR antibody has two synthetic peptides (C (correction) DKWKPPQGTDSIKME (a Minoic acid 30-45) and C (additional caro) EVYDQERFDNSDLSA (amino acids 528-542)) were immunized simultaneously.
  • RNA and genomic DNA were extracted from cancerous and non-cancerous parts using RNeasy TM Mini Kit and DNeasy TM Tis sues Kit (Qiagen).
  • cDNA was synthesized from total RNA using the 1 st strand cDNA Synthesis Kit (Roche, Mannheim, Germany).
  • FIR cDNA was amplified by RT-PCR using the prepared cDNA.
  • the primers used were: forward 5,-GGCCCCATC AAGAGCATC -3 '(SEQ ID NO: 5), reverse 5,-GGGGCTGGGCCAGGGTCAG -3' (SEQ ID NO: 6).
  • GAPDH cDNA was used as a control.
  • FIR cDNA amplified by ReaH ime quantitative PCR was accurately quantified with LightCycler TM (Roche, Mannheim, Germany). We asked Gene research laboratories Inc (Sendai, Japan) to evaluate the optimal primer conditions. PIR for amplification of FIR cDNA ⁇ [MA (PCR product size is 275 base pairs): forward 5, — G and ACCTGGAGTC ATCACA-3, (SEQ ID NO: 7), reverse 5, -CGCAGAACCATCACTGTAG-3 '(SEQ ID NO: 8) was used.
  • Primers for human c-myc cDNA and human j8-actin cDNA are: forward 5,-GCCTCAGAGTGCATCGAC-3, (SEQ ID NO: 9), reverse 5, -TCCACA GAAACAACATCG-3 '(SEQ ID NO: 10) (c-myc), forward 5 '-TGGAGAAAATCTGG CACCAC-3' (SEQ ID NO: 11), reverse 5, -AATGGTGATGACCTGGCCGT-3, (SEQ ID NO: 12) ( ⁇ -actin) were used.
  • the following primer set was used for detecting the repeated sequence of FIR intron 2.
  • R04 5 '-AAGCCCATTCCTCCTTA-3' (SEQ ID NO: 14)
  • R05 5 '-TTTGACTTGCGCATTGC-3' (SEQ ID NO: 15)
  • PCR was performed using the IF-R04 primer set, and 2nd PCR was performed using the 1F-R05 primer set.
  • This PCR product was directly subjected to DNA sequencing according to a conventional method. PCR conditions are 95 ° C; 5 min, [95 ° C; 30 sec-56 ° C; 30 sec-72 ° C; 1 min] x35 cycles, 72. C; performed at 2 min.
  • DNA polymerase is Pfo DNA polymerase (Stratagene, La Jolla, CA) used.
  • an IF primer was used for the DNA sequence.
  • intron 2 of the FIR gene were significantly different in the number of repeats between genomic DNA derived from peripheral blood mononuclear suddenly in healthy subjects and genomic DNA derived from colon cancer tissue (FIGS. 7 and 8). That is, the proportion of cancer patients with 9 repeat sequences (homo) was significantly higher in cancer patients than in healthy individuals.
  • cancer detection can be diagnosed by examining the number of repetitive sequences present in intron 2 of the FIR gene.
  • the present invention provides a more effective and reliable detection 'diagnosis' treatment of cancer, particularly digestive organ cancer represented by colorectal cancer, as well as development of cancer gene diagnosis system and gene therapy clinical research system.
  • Various biomolecules used in the field are provided.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Oncology (AREA)
  • Wood Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Urology & Nephrology (AREA)
  • Physics & Mathematics (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

[PROBLEMS] To provide a method of more effectively and surely detecting, diagnosing and/or treating cancer, in particular, digestive system cancer typified by colon cancer; and various biomolecules to be used in developing such gene diagnosis systems for cancer as described above and clinical research systems of gene therapy. [MEANS FOR SOLVING PROBLEMS] A splicing variant of c-myc gene transcriptional regulator FIR, a cDNA encoding the same or an mRNA corresponding to the same; a DNA containing a four-base repetitive sequence in intron 2 of FIR gene; a method of detecting cancer by detecting the occurrence thereof; and an assay kit to be used therefor.

Description

明 細 書  Specification
c-myc遺伝子転写抑制因子 FIRのスプライシングバリアント又はイントロン 2内の 4塩基繰り返し配列による癌検出方法  Cancer detection method by splicing variant of c-myc gene transcription repressor FIR or 4-base repeat sequence in intron 2
技術分野  Technical field
[0001] 本発明は、 c-myc遺伝子転写抑制因子 FIRのスプライシングバリアント若しくはそれ をコードする cDNA又はそれに対応する mRNA、 FIR遺伝子のイントロン 2内の 4塩 基繰り返し配列を含む DNA、それらの存在を検出する癌検出方法、それに使用す る測定キット等に関する。  [0001] The present invention relates to a splicing variant of a c-myc gene transcription repressor FIR or a cDNA encoding the same or a mRNA corresponding thereto, a DNA containing a 4-base repeat sequence in intron 2 of the FIR gene, and the presence thereof The present invention relates to a cancer detection method to be detected and a measurement kit to be used for the method.
背景技術  Background art
[0002] c-Myc蛋白は細胞の生命活動に極めて重要であり、細胞の増殖、分化、細胞死(ァ ポトーシス)を制御する転写因子であり、それ自身、細胞周期や分化に伴い、多くの 転写因子によって厳密に制御されている。一方細胞内、外からの刺激に素早く対応 し恒常性を維持するために、 c-myc遺伝子には蛋白合成を介しない、転写により発生 する 2重鎖 DNAの物理的 ·力学的変化を感知する未知のメカニズムが存在し、それ によって転写レベルが一定に維持されていると考えられる。これまでに、 c-myc遺伝 子の上流のどの部位力 ¾-myc遺伝子の転写に影響を与える力調べられ、 c-myc遺伝 子の転写開始部位の 1.5kb上流のわずか百数十 baseの部位が c-myc遺伝子の転写 に極めて重要であることが示され FUSE (Far Upstream Element)と名付けられた(1)。 即ち転写の活性ィヒによって発生する 2重鎖 DNAの物理的ひずみがプロモーターの 上流に negative supercoilingを生じさせ、これにより 2重鎖 DNAが 1本鎖 DNAにほど ける部位があり、同部位が c-myc遺伝子の転写制御に極めて重要であることが報告さ れた(1-7)。この FUSEに結合する蛋白質を oligonucleotide affinity chromatographyに よって同定したところ、 70kDaの分子量を有する FBP (FUSE結合蛋白; FUSE Binding Protein)であった。この FBP蛋白は強力な転写活性を有しており (2_4)、 FIRはこの FB Pに結合する蛋白質として Yeast Two-Hybrid Systemにより同定された (5)。 FIR (FBP Interacting Repressor)は TFIIHの helicase活性を抑制することにより c- myc遺伝子の 転写を抑制すると考えられて 、る (6)。 [0003] このような c-Myc蛋白は細胞内で増加しても減少してもアポトーシスを誘導する力 そ の詳細なメカニズムや癌化との関わりついては不明の点が多い。これまでに、本発明 者自身の発明に関する国際特許出願 PCT/JP2003/5046 (特許文献 1)及び PCT/JP 2003/10676 (特許文献 2)において、抗 FIR抗体を用いたウェスタンブロット及び RT — PCRにより癌組織での FIRの発現が増加していることが記載されている。 [0002] c-Myc protein is extremely important for cell life activity and is a transcription factor that controls cell proliferation, differentiation and cell death (apoptosis). It is strictly controlled by transcription factors. On the other hand, the c-myc gene senses physical and mechanical changes in double-stranded DNA generated by transcription, not through protein synthesis, in order to respond quickly to intracellular and external stimuli and maintain homeostasis. There is an unknown mechanism, which is thought to keep the transcription level constant. So far, any site force upstream of the c-myc gene ¾-myc gene has been examined for its influence on transcription, and only a few tens of bases 1.5 kb upstream of the transcription start site of the c-myc gene. Was shown to be very important for c-myc gene transcription and named FUSE (Far Upstream Element) (1). That is, the physical strain of double-stranded DNA generated by transcriptional activity causes negative supercoiling upstream of the promoter, which causes the double-stranded DNA to unfold into single-stranded DNA, which is c -It was reported to be extremely important for the transcriptional control of the myc gene (1-7). The protein that binds to FUSE was identified by oligonucleotide affinity chromatography and found to be FBP (FUSE Binding Protein) having a molecular weight of 70 kDa. This FBP protein has a strong transcriptional activity (2_4), and FIR was identified by the Yeast Two-Hybrid System as a protein that binds to this FBP (5). FIR (FBP Interacting Repressor) is thought to suppress c-myc gene transcription by suppressing the helicase activity of TFIIH (6). [0003] The ability of such c-Myc protein to induce apoptosis regardless of whether it increases or decreases in a cell has many unclear points regarding its detailed mechanism and its relation to canceration. So far, in the international patent applications PCT / JP2003 / 5046 (Patent Document 1) and PCT / JP 2003/10676 (Patent Document 2) relating to the inventors' own invention, Western blot and RT-PCR using anti-FIR antibodies have been carried out. Describes that FIR expression in cancer tissues is increased.
[0004] 更に、 PCT/JP2003/10676 (特許文献 2)には、大腸癌 3例からの FIR配列決定により FIRアミノ酸配列の第 1〜135番目における複数の塩基又はアミノ酸の置換が検出さ れたこと、 FIRを HeLa細胞に発現誘導するとアポトーシスが誘導される一方、 FIRの転 写活性部位である N末端 77個のアミノ酸を削除した変異 FIR蛋白ではアポトーシスが 誘導されず、 c-Myc発現プラスミドを共発現させると FIRによるアポトーシス誘導は阻 害されたこと、更には、結腸直腸癌において FIRタンパク及び mRN Aの発現が増加 して 、ること等が記載されて 、る。  [0004] Furthermore, in PCT / JP2003 / 10676 (Patent Document 2), substitution of a plurality of bases or amino acids at positions 1 to 135 of the FIR amino acid sequence was detected by FIR sequencing from 3 cases of colorectal cancer. When FIR is induced to induce expression in HeLa cells, apoptosis is induced. On the other hand, a mutant FIR protein that deletes the N-terminal 77 amino acids of the FIR transcriptional activity does not induce apoptosis. It is described that, when co-expressed, apoptosis induction by FIR was inhibited, and furthermore, the expression of FIR protein and mRNA was increased in colorectal cancer.
特許文献 1:国際公開第 WO2004Z018518号パンフレット  Patent Document 1: International Publication No. WO2004Z018518 Pamphlet
特許文献 2:国際公開第 WO2004Z018679号パンフレット  Patent Document 2: International Publication No. WO2004Z018679 Pamphlet
発明の開示  Disclosure of the invention
発明が解決しょうとする課題  Problems to be solved by the invention
[0005] 本発明の目的は、癌、特に大腸癌に代表される消化器癌等のより有効で確実な検出 •診断 '治療を提供すること、及び、このような癌の遺伝子診断システムと遺伝子治療 臨床研究システムの開発に利用される各種の生体分子を提供すること等を目的とす る。 [0005] An object of the present invention is to provide more effective and reliable detection / diagnosis of cancer, particularly digestive organ cancer represented by colorectal cancer, and to provide a genetic diagnosis system and gene for such cancer. Treatment The purpose is to provide various biomolecules used in the development of clinical research systems.
課題を解決するための手段  Means for solving the problem
[0006] 本発明者は上記課題を解決すベぐ研究した結果、 c-Mycの発現増大が認められる 消ィ匕器 (大腸)癌組織中では転写抑制部位を含むェクソン 2を欠損した FIR^プライシ ングバリアント (FIR Δ exon2)が癌特異的に発現増大して 、ることを新たに見出し、更 に、 FIR遺伝子のスプライシング発現メカニズムを調べる過程で、 FIRのイントロン 2に 4塩基の繰り返し配列(Microsatellite Instability (MIN)/Simple Sequence Repeat (SSR ))が存在することを発見した。本発明はこれらの新たな知見に基づき完成されたもの である。 即ち、本発明は、以下の各態様に係るものである。 [0006] As a result of research that solves the above problems, the present inventor has found that c-Myc expression is increased. In the cancer tissue of colon (colon), FIR ^ lacking exon 2 containing a transcriptional repression site. In the process of investigating the mechanism of splicing expression of FIR gene, it was newly found that the expression of the pricing variant (FIR Δ exon2) was increased specifically in cancer. I discovered that Microsatellite Instability (MIN) / Simple Sequence Repeat (SSR) exist. The present invention has been completed based on these new findings. That is, the present invention relates to the following aspects.
[態様 l]c myc遺伝子の転写抑制因子である FIRのェクソン 2が欠失したスプライ シングバリアントである蛋白質。  [Aspect l] A protein which is a splicing variant in which exon 2 of FIR which is a transcription repressor of c myc gene is deleted.
[態様 2]以下のアミノ酸配列を有する蛋白質:  [Aspect 2] Protein having the following amino acid sequence:
(1)配列番号 1に示されたアミノ酸配列から成る蛋白質、又は  (1) a protein comprising the amino acid sequence shown in SEQ ID NO: 1, or
(2)配列番号 1に示されるアミノ酸配列において、 1個若しく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、又は、配列番号 1に示されるアミノ酸配列と 相同性が 95%以上のアミノ酸配列からなり、且つ、配列番号 1に示されるアミノ酸配 列を有するポリペプチドと実質的に同等の活性を有する蛋白質。  (2) In the amino acid sequence shown in SEQ ID NO: 1, the amino acid sequence in which one or several amino acids are deleted, substituted or added, or homology with the amino acid sequence shown in SEQ ID NO: 1 is 95% or more And a protein having substantially the same activity as a polypeptide having the amino acid sequence shown in SEQ ID NO: 1.
[態様 3]態様 1又は 2記載の蛋白質をコードする cDNA又はそれに対応する mRNA  [Aspect 3] cDNA encoding the protein according to aspect 1 or 2, or mRNA corresponding thereto
[態様 4]以下の塩基配列から成る cDNA又はそれに対応する mRNA: [Aspect 4] cDNA consisting of the following nucleotide sequences or mRNA corresponding thereto:
(1)配列番号 2に示された塩基配列、又は  (1) the base sequence shown in SEQ ID NO: 2, or
(2)配列番号 2に示される塩基配列と相補的な塩基配列力 なる DNAとストリンジ ントな条件下でハイブリダィズし、且つ、態様 1又は 2記載の蛋白質をコードする塩基 配列。  (2) A base sequence that hybridizes under stringent conditions with DNA having a base sequence ability complementary to the base sequence shown in SEQ ID NO: 2 and encodes the protein according to aspect 1 or 2.
[態様 5]FIR遺伝子のイントロン 2における 4塩基繰り返し配列における遺伝子多型 を示す塩基配列を含む連続した塩基配列から成る DNA。  [Aspect 5] DNA comprising a continuous base sequence including a base sequence indicating a gene polymorphism in a 4-base repeat sequence in intron 2 of the FIR gene.
[態様 6]配列番号 3 (FIR遺伝子のェクソン 2及びイントロン 2を含むゲノム DNA:合 計 2487bp)における 118番目〜153番目の塩基配列に見られる 4塩基の 9回繰り返 し配列、又は、その領域における該 4塩基の 5回若しくは 6回繰り返し配列を含む、態 様 5記載の DNA。  [Aspect 6] 4 bases 9 times repeated sequence found in the base sequence from position 118 to position 153 in SEQ ID NO: 3 (genomic DNA containing exon 2 and intron 2 of FIR gene: total 2487 bp), or its The DNA according to aspect 5, comprising 5 or 6 repeat sequences of the 4 bases in the region.
[態様 7]4塩基繰り返しの塩基配列が CCCG又は CCTGである、態様 6記載の DNA  [Aspect 7] A DNA according to Aspect 6, wherein the base sequence of the 4-base repeat is CCCG or CCTG
[態様 8]態様 1若しくは 2記載の蛋白質又はその断片ペプチド、態様 3又は 4記載の cDNA若しくはそれに対応する mRNA、又はそれらの一部からなるオリゴヌクレオチ ド、又は、態様 5〜7のいずれか一項記載の 4塩基繰り返し配列を含む連続した塩基 配列から成る DNAの存在を測定することから成る、癌の検出方法。 [態様 9]FIR又はそのスプライシングバリアントに特異的な抗体との抗原抗体反応を 利用する、態様 8記載の検出方法。 [Aspect 8] The protein according to Aspect 1 or 2, or a fragment peptide thereof, the cDNA according to Aspect 3 or 4, or the mRNA corresponding thereto, or an oligonucleotide comprising a part thereof, or any one of Aspects 5-7 A method for detecting cancer, comprising measuring the presence of DNA comprising a continuous base sequence comprising the 4-base repeat sequence described in the section. [Aspect 9] The detection method according to Aspect 8, using an antigen-antibody reaction with an antibody specific for FIR or a splicing variant thereof.
[態様 10]ウェスタンプロット法により測定することを特徴とする、態様 9記載の検出方 法。  [Aspect 10] The detection method according to Aspect 9, characterized by measurement by Western plotting.
[態様 11 ]態様 3又は 4記載の cDNA若しくはそれに対応する mRNA、又はそれらの 一部からなるオリゴヌクレオチドを含む核酸分子を増幅する為のプライマーセットを使 用する PCRを利用する、態様 8記載の検出方法。  [Aspect 11] Aspect 8 according to aspect 8, wherein PCR using a primer set for amplifying a nucleic acid molecule comprising the cDNA according to aspect 3 or 4 or the mRNA corresponding thereto or an oligonucleotide comprising a part thereof is used. Detection method.
[態様 12]RT— PCRである態様 11記載の検出方法。  [Aspect 12] The detection method according to Aspect 11, which is RT—PCR.
[態様 13]リアルタイム定量的 PCRである態様 11記載の検出方法。  [Aspect 13] The detection method according to Aspect 11, which is real-time quantitative PCR.
[態様 14]態様 5〜7の 、ずれか一項記載の DNAを含む核酸分子を増幅する為の プライマーセットを使用する PCRを利用する、態様 8記載の検出方法。  [Aspect 14] A detection method according to Aspect 8, which utilizes PCR using a primer set for amplifying a nucleic acid molecule comprising the DNA according to any one of Aspects 5-7.
[態様 15]癌が消ィヒ器癌である、態様 8〜14のいずれか一項記載の検出方法。  [Aspect 15] The detection method according to any one of Aspects 8 to 14, wherein the cancer is a cancer of the extinct organ.
[態様 16]癌が大腸癌である、態様 8〜14のいずれか一項記載の検出方法。  [Aspect 16] The detection method according to any one of Aspects 8 to 14, wherein the cancer is colorectal cancer.
[態様 17]態様 8〜14のいずれか一項に記載の検出方法に使用する測定キット。  [Aspect 17] A measurement kit used in the detection method according to any one of Aspects 8 to 14.
[態様 18]抗体 FIR又はそのスプライシングバリアントに特異的な抗体、又は、態様 3 [Aspect 18] Antibody specific to antibody FIR or a splicing variant thereof, or Aspect 3
〜7のいずれか一項記載の核酸分子を増幅する為のプライマーセットを要素として含 む、態様 17記載の測定キット。 The measurement kit according to embodiment 17, comprising a primer set for amplifying the nucleic acid molecule according to any one of -7 as an element.
発明の効果  The invention's effect
[0008] 本発明の c myc遺伝子の転写抑制因子である FIRのェクソン 2が欠失したスプラ イシングバリアント蛋白質は癌糸且織にのみに特異的に発現しており、外蛋白質及び それをコードする mRNA (cDNA)レベルで有意に検出することが出来る。更に、 FI R遺伝子のイントロン 2における 4塩基繰り返し配列における遺伝子多型に基づき、癌 の検出'診断を行うことが出来る。 [0008] The splicing variant protein lacking FIR exon 2 which is a transcriptional repressor of the c myc gene of the present invention is specifically expressed only in cancer thread and tissue, and encodes an external protein and the protein. m RNA (cDNA) can be significantly detected at level. Furthermore, cancer detection 'diagnosis can be performed based on the gene polymorphism in the 4-base repeat sequence in intron 2 of the FIR gene.
図面の簡単な説明  Brief Description of Drawings
[0009] [図 1]消ィ匕器 (大腸)癌組織中では c-myc遺伝子転写抑制部位であるェクソン 2の欠 損した FIRスプライシングバリアント(FIR Δ exon2)が特異的に高発現して!/、たことを示 す RT— PCRの結果 (2. 5%ァガロースゲル電気泳動の写真)。  [0009] [Fig. 1] FIR splicing variant (FIR Δ exon2) lacking exon 2, which is a transcriptional repression site of c-myc gene, is specifically expressed in cancer tissue (colon) cancer tissue! RT—PCR results (2.5% agarose gel electrophoresis photograph) showing that.
[図 2]リアルタイム定量的 PCRで測定した消ィ匕器癌患者における FIR Δ exon2の発現 の様子を示す。 [Figure 2] Expression of FIR Δ exon2 in gastrointestinal cancer patients measured by real-time quantitative PCR The state of is shown.
[図 3]FIR A exon2は FIRと異なり内因性 c-Mycの発現を増大させることを示す各種実 験のデータである。 A:RT— PCR、 B :免疫組織ィ匕学染色、 C及び D :フローサイトメト リー。  [Figure 3] FIR A exon2 is data from various experiments showing that, unlike FIR, it increases the expression of endogenous c-Myc. A: RT—PCR, B: Immunohistochemical staining, C and D: Flow cytometry.
[図 4]FIR A exon2はアポトーシスを誘導することが出来ないことを示す各種実験のデ ータである。 A:免疫組織化学染色、 B :フローサイトメトリー。  [Fig. 4] FIR A exon2 is data of various experiments showing that apoptosis cannot be induced. A: Immunohistochemical staining, B: Flow cytometry.
[図 5]癌糸且織では FIR Δ exon2が特異的に発現することにより正常型 FIRによる c- myc 遺伝子転写抑制を競合阻害し、アポトーシス誘導阻害と c-Myc発現増大を同時に惹 起することが、発癌の原因になって ヽることを示すイメージ図である。  [Fig.5] In cancer thread and tissue, FIR Δ exon2 is specifically expressed to competitively inhibit c-myc gene transcription repression by normal FIR, and simultaneously induce apoptosis induction and increase c-Myc expression. FIG. 4 is an image diagram showing that the cause of carcinogenesis.
[図 6]FIRイントロン 2(2400bp)には CCCG(/CCTG)の 9回繰り返し配列が存在する。 FI [FIG. 6] FIR intron 2 (2400 bp) has a CCCG (/ CCTG) repeat sequence of 9 times. FI
Rイントロン 2の最初の部分(12-206)とイントロン l(4880bp)の最後部分 (4682-4880)に は、 CCCG(/CCTG)の 9回繰り返し配列を含み 121bp/195bp=62.1%の高い相同性が 認められた。 The first part of R intron 2 (12-206) and the last part of intron l (4880bp) (4682-4880) contain 9 repeats of CCCG (/ CCTG), 121bp / 195bp = 62.1% high homology Sex was recognized.
[図 7]健常者と癌患者との間で有意の差が認められた FIRイントロン 2の CCCG(/CCT G)の繰り返し配列の回数を示す。  FIG. 7 shows the number of FCG intron 2 CCCG (/ CCT G) repeat sequences for which significant differences were observed between healthy subjects and cancer patients.
[図 8]図 7に示した健常者及び癌患者における FIRイントロン 2の CCCG /CCTG)の繰 り返し配列の詳細な構造を示す。  FIG. 8 shows the detailed structure of the repeating sequence of FIR intron 2 (CCCG / CCTG) in healthy subjects and cancer patients shown in FIG.
発明を実施するための最良の形態  BEST MODE FOR CARRYING OUT THE INVENTION
[0010] [態様 1及び 2]  [0010] [Aspects 1 and 2]
c myc遺伝子の転写抑制因子である FIR (FBP Interacting Repressor)のスプライシ ングバリアントであって、ェクソン 2が欠失した蛋白質それ自体に関する。このような蛋 白質の具体例として、配列番号 1に示された 513個のアミノ酸配列力 成る蛋白質を 挙げることが出来る。  c A splicing variant of FIR (FBP Interacting Repressor), which is a transcriptional repressor of myc gene, and relates to the protein itself lacking exon 2. As a specific example of such a protein, there can be mentioned a protein having a sequence ability of 513 amino acids shown in SEQ ID NO: 1.
[0011] S列番号 1 :アミノ酸一文字表記] [0011] S column number 1: single letter amino acid]
[0012] 上記配列番号 1における 1〜8番目の「MATATIAL」がェクソン 1であり、その後に 続く 29個のアミノ酸からなるェクソン 2「QVNGQQGGGSEPAAAAAWAAGDK WKPPQ」が配列番号 1では欠損している。尚、ェクソン 2中のァラニン連続配列「AA AAAWAA」は転写抑制因子に特徴的配列である。 [0012] The first to eighth “MATATIAL” in SEQ ID NO: 1 is exon 1, followed by exon 2 consisting of 29 amino acids “QVNGQQGGGSEPAAAAAWAAGDK WKPPQ” is missing in SEQ ID NO: 1. In addition, the alanine continuous sequence “AA AAAWAA” in Exon 2 is a characteristic sequence for transcription repressor.
[0013] 本発明の蛋白質には、配列番号 1に示されるアミノ酸配列において、 1個若しくは 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、又は、配列番号 1に 示されるアミノ酸配列と相同性が 95%以上、好ましくは 98%以上のアミノ酸配列から なり、且つ、配列番号 1に示されるアミノ酸配列を有するポリペプチドと実質的に同等 の活性を有する蛋白質が含まれる。  [0013] The protein of the present invention includes an amino acid sequence in which one or several amino acids are deleted, substituted, or added in the amino acid sequence shown in SEQ ID NO: 1, or the amino acid sequence shown in SEQ ID NO: 1. And a protein having an amino acid sequence of 95% or more, preferably 98% or more, and having substantially the same activity as the polypeptide having the amino acid sequence shown in SEQ ID NO: 1.
[0014] ここで、「相同性」とは、ポリペプチド配列(あるいはアミノ酸配列)又はポリヌクレオチド 配列(あるいは塩基配列)における 2本の鎖の間で該鎖を構成して 、る各アミノ酸残 基同志又は各塩基同志の互いの適合関係において同一であると決定できるようなも のの量 (数)を意味し、二つのポリペプチド配列又は二つのポリヌクレオチド配列の間 の配列相関性の程度を意味するものである。相同性は容易に算出できる。二つのポ リヌクレオチド配列又はポリペプチド配列間の相同性を測定する方法は数多く知られ ており、「相同性」(「同一性」とも言われる)なる用語は、当業者には周知である(例え 【 、 Lesk, A. M. (Ed.), し omputational Molecular Biology, Oxford University Press, New York, (1988);Smith, D. W. (Ed.), Biocomputing: Informatics and Genome Projec ts, Academic Press, New York, (1993); Grifin, A. M. & Grifin, H. G. (Ed.), Compute r Analysis of Sequence Data: Part I, Human Press, New Jersey, (1994);von Heinje, G., Sequence Analysis in Molecular Biology, Academic Press, New York, (1987); Gri bskov, M. & Devereux, J. (Ed.), Sequence Analysis Primer, M- Stockton Press, New York, (1991)等)。二つの配列の相同性を測定するのに用いる一般的な方法には、 Martin, J. Bishop (Ed.), Guide to Huge Computers, Academic Press, San Diego, (19 94);Carillo, H. & Lipman, D., SIAM J. Applied Math., 48: 1073 (1988)等に開示され ているものが挙げられる力 これらに限定されるものではない。相同性を測定するた めの好まし!/、方法としては、試験する二つの配列間の最も大きな適合関係部分を得 るように設計したものが挙げられる。このような方法は、コンピュータープログラムとし て組み立てられているものが挙げられる。二つの配列間の相同性を測定するための 好ましいコンピュータープログラム法としては、 GCGプログラムパッケージ(Devereux, J. et al., Nucleic Acids Research, 12(1): 387 (1984))、 BLASTP、 BLASTNゝ FASTA ( Atschul, S. F. et al., J. Molec. Biol, 215: 403 (1990》等が挙げられる力 これらに 限定されるものでなぐ当該分野で公知の方法を使用することができる。 [0014] Here, "homology" means each amino acid residue comprising the chain between two chains in a polypeptide sequence (or amino acid sequence) or polynucleotide sequence (or base sequence). This means the amount (number) of things that can be determined to be the same in the mutual relationship between each other or each base, and the degree of sequence correlation between two polypeptide sequences or two polynucleotide sequences. That means. Homology can be easily calculated. Many methods for measuring homology between two polynucleotide or polypeptide sequences are known, and the term “homology” (also referred to as “identity”) is well known to those skilled in the art ( For example,, Lesk, AM (Ed.), Omputational Molecular Biology, Oxford University Press, New York, (1988); Smith, DW (Ed.), Biocomputing: Informatics and Genome Projects, Academic Press, New York, ( 1993); Grifin, AM & Grifin, HG (Ed.), Computer Analysis of Sequence Data: Part I, Human Press, New Jersey, (1994); von Heinje, G., Sequence Analysis in Molecular Biology, Academic Press, New York, (1987); Gri bskov, M. & Devereux, J. (Ed.), Sequence Analysis Primer, M-Stockton Press, New York, (1991), etc. ). Common methods used to measure the homology of two sequences include Martin, J. Bishop (Ed.), Guide to Huge Computers, Academic Press, San Diego, (19 94); Carillo, H. & Lipman, D., SIAM J. Applied Math., 48: 1073 (1988) and the like. The power is not limited to these. Preferred / methods for measuring homology are those designed to obtain the largest match between the two sequences tested. Such methods include those assembled as computer programs. Preferred computer programming methods for measuring homology between two sequences include the GCG program package (Devereux, J. et al., Nucleic Acids Research, 12 (1): 387 (1984)), BLASTP, BLASTN ゝForces such as FASTA (Atschul, SF et al., J. Molec. Biol, 215: 403 (1990) and the like can be used methods known in the art without being limited thereto.
[0015] 本明細書において、「配列番号 1に示されるアミノ酸配列を有するポリペプチドと実質 的に同等の活性」とは、蛋白質の生理的活性、生物学的活性及び物理化学的活性 等の各種活性が実質的に同等又は同質であることを意味する。このような活性の具 体例として、例えば、配列番号 1に示されるアミノ酸配列を有するポリペプチド、即ち、 FIRのェクソン 2が欠失したスプライシングバリアントと同一の抗原性 (該スプライシン グバリアントと反応する抗体と特異的に結合する)、並びに、 c-myc遺伝子の発現増 大、アポトーシス誘導阻害、細胞増殖及び癌化等の機能を挙げることが出来る。 In the present specification, “substantially the same activity as the polypeptide having the amino acid sequence represented by SEQ ID NO: 1” refers to various protein activities such as physiological activity, biological activity, and physicochemical activity. It means that the activity is substantially equivalent or homogeneous. Specific examples of such activity include, for example, a polypeptide having the amino acid sequence shown in SEQ ID NO: 1, that is, the same antigenicity as a splicing variant lacking exon 2 of FIR (an antibody that reacts with the splicing variant). As well as functions such as increased expression of c-myc gene, inhibition of apoptosis induction, cell proliferation and canceration.
[0016] [態様 3及び 4] [Aspects 3 and 4]
本発明の FIRのェクソン 2が欠失したスプライシングノ リアントをコードする cDNAの 塩基配列 (コード領域)の具体例として以下の配列番号 2を示す。  The following SEQ ID NO: 2 is shown as a specific example of the base sequence (coding region) of a cDNA encoding a splicing noreant lacking exon 2 of FIR of the present invention.
[0017] 己列番号 2 : 1542bp] atggcg acggcgacca tagctctc ggcac  [0017] self-row number 2: 1542bp] atggcg acggcgacca tagctctc ggcac
agactccatc aagatggaga acgggcagag cacagccgcc aagctggggc tgcctcccct gacgcccgag cagcaggagg cccttcagaa ggccaagaag tacgccatgg agcagagcat agactccatc aagatggaga acgggcagag cacagccgcc aagctggggc tgcctcccct gacgcccgag cagcaggagg cccttcagaa ggccaagaag tacgccatgg agcagagcat
caagagtgtg ctggtgaagc agaccatcgc gcaccagcag cagcagctca ccaacctgca caagagtgtg ctggtgaagc agaccatcgc gcaccagcag cagcagctca ccaacctgca
gatggcggct cagcggcagc gggcgctggc catcatgtgc cgcgtctacg tgggctctat gatggcggct cagcggcagc gggcgctggc catcatgtgc cgcgtctacg tgggctctat
ctactatgag ctgggggagg acaccatccg ccaggccttt gccccctttg gccccatcaa ctactatgag ctgggggagg acaccatccg ccaggccttt gccccctttg gccccatcaa
gagcatcgac atgtcctggg actccgtcac catgaagcac aagggctttg ccttcgtgga gagcatcgac atgtcctggg actccgtcac catgaagcac aagggctttg ccttcgtgga
gtatgaggtc cccgaagctg cacagctggc cttggagcag atgaactcgg tgatgctggg gtatgaggtc cccgaagctg cacagctggc cttggagcag atgaactcgg tgatgctggg
gggcaggaac atcaaggtgg gcagacccag caacataggg caggcccagc ccatcataga gggcaggaac atcaaggtgg gcagacccag caacataggg caggcccagc ccatcataga
ccagttggct gaggaggcac gggccttcaa ccgcatctac gtggcctctg tgcaccagga ccagttggct gaggaggcac gggccttcaa ccgcatctac gtggcctctg tgcaccagga
cctctcagac gatgacatca agagcgtgtt tgaggccttt ggcaagatca agtcctgcac cctctcagac gatgacatca agagcgtgtt tgaggccttt ggcaagatca agtcctgcac
actggcccgg gaccccacaa ctggcaagca caagggctac ggcttcattg agtacgagaa actggcccgg gaccccacaa ctggcaagca caagggctac ggcttcattg agtacgagaa
ggcccagtcg tcccaagatg ctgtgtcttc catgaacctc tttgacctgg gtggccagta ggcccagtcg tcccaagatg ctgtgtcttc catgaacctc tttgacctgg gtggccagta
cttgcgggtg ggcaaggctg tcacaccgcc catgccccta ctcacaccag ccacgcctgg cttgcgggtg ggcaaggctg tcacaccgcc catgccccta ctcacaccag ccacgcctgg
aggcctccca cctgccgctg ctgtggcagc tgctgcagcc actgccaaga tcacagctca aggcctccca cctgccgctg ctgtggcagc tgctgcagcc actgccaaga tcacagctca
ggaagcagtg gccggagcag cggtgctggg taccctgggc acacctggac tggtgtcccc ggaagcagtg gccggagcag cggtgctggg taccctgggc acacctggac tggtgtcccc
agcactgacc ctggcccagc ccctgggcac tttgccccag gctgtcatgg ctgcccaggc agcactgacc ctggcccagc ccctgggcac tttgccccag gctgtcatgg ctgcccaggc
acctggagtc atcacaggtg tgaccccagc ccgtcctcct atcccggtca ccatcccctc acctggagtc atcacaggtg tgaccccagc ccgtcctcct atcccggtca ccatcccctc
ggtgggagtg gtgaacccca tcctggccag ccctccaacg ctgggtctcc tggagcccaa ggtgggagtg gtgaacccca tcctggccag ccctccaacg ctgggtctcc tggagcccaa
gaaggagaag gaagaagagg agctgtttcc cgagtcagag cggccagaga tgctgagcga gaaggagaag gaagaagagg agctgtttcc cgagtcagag cggccagaga tgctgagcga
gcaggagcac atgagcatct cgggcagtag cgcccgacac atggtgatgc agaagctgct gcaggagcac atgagcatct cgggcagtag cgcccgacac atggtgatgc agaagctgct
ccgcaagcag gagtctacag tgatggttct gcgcaacatg gtggacccca aggacatcga ccgcaagcag gagtctacag tgatggttct gcgcaacatg gtggacccca aggacatcga
tgatgacctg gaaggggagg tgacagagga gtgtggcaag ttcggggccg tgaaccgcgt tgatgacctg gaaggggagg tgacagagga gtgtggcaag ttcggggccg tgaaccgcgt
catcatctac caagagaaac aaggcgagga ggaggatgca gaaatcattg tcaagatctt catcatctac caagagaaac aaggcgagga ggaggatgca gaaatcattg tcaagatctt
tgtggagttt tccatagcct ctgagactca taaggccatc caggccctca atggccgctg tgtggagttt tccatagcct ctgagactca taaggccatc caggccctca atggccgctg
gtttgctggc cgcaaggtgg tggctgaagt gtacgaccag gagcgttttg ataacagtga gtttgctggc cgcaaggtgg tggctgaagt gtacgaccag gagcgttttg ataacagtga
cctctctgcg tga 尚、ェクソン 2をコードする塩基配列(配列番号 4 : 87bp)の一例は以下のとおりであ る: AGTGGTGGCAGCGGGAGACAAATGGAAACCTCCACAG0 cctctctgcg tga An example of the base sequence encoding exon 2 (SEQ ID NO: 4: 87 bp) is as follows: AGTGGTGGCAGCGGGAGACAAATGGAAACCTCCACAG 0
[0019] 本発明の蛋白質 (又は、ポリペプチド)をコードする DNAは当業者であれば、本明 細書の記載及び当該技術分野における公知技術に基き容易に調製することが可能 である。例えば、癌組織力ゝら調製された mRNAを铸型として使用する RT— PCR、及 び、適当な cDNAライブラリーに対してその他の NASBA (Nucleic acid sequence base d amplincation)法、 TMA (Transcription-mediated amplification)法、 SDA (Strand Dis placement Amplification)法及び ICAN(isothermal and chimeric primer-initiated ampli fication of nucleic acids)法等の当業者に公知の任意の DNA増幅技術を用いること により、 cDNAとして容易に得ることが可能である。尚、ェクソン 2部分を含むヒトの FI R遺伝子は、 NCBIの GenBankに登録番号 NM_014281として登録されている。従つ て、 RT— PCR等に使用するプライマーの塩基配列は、この登録された塩基配列、及 び特許文献 1又は特許文献 2に開示された情報に基き適宜設計 '選択することが出 来る。 [0019] A DNA encoding the protein (or polypeptide) of the present invention can be easily prepared by those skilled in the art based on the description in the present specification and known techniques in the technical field. For example, RT-PCR using mRNA prepared from cancer tissue force as a template, and other NASBA (Nucleic acid sequence base amplification) methods and TMA (Transcription-mediated) for appropriate cDNA libraries. Amplification), SDA (Strand Displacement Amplification) and ICAN (isothermal and chimeric primer-initiated amplification of nucleic acids) methods, etc. It is possible. The human FIR gene containing the exon 2 part is registered under NCBI GenBank as registration number NM_014281. Therefore, the base sequence of a primer used for RT-PCR or the like can be appropriately designed and selected based on the registered base sequence and the information disclosed in Patent Document 1 or Patent Document 2.
[0020] 当業者に公知の部位特異的突然変異誘発に基づき、市販のミューテーシヨンシステ ム等を用いて該 DNAに塩基変異を導入して調製することも可能である。更には、 PC R又は適当な制限酵素で処理することによって得られる、このような DNAの一部から 成るオリゴヌクレオチドも上記の DNAに含まれる。  [0020] Based on site-directed mutagenesis known to those skilled in the art, it is also possible to prepare by introducing a base mutation into the DNA using a commercially available mutation system or the like. Furthermore, an oligonucleotide consisting of a part of such DNA obtained by treating with PCR or an appropriate restriction enzyme is also included in the above DNA.
[0021] 又、上記 DNAは、公知の方法(例えば、 Carruthers (1982) Cold Spring Harbor Sym p. Quant. Biol. 47:411— 418;Adams (1983) J. Am. Chem. Soc. 105:661; Belousov(19 97) Nucleic Acid Res. 25:3440-3444; Frenkel (1995) Free Radic. Biol. Med. 19:373— 380; Blommers (1994) Biochemistry 33:7886—7896; Narang (1979) Meth. Enzymol. 68 :90; Brown (1979) Meth. Enzymol. 68: 109; Beaucage (1981) Tetra. Lett. 22:1859;米 国特許第 4,458,066号)に記載されているような周知の化学合成技術により、 in vitro において合成することもできる。また、本発明のポリヌクレオチドを適当な制限酵素で 切断する等の方法によって作製することもできる。 [0022] 本明細書において、「ストリンジェント(stringent)な条件」とは、前記のポリヌクレオチド またはオリゴヌクレオチドと、ゲノム DNAとの選択的かつ検出可能な特異的結合を可 能とする条件である。ストリンジェント条件は、塩濃度、有機溶媒 (例えば、ホルムアミ ド)、温度、およびその他公知の条件の適当な組み合わせによって定義される。すな わち、塩濃度を減じるか、有機溶媒濃度を増加させるカゝ、またはハイブリダィゼーショ ン温度を上昇させるかによつてストリンジエンシー(stringency)は増加する。更に、ノヽ イブリダィゼーシヨン後の洗浄の条件もストリンジエンシーに影響する。この洗浄条件 もまた、塩濃度と温度によって定義され、塩濃度の減少と温度の上昇によって洗浄の ストリンジエンシーは増加する。 [0021] In addition, the above DNA can be obtained by known methods (for example, Carruthers (1982) Cold Spring Harbor Symp. Quant. Biol. 47: 411—418; Adams (1983) J. Am. Chem. Soc. 105: 661 ; Belousov (19 97) Nucleic Acid Res. 25: 3440-3444; Frenkel (1995) Free Radic. Biol. Med. 19: 373—380; Blommers (1994) Biochemistry 33: 7886—7896; Narang (1979) Meth. Enzymol. 68: 90; Brown (1979) Meth. Enzymol. 68: 109; Beaucage (1981) Tetra. Lett. 22: 1859; US Pat. No. 4,458,066). It can also be synthesized in vitro. It can also be produced by a method such as cleaving the polynucleotide of the present invention with an appropriate restriction enzyme. In the present specification, “stringent conditions” are conditions that enable selective and detectable specific binding between the polynucleotide or oligonucleotide and genomic DNA. . Stringent conditions are defined by a suitable combination of salt concentration, organic solvent (eg, formamide), temperature, and other known conditions. That is, stringency increases by decreasing salt concentration, increasing organic solvent concentration, or increasing hybridization temperature. In addition, the washing conditions after the noisyization also affect the stringency. This wash condition is also defined by salt concentration and temperature, and the stringency of the wash increases with decreasing salt concentration and increasing temperature.
[0023] 従って、「ストリンジ ントな条件」とは、各塩基配列間の相同性の程度力 例えば、全 体の平均で約 80%以上、好ましくは約 90%以上、より好ましくは約 95%以上である ような、高い相同性を有する塩基配列間のみで、特異的にハイブリッドが形成される ような条件を意味する。具体的には、例えば、温度 60°C〜68°Cにおいて、ナトリウム 濃度 150〜900mM、好ましくは 600〜900mM、 pH 6〜8であるような条件を挙げること が出来る。ストリンジェントな条件の一具体例としては、 5 X SSC (750 mM NaCl、 75 m Mクェン酸三ナトリウム)、 1% SDS、 5 Xデンハルト溶液 50%ホルムアルデヒド、及び 42 °Cの条件でハイブリダィゼーシヨンを行い、 0.1 X SSC (15 mM NaCl、 1.5 mMクェン 酸三ナトリウム)、 0.1% SDS、及び 55°Cの条件で洗浄を行うものである。  Therefore, the “stringent condition” means the degree of homology between each base sequence, for example, about 80% or more, preferably about 90% or more, more preferably about 95% or more on average in the whole. This means that the hybrid is specifically formed only between the base sequences having high homology. Specifically, for example, the conditions include a sodium concentration of 150 to 900 mM, preferably 600 to 900 mM, and a pH of 6 to 8 at a temperature of 60 ° C to 68 ° C. Specific examples of stringent conditions include 5 X SSC (750 mM NaCl, 75 mM trisodium citrate), 1% SDS, 5 X Denhardt's solution 50% formaldehyde, and 42 ° C. The sample is washed and washed under the conditions of 0.1 X SSC (15 mM NaCl, 1.5 mM trisodium citrate), 0.1% SDS, and 55 ° C.
[0024] ハイブリダィゼーシヨンは、例えば、カレント 'プロトコーノレズ'イン'モレキュラ^ ~ ·バイ ォロン■ ~" (Current protocols in molecular biology (.edited by Frederick M. Ausubel et al, 1987) )に記載の方法等、当業界で公知の方法あるいはそれに準じる方法に従 つて行なうことができる。また、市販のライブラリーを使用する場合、添付の使用説明 書に記載の方法に従って行なうことができる。  [0024] Hybridization is described, for example, in Current Protocol in Molecular Biology (.edited by Frederick M. Ausubel et al, 1987). It can be carried out according to a method known in the art such as the method described, or a method analogous thereto, and when a commercially available library is used, it can be carried out according to the method described in the attached instruction manual.
[0025] 更に、こうして取得された本発明蛋白質をコードする DNAを、プラスミドベクター、フ ァージベクター、及び各種の混成ベクター等の適当な組換え用 DNAに挿入し、こう して得られた発現ベクターを用いて各種の細胞を形質転換させ、該形質転換体を適 当な培養条件で培養することにより、本発明の蛋白質を産生させることが出来る。こ の組換え用 DNAは、当業者に公知の通常の組換え DNA手法によって取り扱うこと が可能な任意のベクターである。 [0025] Furthermore, the thus obtained DNA encoding the protein of the present invention is inserted into an appropriate recombination DNA such as a plasmid vector, a fuzzy vector, and various hybrid vectors, and the expression vector thus obtained is obtained. The protein of the present invention can be produced by transforming various cells using, and culturing the transformant under appropriate culture conditions. This recombination DNA should be handled using conventional recombinant DNA techniques known to those skilled in the art. Can be any vector.
[0026] 本発明において蛋白質をコードする DNAを含むベクターによって形質転換される 宿主細胞に特に制限はないが、 HeLa等のヒト由来細胞、 COS7、及び CHO細胞等 の哺乳類細胞、昆虫細胞、並びに酵母等の真核細胞、更に、大腸菌及び枯草菌等 の各種の細菌を含む各種の原核細胞を用いることができる。  [0026] In the present invention, host cells transformed with a vector containing a DNA encoding a protein are not particularly limited, but human-derived cells such as HeLa, mammalian cells such as COS7 and CHO cells, insect cells, and yeasts In addition, various prokaryotic cells containing various bacteria such as E. coli and Bacillus subtilis can be used.
[0027] 或いは、ゥサギ網状赤血球溶解物又は小麦胚芽抽出物などの当業者に公知の適 当なインビトロ翻訳系を用いて本発明の蛋白質を産生させることも出来る。  [0027] Alternatively, the protein of the present invention can be produced using an appropriate in vitro translation system known to those skilled in the art, such as a rabbit reticulocyte lysate or a wheat germ extract.
[0028] 尚、本発明の蛋白質は、精製操作及び検出等の便宜の為に、 HAタグ、 Hisタグ、 FLAGタグ、及び mycタグ等の当業者に公知の任意のタグとの融合蛋白質として産 生させることも可能である。更には、蛋白質の測定 '検出の便宜の為に、 GSTなどの 酵素、及び各種の蛍光蛋白質との融合蛋白質として産生することも可能である。  [0028] The protein of the present invention is produced as a fusion protein with any tag known to those skilled in the art such as HA tag, His tag, FLAG tag, and myc tag for convenience of purification operation and detection. It is also possible to make it live. Furthermore, for convenience of protein measurement and detection, it can also be produced as a fusion protein with enzymes such as GST and various fluorescent proteins.
[0029] 上記の発現ベクターには、使用する発現系、宿主細胞等に応じて、当業者に公知 の、構成的発現プロモーター又は各種の誘導型発現プロモーター等の各種プロモ 一ター、ェンノヽンサ一及びサイレンサー等の各種調節配列、リボソーム結合部位、シ グナル配列、複製オリジン、クローユング部位、翻訳開始配列、ターミネータ一部位、 及びポリ A付加部位等の各種要素ならびにその他の外来性あるいは内在性タンパク 質をコードする遺伝子、各種薬剤耐性遺伝子、栄養要求性を相補する遺伝子等を 適宜含むことができる。  [0029] The above expression vectors include various promoters such as constitutive expression promoters or various inducible expression promoters known to those skilled in the art, depending on the expression system used, host cells, and the like. And various regulatory elements such as silencers, ribosome binding sites, signal sequences, replication origins, cloning sites, translation initiation sequences, partial positions of terminators, and poly A addition sites, as well as other foreign or endogenous proteins. It can appropriately contain genes to be encoded, various drug resistance genes, genes that complement auxotrophy, and the like.
[0030] 即ち、当業者に周知の任意の方法に従い、本発明の蛋白質をコードする DNAを、 これらのベクターは、その導入すべき宿主細胞に依存して適当に選択することが出 来る。一例として、真核細胞系では、 pCGNM2ベクタープラスミド(6)、 pKAl, pCDM8 ,及び pSVK3等の当業者に公知のベクターを挙げることが出来る。このようなベクター は、宿主細胞の中に導入され、本発明の蛋白質を一過性で発現させたり、或いは、 宿主細胞のゲノムの中にその全体あるいはその一部がゲノム中の 1箇所以上に糸且込 まれることができる。このようなベクターとして、当業者に公知の各種の市販のベクタ 一を使用することが出来る。  That is, according to any method known to those skilled in the art, DNA encoding the protein of the present invention can be appropriately selected depending on the host cell into which these vectors are to be introduced. As an example, in eukaryotic cell systems, vectors known to those skilled in the art such as pCGNM2 vector plasmid (6), pKAl, pCDM8, and pSVK3 can be mentioned. Such a vector can be introduced into a host cell to transiently express the protein of the present invention, or the entire or part of the host cell genome can be located in one or more places in the genome. Can be threaded. As such a vector, various commercially available vectors known to those skilled in the art can be used.
[0031] 上記発現ベクターの代わりに、 PCR増幅等により取得される本発明の蛋白質をコ ードする遺伝子を含む適当な DNA断片自体を用いて本発明の形質転換体を得るこ とも可能である。そのような場合には、力かる DNA断片に加えてさらに適当な緩衝液 及びその他の助剤を任意に含む溶液等の組成物として形質転換に使用することが できる。 [0031] The transformant of the present invention can be obtained by using an appropriate DNA fragment itself containing a gene encoding the protein of the present invention obtained by PCR amplification or the like instead of the expression vector. Both are possible. In such a case, the composition can be used for transformation as a composition such as a solution optionally containing an appropriate buffer and other auxiliaries in addition to a strong DNA fragment.
[0032] 本発明の DNAを含む発現ベクター (組換え用 DNA)又は DNA断片自体、例えば 、 し法、 elctroporation)、 Lipofectamine Plus reagents (Gibco BRL入 ¾ [匕力ノレ シゥム法、プロトプラスト- PEG法、 Tiプラスミド法、パーティクルガン法、ノ キュロウィ ルス法などの公知の任意の方法によって宿主細胞へと導入でき、形質転換体を作成 することができる。更に、複数種の組換え DNAを用いるコトランスフエクシヨン法によ つても可能である。  [0032] The expression vector (DNA for recombination) containing the DNA of the present invention or the DNA fragment itself, such as, for example, the method, elctroporation, Lipofectamine Plus reagents (Gibco BRL input [repulsive norme method, protoplast-PEG method, It can be introduced into a host cell by any known method such as Ti plasmid method, particle gun method, nanovirus method, etc. Further, a transformant can be prepared. It is also possible by the Yong method.
[0033] 本発明の蛋白質を発現する転換体を該蛋白質の生産に好ましい条件で培養して 該蛋白質を発現させ、その宿主細胞および Zまたは培地力 回収することにより製造 することができる。宿主細胞の培養に用いる培地は、当業者に公知である任意の培 地の中から、使用する発現ベクターの構成 (プロモーターの種類等)及び宿主の種類 等に応じて適当なものを適宜選択することができる。  [0033] A transformant that expresses the protein of the present invention can be produced by culturing the protein under conditions favorable for production of the protein, expressing the protein, and recovering the host cell and Z or medium power. The medium used for host cell culture is appropriately selected from any medium known to those skilled in the art, depending on the configuration of the expression vector used (type of promoter, etc.) and the type of host. be able to.
[0034] 宿主細胞により産生された本発明の蛋白質は、当業者に公知の任意の手段の適 当な組み合わせ、例えば、遠心または濾過による培地と細胞の分離、および硫酸ァ ンモ-ゥムの様な塩による培地のタンパク質成分の沈殿、及びこれに続く疎水クロマ トグラフィー、イオン交換クロマトグラフィー、ァフィユティークロマトグラフィー、又はそ の他のクロマトグラフィーの使用により培地から回収することができる。或いは、本発 明の蛋白質は化学合成法により製造することも可能である。  [0034] The protein of the present invention produced by a host cell is used in an appropriate combination of any means known to those skilled in the art, for example, separation of a medium and cells by centrifugation or filtration, and an ammonium sulfate-like protein. It can be recovered from the medium by precipitation of the protein components of the medium with the appropriate salt and subsequent use of hydrophobic chromatography, ion exchange chromatography, affinity chromatography, or other chromatography. Alternatively, the protein of the present invention can be produced by a chemical synthesis method.
[0035] [態様 5〜7] [0035] [Aspects 5-7]
本発明の FIR遺伝子のイントロン 2における 4塩基繰り返し配列における遺伝子多型 を示す塩基配列を含む DNA (オリゴヌクレオチド)は、例えば、 1〜2100個(例えば、 2058個程度)、好ましくは 1〜: L 100個(例えば、 1032個程度)、より好ましくは 1〜6 00個、更に好ましくは 40〜600個(例えば、 513個程度)の連続した塩基配列から成 る。このような DNAの例として、以下に示す配列番号 3 (FIR遺伝子のェクソン 2及び イントロン 2を含むゲノム DNA)に含まれる配列をあげることが出来る。  The DNA (oligonucleotide) containing a base sequence showing a gene polymorphism in the 4-base repeat sequence in intron 2 of the FIR gene of the present invention is, for example, 1 to 2100 (for example, about 2058), preferably 1 to: L It consists of a continuous base sequence of 100 (for example, about 1032), more preferably 1 to 600, and still more preferably 40 to 600 (for example, about 513). As an example of such DNA, a sequence contained in SEQ ID NO: 3 (genomic DNA containing exon 2 and intron 2 of the FIR gene) shown below can be mentioned.
[0036] 己列番号 3 : 2486bp] Exon2[0036] Self-row number 3: 2486bp] Exon2
Figure imgf000015_0001
Figure imgf000015_0001
catcctctccctcagg[cccgcccgcctgcccgcccgcccgcccgcctgcctg]tgcgtccctgggcccagagagggc tggctgggggcggggcgggaggaacctcgggaacccttctctgcctctgggcagacaactgcccgcaggaccacggc cctctccccgcaccagccctgcaccctccccacccaggcactgagctggcctttgccctctgcatctgccctgctccaa gcaccaggcggggcctgttgacagtctggctgggcggggacgccggggcctggagctcgcgcgttcttaggcatgagc gtcttgcaggaagggtgggccaagcttcccagaactcgcagtgcttgaggagccagaaggacagatttcatttcagacc tctgtgttttcatttaaataaaacagtgaagcaatgcgcaagtcaaagcacaggtctggttgggactcagccctccccat cttttttcggccccaggctccagagtgggtgcaggtgggtcttggtctgttcgatggcccagccttgggagcaggtcctc tcagaggtggagctgggcctgtggctgggtagttgggttctttgtgcttctccatggtagcgcccgcctccagagcattc cagtggggcctcagcaaggtggcctggcagcctcttggggctgggccctcctcttggtcccccaggcacactcagggg gtgggactgggacatggtatgcagcttggatcctgggagatgtgggggctggtgctccctggcccttggctggcgccca gccggccattggttggtgggagaagggccttttgtttaccaagacaaagaggagattgaaaaatgtctgggggctgagg gggttggcattgattaccatggtgacagcctgctctcaagtcccagcctctggctcttgggccctctttcccactaagga ggaatgggcttggggccagctgtgagggtggtgaggacaggcctagtgtgagccggactggggcacctggtgctgagg gtggtgaggacaggcctagtgtgagccggactggggcacctggtgctgagggtggtgaagacaggcctagtgtgagcc ggactggggcacctggtgctgagggtgattaggacaggcctggtgtgagccggactggggcacctggtgccacccaca gccagcctttgaccttcagggactggttggggctctggttgcctggggaggggtctccagggcttcctgccctttgagcc tgggccagcatcccagcacttggcaggcaggagggggttcttggggcttgggccagcctgtcccttcctgcctcctgtg tccctctgtccttgctcatcacttctgtgctggggacccttctctcttctctgctgtccctcaaggctgtgtctggaagtct cggagggcccggttggtgggccagtgatctttgtgcttgctgagctggaggacaggcgcaggcatgctgtttgcccatc ccacactctgatgggcctgctagccatgcctccctgctcacacagctgcgttcttccctctgcaaaaagcttagagatgg attttgacaggagggacgtaaacataatcctggttggcatcagcttgggtctgcagtggctgtgactgcccaagggcctc ttggtccccaggcccacctggctagggtgccagtggggtctgctcctggcttctcccagtgggccaggcaaagcccatg caagagtgtgtggaagtgagctggagccctcttcagggcttgtgtggcccgagagaggcctgtgttctctgggaggtgt gcacagtgcctgcggggacggtggggggagcactgttgggagagaggcccttgtattagctccttgggtgggccccttg aaactccagggaagctgagcctgtgtctgtggcagggactgtgaggaagcctggaggagcctgggagcaaggagtccc agaacgtccagagagccgtggcccttttctcctgccttggaggacggctgaggctggctctcctgccgggcatgtgata gcagctctggggtcagactgacaacaaggggctggaccccttttctgcctccagctgtttatcttggtggttcccgagga ggccactgtggcctgggccccacaactgctcgggttggcccagggcctccagggcacctgcctgcctcagtctgtgttc taccccatgggtcccgggggagacagtggcctgggctggggacagcaatgtcagaagggctggcctgagtacaggcc aggcaggggacagtctgtgctaggggtggccctcctggggccacgtgaaggagaacttgagcttgctgctgatggattc ccctgctcccttaccag catcctctccctcagg [cccgcccgcctgcccgcccgcccgcccgcctgcctg] tgcgtccctgggcccagagagggc tggctgggggcggggcgggaggaacctcgggaacccttctctgcctctgggcagacaactgcccgcaggaccacggc cctctccccgcaccagccctgcaccctccccacccaggcactgagctggcctttgccctctgcatctgccctgctccaa gcaccaggcggggcctgttgacagtctggctgggcggggacgccggggcctggagctcgcgcgttcttaggcatgagc gtcttgcaggaagggtgggccaagcttcccagaactcgcagtgcttgaggagccagaaggacagatttcatttcagacc tctgtgttttcatttaaataaaacagtgaagcaatgcgcaagtcaaagcacaggtctggttgggactcagccctccccat cttttttcggccccaggctccagagtgggtgcaggtgggtcttggtctgttcgatggcccagccttgggagcaggtcctc tcagaggtggagctgggcctgtggctgggtagttgggttctttgtgcttctccatggtagcgcccgcctccagagcattc cagtggggcctcagcaaggtggcctggcagcctcttggggctgggccctcctcttggtcccccaggcacactcagggg gtgggactgggacatggtatgcagcttggatcctgggagatgtgggggctggtgctccctggcccttggctggcgccca gccggccattggttggtgggagaagggccttttgtttaccaagacaaagaggagattgaaaaatgtctgggggctgagg gggttggcattgattaccatggtgacagcctgctctcaagtcccagcctctggctcttgggccctctttcccactaagga ggaatgggcttggggccagctgtgagggtggtgaggacagg cctagtgtgagccggactggggcacctggtgctgagg gtggtgaggacaggcctagtgtgagccggactggggcacctggtgctgagggtggtgaagacaggcctagtgtgagcc ggactggggcacctggtgctgagggtgattaggacaggcctggtgtgagccggactggggcacctggtgccacccaca gccagcctttgaccttcagggactggttggggctctggttgcctggggaggggtctccagggcttcctgccctttgagcc tgggccagcatcccagcacttggcaggcaggagggggttcttggggcttgggccagcctgtcccttcctgcctcctgtg tccctctgtccttgctcatcacttctgtgctggggacccttctctcttctctgctgtccctcaaggctgtgtctggaagtct cggagggcccggttggtgggccagtgatctttgtgcttgctgagctggaggacaggcgcaggcatgctgtttgcccatc ccacactctgatgggcctgctagccatgcctccctgctcacacagctgcgttcttccctctgcaaaaagcttagagatgg attttgacaggagggacgtaaacataatcctggttggcatcagcttgggtctgcagtggctgtgactgcccaagggcctc ttggtccccaggcccacctggctagggtgccagtggggtctgctcctggcttctcccagtgggccaggcaaagcccatg caagagtgtgtggaagtgagctggagccctcttcagggcttgtgtggcccgagagaggcctgtgttctctgggaggtgt gcacagtgcctgcggggacggtggggggagcactgttgggagagaggcccttgtattagctccttgggtgggccccttg aaactccagggaagctgagcctgtgtctgtggcagggactgtgaggaagcctggaggagcctgggagcaaggagtccc agaacgtccagagagccgtggcccttttctcctgccttggaggacggctgaggctggctctcctgccgggcatgtgata gcagctctggggtcagactgacaacaaggggctggaccccttttctgcctccagctgtttatcttggtggttcccgagga ggccactgtggcctgggccccacaactgctcgggttggcccagggcctccagggcacctgcctgcctcagtctgtgttc taccccatgggtcccgggggagacagtggcctgggctggggacagcaatgtcagaagggctggcctgagtacaggcc aggcaggggacagtctgtgctaggggtggccctcctggggccacgtgaaggagaacttgagcttgctgctgatggattc ccctgctcccttaccag
[0037] 上記の配列番号 3にお 、て、大文字で示した最初の配列がェクソン 2 (87bp)であ り、それに続く小文字で表した配列がイントロン 2の配列(2399bp)である。更に、括 弧で囲まれた部分力 塩基繰り返し (9回)配列における遺伝子多型を示す配列であ る。 [0037] In SEQ ID NO: 3, the first sequence shown in capital letters is exon 2 (87 bp), and the sequence shown in lower case following is the sequence of intron 2 (2399 bp). Furthermore, it is a sequence showing a gene polymorphism in a partial force base repeat (9 times) sequence surrounded by parentheses.
[0038] この 4塩基繰り返し配列には、 9回、 6回、及び 5回の繰り返しが見られ、 4塩基繰り返 しの塩基配列の具体例として、 CCCG又は CCTGをあげることが出来る。尚、図 8に示 されるように、これらの 4塩基繰り返し配列は、その繰り返し回数又は繰り返し単位の 塩基配列の種類においてホモ又はへテロの場合が見られる。  [0038] In this 4-base repeat sequence, 9-, 6-, and 5-fold repeats are observed, and CCCG or CCTG can be given as a specific example of the base sequence of 4 base repeats. In addition, as shown in FIG. 8, these 4-base repeat sequences are sometimes homozygous or heterozygous in the number of repeats or the type of base sequence of the repeat unit.
[0039] 以下の実施例に示されるように、 FIRは癌組織のみならず、正常組織においても発 現して 、る例がみられるのに対して、 FIRのェクソン 2が欠失したスプライシングバリア ントである本発明の蛋白質は、癌患者の癌組織細胞においてのみ発現され、癌組織 に隣接する正常組織細胞又は血液細胞では発現していない。従って、このような蛋 白質及びそれをコードする cDNA若しくは対応する mRNA又はそれらの一部力ゝらな るオリゴヌクレオチドは、癌の検出 ·診断等において非常に特異的な癌の検出マーカ 一としての有用性を有する物質である。  [0039] As shown in the following Examples, FIR is expressed not only in cancer tissues but also in normal tissues, whereas in some cases, splicing variants lacking exon 2 of FIR are observed. The protein of the present invention is expressed only in cancer tissue cells of cancer patients and not expressed in normal tissue cells or blood cells adjacent to the cancer tissue. Therefore, such a protein and its encoding cDNA or corresponding mRNA, or an oligonucleotide that partially relies on them, serve as a very specific cancer detection marker in cancer detection and diagnosis. It is a substance that has utility.
[0040] 更に、癌組織において、 FIR遺伝子のイントロン 2における 4塩基繰り返し配列にお いて 9回の繰り返しのホモ (繰り返し単位の塩基配列の種類におけるヘテロを含む) の存在する割合が正常組織と比較して有意に高い為、このような遺伝子多型を示す 塩基配列を含む上記の DNAも、癌の検出'診断等において癌の検出マーカーとし て有用な物質である。  [0040] Furthermore, in the cancer tissue, the proportion of 9 homo repeats (including hetero in the base sequence type of repeat units) in the 4-base repeat sequence in intron 2 of the FIR gene is compared with that in normal tissues. Therefore, the above DNA containing a nucleotide sequence showing such a gene polymorphism is also a useful substance as a cancer detection marker in cancer detection and diagnosis.
[0041] [態様 8〜18] 従って、以上の FIRのェクソン 2が欠失したスプライシングバリアント蛋白質又はその 断片ペプチド、それらをコードする cDNA若しくはそれに対応する mRNA、又はそれ らの一部力もなるオリゴヌクレオチド、又は、 FIR遺伝子のイントロン 2における 4塩基 繰り返し配列における遺伝子多型を示す塩基配列を含む連続した塩基配列から成 る DNA (ゲノム DNA)の存在を測定することによって、癌を有意に検出又は診断する ことが可能となる。対象となる癌の種類に特に制限はないが、特に、大腸癌等に代表 される消ィ匕器癌が好適である。 [0041] [Aspects 8 to 18] Therefore, the splicing variant protein or its fragment peptide lacking exon 2 of the above FIR, the cDNA encoding them or the mRNA corresponding thereto, or a partial oligonucleotide thereof, or in the intron 2 of the FIR gene Cancer can be detected or diagnosed significantly by measuring the presence of DNA (genomic DNA) consisting of a continuous base sequence containing a base sequence indicating a gene polymorphism in a 4-base repeat sequence. There are no particular restrictions on the type of cancer to be treated, but particularly cancer cancer typified by colorectal cancer is preferred.
[0042] 癌の検出は、例えば、癌糸且織における上記のスプライシングバリアント蛋白質、 mRN A (cDNA)又はオリゴヌクレオチドの発現量を測定し、これを同一の癌患者由来の正 常組織又は正常人の組織における発現量を測定し、これらの値を比較すること等に よって行うことが出来る。尚、このような発現量の測定は、測定方法'原理に応じて、 定量的、半定量的、又は定性的であり得る。特に、本発明の FIRのェクソン 2が欠失 したスプライシングバリアント蛋白質又はそれをコードする mRNA(cDNA)は癌糸且織 のみにおいて特異的に発現しているので、癌を簡便に且つ高感度 (高特異性)で検 出することが可能となる。  [0042] For example, cancer is detected by measuring the expression level of the above-mentioned splicing variant protein, mRNA (cDNA) or oligonucleotide in cancer thread and tissue, and using this as a normal tissue or normal person derived from the same cancer patient. This can be done by measuring the expression level in the tissues and comparing these values. Such measurement of the expression level can be quantitative, semi-quantitative, or qualitative depending on the principle of the measurement method. In particular, the splicing variant protein lacking exon 2 of the FIR of the present invention or the mRNA (cDNA) encoding the same is expressed specifically only in the cancer thread and tissue, so that cancer can be conveniently and highly sensitive (highly sensitive). (Specificity) can be detected.
[0043] スプライシングバリアント蛋白質の発現量は当業者に公知の任意の方法で測定する ことが可能である。例として、例えば、適当な抗体を用いたウェスタンプロット法、免疫 染色及び EIA等の各種の免疫学的特異反応を利用する方法、エドマン法を用いた 気相シークェンサ一等ペプチドのアミノ酸配列分析法、更には、 MALDI-TOF/ MS及び ESI Q— TOFZMS法等に代表される質量分析によって検出することが 出来る。  [0043] The expression level of the splicing variant protein can be measured by any method known to those skilled in the art. Examples include Western plotting using appropriate antibodies, immunostaining and methods using various immunological specific reactions such as EIA, amino acid sequence analysis of gas phase sequencer etc. peptides using Edman's method, Furthermore, it can be detected by mass spectrometry represented by MALDI-TOF / MS and ESI Q-TOFZMS methods.
[0044] 上記の方法の中でも、 FIR又はそのスプライシングバリアントに特異的な抗体との抗 原抗体反応によって該白質の発現量を測定する検査方法が好適である。このような 抗体には、例えば、 FIR及び FIRのェクソン 2が欠失したスプライシングバリアントの 双方を抗原として認識する抗体が含まれる。その他に、例えば、 FIRのェクソン 2に特 異的な抗体及びェクソン 5に特異的な抗体等の複数種類の抗体を組み合わせて測 定し、それらの結果力 該白質の発現を検出することも可能である。  [0044] Among the above methods, a test method is preferred in which the expression level of the white matter is measured by an antigen antibody reaction with an antibody specific for FIR or a splicing variant thereof. Such antibodies include, for example, antibodies that recognize both FIR and a splicing variant lacking exon 2 of FIR as antigens. In addition, it is possible to measure multiple types of antibodies, such as antibodies specific to FIR exon 2 and antibodies specific to exon 5, and detect the expression of the resulting white matter. It is.
[0045] 従って、上記抗体は、 FIR若しくは FIRのェクソン 2が欠失したスプライシングバリアン ト蛋白質、又はその適当な部分ポリペプチド (ペプチド断片)又はそれらの各種誘導 体又は複合体等を抗原物質又は免疫原として用いて、当業者に公知の適当な方法 で調製することが可能である。例えば、ポリクローナル抗体の場合には、マウス、ラット 、ゥサギ、ャギ、 -ヮトリ等の適当な動物に投与し、その抗血清力も調製することが可 能である。或いは、モノクローナル抗体作成法(「単クローン抗体」、長宗香明、寺田 弘共著、廣川書店、 1990年; "Monoclonal Antibody" James W. Goding, third edition , Academic Press, 1996)等に記載の公知の細胞融合を用いる方法でモノクローナル 抗体として調製することも可能である。例えば、特許文献 1又は特許文献 2に記載の 方法で調製することが可能である。 [0045] Therefore, the above-mentioned antibody is a splicing variant that lacks FIR or exon 2 of FIR. Or a suitable partial polypeptide (peptide fragment) thereof or various derivatives or complexes thereof as antigenic substances or immunogens, and can be prepared by an appropriate method known to those skilled in the art. . For example, in the case of a polyclonal antibody, it can be administered to an appropriate animal such as mouse, rat, rabbit, goat, or chicken, and its antiserum can be prepared. Alternatively, known methods described in monoclonal antibody production methods (“monoclonal antibodies”, Nagamune Kamei, Hiroshi Terada, Yodogawa Shoten, 1990; “Monoclonal Antibody” James W. Goding, third edition, Academic Press, 1996) It is also possible to prepare a monoclonal antibody by a method using cell fusion. For example, it can be prepared by the method described in Patent Document 1 or Patent Document 2.
[0046] 尚、このような抗体は、その元来の抗体活性を失わない限り、遺伝子工学 (DNA組 換え技術)により、例えば、 Fab, F(ab')、 Fv断片等の完全な抗体由来の各種 [0046] Such an antibody is derived from a complete antibody such as Fab, F (ab '), Fv fragment, etc. by genetic engineering (DNA recombination technology) as long as the original antibody activity is not lost. Various
2  2
誘導体を含む、当業者に公知の様々な形態に改変された誘導体、組換え体又はフ ラグメントであっても良い。尚、このような抗体等には、酵素、放射性同位体蛍光色素 、及び金属原子等の当業者に公知の各種の標識物質で標識されているものも含ま れる。  It may be a derivative, recombinant or fragment modified into various forms known to those skilled in the art, including derivatives. Such antibodies and the like include those labeled with various labeling substances known to those skilled in the art, such as enzymes, radioisotope fluorescent dyes, and metal atoms.
[0047] 抗 FIR抗体との抗原抗体反応として、例えば、ウェスタンプロット法により測定する場 合には、 FIRのェクソン 2が欠失したスプライシングバリアント蛋白質(513個のアミノ 酸)と FIR (542個のアミノ酸)とを分子量に相違によって特異的に検出することが可 能となる。  [0047] As an antigen-antibody reaction with an anti-FIR antibody, for example, when measured by Western plotting, a splicing variant protein (513 amino acids) lacking FIR exon 2 and FIR (542 Amino acid) can be specifically detected by the difference in molecular weight.
[0048] 更に、 FIRのェクソン 2が欠失したスプライシングバリアント蛋白質にのみ特異的に反 応する抗体を使用して、例えば、 EIA等の酵素免疫測定法により測定することが可能 である。  [0048] Furthermore, using an antibody that specifically reacts only with a splicing variant protein lacking exon 2 of FIR, for example, it can be measured by an enzyme immunoassay such as EIA.
[0049] このような FIRのェクソン 2が欠失したスプライシングノリアントをコードする cDNA又 はそれに対応する mRNA、又はそれらの一部力 なるオリゴヌクレオチド、又は、 FI R遺伝子のイントロン 2における 4塩基繰り返し配列における遺伝子多型を示す塩基 配列を含む連続した塩基配列から成る DNAを含む核酸分子は、 RT— PCR法及び リアルタイム定量的 PCR等の各種 PCR法、並びに各種のマイクロアレイ(DNAチッ プ)法等の当業者に公知の方法で増幅することが出来る。 [0050] 例えば、 cDNA若しくは mRNA又はそれらの一部力 なるオリゴヌクレオチドを含む 核酸分子を増幅する為のプライマーセットを使用する RT— PCR法によって上記の 各 DNAを含む核酸分子を増幅し、電気泳動と組み合わせて測定されるそれらの分 子量から、当該核酸分子の検出 ·同定をすることができる。リアルタイム定量的 PCR ( インターカレーター法、 TaqManプローブ、サイクリングプローブ等)では、 FIRのエタ ソン 2が欠失したスプライシングバリアントに対応する mRNAの塩基配列(例えば、実 質的に FIRのェクソン 1及びェクソン 3をコードする連続する塩基配列力 成るオリゴ ヌクレオチド又はその部分オリゴヌクレオチド)のみを含む核酸分子を特異的に増幅 するようなプライマーを使用することによって、このような核酸分子を直接定量的に測 定することが可能となる。さらには、増幅された DNAの塩基配列を直接決定する方 法 (シークェンス法)を用いることも可能である。 [0049] A cDNA encoding a splicing noreant lacking exon 2 of such FIR or a corresponding mRNA, or a partial oligonucleotide thereof, or a 4-base repeat in intron 2 of the FIR gene Nucleic acid molecules containing DNA consisting of a contiguous base sequence that includes a base sequence that indicates a gene polymorphism in the sequence include various PCR methods such as RT-PCR and real-time quantitative PCR, and various microarray (DNA chip) methods. Can be amplified by methods known to those skilled in the art. [0050] For example, by using a primer set for amplifying a nucleic acid molecule containing cDNA or mRNA or an oligonucleotide that is a partial force thereof, a nucleic acid molecule containing each of the above DNAs is amplified by an RT-PCR method and electrophoresed The nucleic acid molecule can be detected and identified from the amount of the molecules measured in combination. In real-time quantitative PCR (intercalator method, TaqMan probe, cycling probe, etc.), the base sequence of mRNA corresponding to the splicing variant lacking FIR etason 2 (for example, FIR exon 1 and exon 3) Such a nucleic acid molecule is directly and quantitatively measured by using a primer that specifically amplifies a nucleic acid molecule containing only a contiguous nucleotide sequence encoding an oligonucleotide or a partial oligonucleotide thereof. It becomes possible. It is also possible to use a method (sequence method) for directly determining the base sequence of the amplified DNA.
[0051] 上記の RT—PCR等に使用するプライマーの塩基配列は、既に記載したような GenB ankに登録された FIRの塩基配列、及び特許文献 1又は特許文献 2に開示された情 報に基き適宜設計'選択することが出来る。尚、プライマーの設計に際しては、铸型と の特異的な結合が可能となるような塩基数、例えば、 15— 40塩基、より具体的には、 15— 25塩基程度を有することが好ましぐ更には、プライマー内でヘアピン構造をと つたり、センス鎖とアンチセンス鎖とが互いにアニーリングしないような塩基配列とする ことも重要である。例えば、 01igoTM(National Bioscience Inc.製)のような市販のプライ マー設計用のソフトウェアを使用することも可能である。  [0051] The base sequence of the primer used in the above RT-PCR and the like is based on the base sequence of FIR registered in GenBank as described above and the information disclosed in Patent Document 1 or Patent Document 2. You can choose 'design as appropriate'. In designing the primer, it is preferable to have a base number that allows specific binding to the cage, for example, 15-40 bases, more specifically, about 15-25 bases. Furthermore, it is also important to have a base sequence that does not have a hairpin structure in the primer or that the sense strand and the antisense strand are not annealed to each other. For example, commercially available primer design software such as 01igo ™ (National Bioscience Inc.) can be used.
[0052] 本発明の検出方法に使用される測定キットは、測定対象又は測定原理等に応じて、 適当な構成をとることが出来る。該キットは、その構成要素として、例えば、抗 FIR抗 体、 FIRのェクソン 2が欠失したスプライシングノリアント蛋白質に対する抗体、各種 の二次抗体 (標識抗体)、上記の mRNA (cDNA)の増幅用プライマー及び DNAチ ップ等で使用するハイブリダィゼーシヨン用のプローブ(例えば、 10〜: L00個程度の 連続した塩基配列から成る)を含むことが出来る。更に、上記キットには、その構成- 使用目的などに応じて、当業者に公知の他の要素又は成分、例えば、各種試薬、酵 素、緩衝液、反応プレート (容器)等が含まれる。  [0052] The measurement kit used in the detection method of the present invention can have an appropriate configuration depending on the measurement object or measurement principle. The kit includes, for example, an anti-FIR antibody, an antibody against a splicing noriant protein lacking exon 2 of FIR, various secondary antibodies (labeled antibodies), and the above-mentioned mRNA (cDNA) amplification. Probes for hybridization used in primers, DNA chips, etc. (for example, consisting of 10 to: about L00 continuous base sequences) can be included. Furthermore, the above kit contains other elements or components known to those skilled in the art, for example, various reagents, enzymes, buffers, reaction plates (containers), etc., depending on the configuration-purpose of use.
実施例 [0053] 以下、本発明を実施例によって詳細に説明するが、本発明の技術的範囲は以下の 実施例の記載によって何ら限定して解釈されるものではない。又、特に記載のない 場合には、以下の実施例は、例えば、 Sambrook and Maniatis, in Molecular Cloning- A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York, 1989; Ausu bel, F. M. et al., Current Protocols in Molecular Biology, John Wiley & Sons, New Y ork, N.Y, 1995等に記載されている、当業者に公知の標準的な遺伝子工学及び分 子生物学的技術に従い、実施することが出来る。又、本明細書中に引用された文献 の記載内容は本明細書の開示内容の一部を構成するものである。 Example Hereinafter, the present invention will be described in detail by way of examples. However, the technical scope of the present invention is not construed as being limited by the description of the following examples. Also, unless otherwise stated, the following examples are for example Sambrook and Maniatis, in Molecular Cloning-A Laboratory Manual, Cold Spring Harbor Laboratory Press, New York, 1989; Ausu bel, FM et al., Current Protocols in Molecular Biology, John Wiley & Sons, New York, NY, 1995, etc. can be performed according to standard genetic engineering and molecular biological techniques known to those skilled in the art. In addition, the contents of the references cited in this specification constitute part of the disclosure of this specification.
[0054] ヒト 組織標本 · mrnm ·健常者末梢血単核球の Β¾  [0054] Human tissue specimen · mrnm · Healthy human peripheral blood mononuclear cells
原発性大腸癌の治療目的で千葉大学病院に入院した患者から、術前に文書による 同意を得て癌部と非癌組織を採取し、マイナス 80°Cに保存した。各種ヒト癌細胞株 は Α し C し ell Biology Collection (http://www.atcc. org/¾earcnし atalogs/し ellBiology. c&i)力 入手した。健常者ボランティアは千葉大学医学研究院および千葉大学医学 部附属病院に勤務する健常者力 文書による同意を得て採取した。  Cancer patients and non-cancerous tissues were collected from patients admitted to Chiba University Hospital for the treatment of primary colorectal cancer with written consent prior to surgery and stored at minus 80 ° C. Various human cancer cell lines were obtained from the Cellellology Collection (http://www.atcc.org/¾earcn and atalogs / ellBiology.c & i). Volunteers for healthy volunteers were collected with the consent of the healthy volunteers working at the Chiba University Medical Research Institute and the Chiba University Hospital.
[0055] プラスミド [0055] Plasmid
癌組織から得られた FIR (完全長 FIR) cDNA及び FIRのェクソン 2が欠失したスプ ライシングバリアント cDNA (HA- FIR Δ exon2)を pCGNM2ベクタープラスミド (9)に導 入して、へマグルチニン (HA)タグがその N末端に付カ卩された、 HA-FIR及び HA-FIR A exon2を発現されるようにした。ヒト c- Myc発現ベクターは市販の pcDNA3.1- c- myc, ueneStorm Expression— Ready Clones (Invitrogen し o., AL) 使用した。  FIR (full-length FIR) cDNA obtained from cancer tissue and splicing variant cDNA (HA-FIR Δ exon2) lacking exon 2 of FIR were introduced into pCGNM2 vector plasmid (9) and hemagglutinin (HA ) HA-FIR and HA-FIR A exon2 with a tag attached to its N-terminus were expressed. The human c-Myc expression vector was a commercially available pcDNA3.1-c-myc, ueneStorm Expression—Ready Clones (Invitrogen, o., AL).
[0056] 免疫組織化学的染色およびフローサイトメトリー解析 [0056] Immunohistochemical staining and flow cytometry analysis
HeLa細胞をカバーグラス上で培養し Lipofectamine Plus reagents (Gibco BRL)により HA-FIRと HA- FIR Δ exon2遺伝子を含む上記のベクター (150 ftnol)を導入し c-Myc の発現変化を調べた。用いた一次抗体は mouse monoclonal抗 HA抗体 (Santa Cruz Biotechnology, C Aと rabbit polyclonal抗 c— My c¾ri体 (Upstate Biotechnology, NY)で それぞれ 500倍、 1,000倍希釈にて使用した。二次抗体 rhodamine- conjugated anti-m ouse IgG (Roche八 fluorescein isothiocyanate (FITし)一 conjugated anti— rabbit IgG (Sig ma)はそれぞれ 1,000倍、 500倍希釈して使用した。フローサイトメトリーは遺伝子導入 後 22時間後の HeLa細胞を trypsine処理し、上記の一次抗体を用いて FIRと c-Mycを それぞれ染色した。二次抗体は FITC- conjugated anti-rabbit IgG (Sigma)と R- PE- co njugated anti-mouse IgG (PharMingen)をそれぞれ 200倍希釈で用いた (km)。 10,000 個の細胞をカウントし c-Myc-FITCを FL1チャンネル HA-PEを FL2チャンネルで検出 した。 PE陽性細胞を X軸で表示した。 HeLa cells were cultured on a cover glass, and the above-mentioned vector (150 ftnol) containing HA-FIR and HA-FIR Δexon2 gene was introduced using Lipofectamine Plus reagents (Gibco BRL) to examine the change in expression of c-Myc. The primary antibodies used were mouse monoclonal anti-HA antibody (Santa Cruz Biotechnology, CA and rabbit polyclonal anti-c-My c¾ri (Upstate Biotechnology, NY), diluted 500-fold and 1,000-fold, respectively. Secondary antibody rhodamine-conjugated Anti-m ouse IgG (Roche 8 fluorescein isothiocyanate (FIT) -one conjugated anti-rabbit IgG (Sigma) was used diluted 1,000-fold and 500-fold, respectively. Flow cytometry was used for gene transfer. Twenty-two hours later, HeLa cells were treated with trypsine and stained with FIR and c-Myc using the primary antibody described above. As secondary antibodies, FITC-conjugated anti-rabbit IgG (Sigma) and R-PE-conjugated anti-mouse IgG (PharMingen) were used at a 200-fold dilution (km). 10,000 cells were counted and c-Myc-FITC was detected in FL1 channel and HA-PE in FL2 channel. PE positive cells were displayed on the X axis.
[0057] アポトーシスの検出  [0057] Detection of apoptosis
ポト ~~シス iiTUNEL assayにより検出し 7こ (Apoptosis Detection system, Fluorescei n. Promega, WI, USA)。 Two- color analysisは、 FITC-ラベルした dUTPを含む 50 μ \ の terminal deoxynucleotidyl transferase (TdT) buffer中で反応させた (MEBSTAIN Ap optosis Kit: Medical & Biological Laboratories, JAPAN)G最後に 250 μ g of DNase- fr ee RNase Aを含む 0.5 mlの propidium iodide (PI)溶液 (freshly diluted to 5 ^ g/ml i n PBS)に細胞を混和した。 10,000個の細胞をカウントし、 FITCを FL1に、 PIを FL2に 対応させた。アポトーシス細胞は Y軸に示される FITC-陽性細胞として示された。さら に Mouse monoclonal anti— caspase 9 (Upstate biotechnology, NY) 用 ヽ 7こウェスタン ブロット法により caspase 9の断片化を調べた。 Potato ~ cis 7 detected by iiTUNEL assay (Apoptosis Detection system, Fluorescein n Promega, WI, USA). Two-color analysis was performed in 50 μ \ terminal deoxynucleotidyl transferase (TdT) buffer containing FITC-labeled dUTP (MEBSTAIN Ap optosis Kit: Medical & Biological Laboratories, JAPAN) G Finally 250 μ g of DNase -Cells were mixed with 0.5 ml propidium iodide (PI) solution (freshly diluted to 5 ^ g / ml in PBS) containing fr ee RNase A. 10,000 cells were counted and FITC corresponded to FL1 and PI corresponded to FL2. Apoptotic cells were shown as FITC-positive cells shown on the Y axis. Furthermore, the fragmentation of caspase 9 was examined by Western blotting for mouse monoclonal anti-caspase 9 (Upstate biotechnology, NY).
[0058] 白皙の柚出 ウェスタン法  [0058] Egret of white birch Western method
凍結組織からの蛋白質の抽出は lysis buffer (7M urea, 2M thiourea, 2% 3-[(3-Chola midopropyl) dimethylammonio— ] 1— propanesulfate (CHAPS), 0.1 M Dithiothreitol (D TT), 2% IPG buffer (Amersham Phrmacia Biothech, Backinghampsnire, UK), 40 mM Tris)中で 500mgの凍結糸且織を Polytron homogenizer (Kinematica, Switzerland)を用 いて溶解し、 100,000 x gで 1時間 4°Cで高速遠沈しその上清を用いた。ウェスタンブ ロットに用いた一次抗体はゥサギポリクローナル抗 -FIR抗体(5)とャギポリクローナ ル抗 13 -actin抗体(Santa Cruz, Santa Cruz, CA)で、それぞれ 1 ,000倍、 500倍で使 用し 7こ。 2次抗体はャキ ゥサャ IgG horseradish peroxidase conjugate (HRP) (Jack son, west Grove, PA)で 3, 000倍希釈、ゥサギ抗ャギ IgG HRP (Cappel, West Chest er, PA)は 500倍希釈で使用した。抗原は ECL™ detection reagents (Amersham Phar macia Biothech)で検出し、発現レベルは NIH Imageにより定量化した。因みに、ポリ クローナル抗 -FIR抗体は 2種類の合成ペプチド(C (訂正) DKWKPPQGTDSIKME (ァ ミノ酸 30-45)及び C (追カロ) EVYDQERFDNSDLSA (アミノ酸 528-542))を同時に免疫し て作製したものである。 Extraction of protein from frozen tissue is lysis buffer (7M urea, 2M thiourea, 2% 3-[(3-Chola midopropyl) dimethylammonio—] 1—propanesulfate (CHAPS), 0.1 M Dithiothreitol (D TT), 2% IPG buffer (Amersham Phrmacia Biothech, Backinghampsnire, UK), 40 mM Tris) Dissolve 500 mg of frozen yarn and weave using Polytron homogenizer (Kinematica, Switzerland) and centrifuge at 100,000 xg for 1 hour at 4 ° C. The supernatant was used. The primary antibodies used for Western blotting are the rabbit anti-FIR antibody (5) and the goat polyclonal anti-13-actin antibody (Santa Cruz, Santa Cruz, CA), which are used at 1,000 and 500 times, respectively. 7 Secondary antibody is 3,000 times diluted with horseradish IgG horseradish peroxidase conjugate (HRP) (Jackson, west Grove, PA), and 500 fold diluted with rabbit horseradish IgG HRP (Cappel, West Chester, PA). used. Antigen was detected with ECL ™ detection reagents (Amersham Pharmacia Biothech) and expression levels were quantified by NIH Image. Polyclonal anti-FIR antibody has two synthetic peptides (C (correction) DKWKPPQGTDSIKME (a Minoic acid 30-45) and C (additional caro) EVYDQERFDNSDLSA (amino acids 528-542)) were immunized simultaneously.
[0059] Reverse transcriptase RT— Pし Rと Real— time quantitative Pし R  [0059] Reverse transcriptase RT— P and R and Real— time quantitative P and R
Total RNAと genomic DNAは癌部、非癌部から RNeasy™ Mini Kit and DNeasy™ Tis sues Kit (Qiagen)を用いて抽出した。 cDNAは total RNAから 1st strand cDNA Synthe sis Kitを用いて合成した(Roche, Mannheim, Germany)。 作製された cDNAを用いて RT-PCRにより FIR cDNAを増幅した。使用した primerは: forward 5,- GGCCCCATC AAGAGCATC -3 ' (配列番号 5)、 reverse 5,- GGGGCTGGGCCAGGGTCAG -3 ' ( 配列番号 6)である。コントロールとして GAPDH cDNAを用いた。 Total RNA and genomic DNA were extracted from cancerous and non-cancerous parts using RNeasy ™ Mini Kit and DNeasy ™ Tis sues Kit (Qiagen). cDNA was synthesized from total RNA using the 1 st strand cDNA Synthesis Kit (Roche, Mannheim, Germany). FIR cDNA was amplified by RT-PCR using the prepared cDNA. The primers used were: forward 5,-GGCCCCATC AAGAGCATC -3 '(SEQ ID NO: 5), reverse 5,-GGGGCTGGGCCAGGGTCAG -3' (SEQ ID NO: 6). GAPDH cDNA was used as a control.
同時に ReaH:ime quantitative PCRにより増幅された FIR cDNAを LightCycler™ (Ro che, Mannheim, Germany)により正確に定量した。 プライマーの至適条件の評価は Gene research laboratories Inc (Sendai, Japan)に依頼した。 FIR cDNA増幅の為の プフイマ ~~【ま (PCR product size is 275 base pairs): forward 5,— Gし ACCTGGAGTC ATCACA- 3,(配列番号 7) , reverse 5, -CGCAGAACCATCACTGTAG- 3 ' (配列番 号 8)を用いた。ヒト c-myc cDNAおよびヒト j8 - actin cDNAのプライマーはそれぞれ : forward 5, - GCCTCAGAGTGCATCGAC- 3,(配列番号 9) , reverse 5,- TCCACA GAAACAACATCG-3 ' (配列番号 10) (c-myc), forward 5 ' -TGGAGAAAATCTGG CACCAC-3 ' (配列番号 11) , reverse 5, - AATGGTGATGACCTGGCCGT- 3,(配 列番号 12) ( β -actin)を用いた。  At the same time, FIR cDNA amplified by ReaH: ime quantitative PCR was accurately quantified with LightCycler ™ (Roche, Mannheim, Germany). We asked Gene research laboratories Inc (Sendai, Japan) to evaluate the optimal primer conditions. PIR for amplification of FIR cDNA ~~ [MA (PCR product size is 275 base pairs): forward 5, — G and ACCTGGAGTC ATCACA-3, (SEQ ID NO: 7), reverse 5, -CGCAGAACCATCACTGTAG-3 '(SEQ ID NO: 8) was used. Primers for human c-myc cDNA and human j8-actin cDNA are: forward 5,-GCCTCAGAGTGCATCGAC-3, (SEQ ID NO: 9), reverse 5, -TCCACA GAAACAACATCG-3 '(SEQ ID NO: 10) (c-myc), forward 5 '-TGGAGAAAATCTGG CACCAC-3' (SEQ ID NO: 11), reverse 5, -AATGGTGATGACCTGGCCGT-3, (SEQ ID NO: 12) (β-actin) were used.
[0060] FIRイントロン 2の繰り返し配列の検出  [0060] Detection of repetitive sequence of FIR intron 2
FIRイントロン 2の繰り返し配列検出には以下のプライマーセットを用いた。  The following primer set was used for detecting the repeated sequence of FIR intron 2.
1F: 5 ' -ATCTCCTTCCTGCCTGCAGCAGGT-3 ' (配列番号 13)  1F: 5 '-ATCTCCTTCCTGCCTGCAGCAGGT-3' (SEQ ID NO: 13)
R04: 5 ' -AAGCCCATTCCTCCTTA-3 ' (配列番号 14)  R04: 5 '-AAGCCCATTCCTCCTTA-3' (SEQ ID NO: 14)
R05: 5 ' -TTTGACTTGCGCATTGC-3 ' (配列番号 15)  R05: 5 '-TTTGACTTGCGCATTGC-3' (SEQ ID NO: 15)
最初に IF- R04のプライマーセットを用いて PCRを行い、 1F-R05のプライマーセットで 2nd PCRを行った。この PCR産物を常法に従い直接 DNAシークェンスした。 PCR条件 はいずれも 95 °C;5min, [95°C;30 sec- 56°C; 30 sec- 72°C; 1 min]x35 cycles, 72。C; 2 minで行った。 DNAポリメラーゼは Pfo DNA polymerase (Stratagene, La Jolla, CA)を 使用した。 DNAシークェンスは IFプライマーを使用した。 First, PCR was performed using the IF-R04 primer set, and 2nd PCR was performed using the 1F-R05 primer set. This PCR product was directly subjected to DNA sequencing according to a conventional method. PCR conditions are 95 ° C; 5 min, [95 ° C; 30 sec-56 ° C; 30 sec-72 ° C; 1 min] x35 cycles, 72. C; performed at 2 min. DNA polymerase is Pfo DNA polymerase (Stratagene, La Jolla, CA) used. For the DNA sequence, an IF primer was used.
[0061] 結果(1) : [0061] Result (1):
我々は癌組織では転写抑制部位であるェクソン 2の欠失した変異 FIR (FIR Δ exon2) が特異的に発現していることを見出した(図 1及び図 2)。興味あることに、この変異 FI R (FIR Δ exon2)は内因性 c-Myc蛋白の発現を抑制せず(図 3 A)、逆に内因性 c-Myc 蛋白の発現を亢進させることが示された(図 3B、 C)。又、 FIR A exon2はアポトーシス を誘導することが出来ないことが確認された(図 4A, B)。更に、 FIRと FIR A exon2を 共発現させると、 c-Myc蛋白の発現を抑制しないのみならず、 FIRのアポトーシス誘導 活性が阻害されることも判明した(図 3D、図 4B)。  We found that a mutant FIR (FIR Δ exon2) lacking exon 2, which is a transcriptional repression site, was specifically expressed in cancer tissues (Figs. 1 and 2). Interestingly, this mutant FI R (FIR Δ exon2) has not been shown to suppress endogenous c-Myc protein expression (Figure 3A), but conversely, has been shown to enhance endogenous c-Myc protein expression. (Fig. 3B, C). It was also confirmed that FIR A exon2 was unable to induce apoptosis (Fig. 4A, B). Furthermore, co-expression of FIR and FIR A exon2 not only suppressed the expression of c-Myc protein, but also found that FIR apoptosis-inducing activity was inhibited (FIGS. 3D and 4B).
以上の結果力 癌では転写抑制部位であるェクソン 2を欠失した FIR Δ exon2が特異 的に発現することにより、 c-myc遺伝子の転写抑制が働かず、発現増大をもたらし、 アポトーシス誘導阻害、細胞増殖および癌化に結びつ!/、て 、るのではな!/、かと考え られた(図 5)。  As a result, in cancer, FIR Δexon2 lacking exon 2 which is a transcriptional repression site is specifically expressed, so that transcriptional repression of c-myc gene does not work, resulting in increased expression, inhibition of apoptosis induction, cell It was thought that this would lead to proliferation and canceration! /, So why not! / (Figure 5).
[0062] 結 (2): [0062] Yui (2):
FIRのスプライシングバリアントの発現メカニズムを解析する過程で、スプライシングを 起こすェクソン周囲のイントロンを調べたところ、先に述べたように 4塩基の繰り返し配 列 (MSI/SSR)が存在し、この MSI/SSRには極めて多くの多様性 (繰り返し数.塩基配列 の違 、)が認められた(図 6)。  In the process of analyzing the expression mechanism of the FIR splicing variant, we examined the intron around the exon that caused splicing, and as described above, there was a 4-base repetitive sequence (MSI / SSR). There was a great deal of diversity (number of repeats, difference in base sequence) (Fig. 6).
FIR遺伝子のイントロン 2内にある繰り返し配列は健常者末梢血単核急由来のゲノム DNAと大腸癌組織由来のゲノム DNAで有意にその繰り返し回数が異なつていた(図 7 、図 8)。即ち、健常者と較べて癌患者では 9回繰り返し配列 (ホモ)をもつ人の割合が 有意に多かった。  The repetitive sequences in intron 2 of the FIR gene were significantly different in the number of repeats between genomic DNA derived from peripheral blood mononuclear suddenly in healthy subjects and genomic DNA derived from colon cancer tissue (FIGS. 7 and 8). That is, the proportion of cancer patients with 9 repeat sequences (homo) was significantly higher in cancer patients than in healthy individuals.
この結果から、 FIR遺伝子のイントロン 2内に存在する繰り返し配列の回数を調べるこ とにより癌の検出'診断を行うことが出来ると考えられる。  From this result, it is considered that cancer detection can be diagnosed by examining the number of repetitive sequences present in intron 2 of the FIR gene.
[0063] [引用文献] [0063] [Cited document]
1. Avigan, M., ι·, Strober, Β·, and Levens, D. A far upstream element stimulates c— myc expression in differentiated leukemia cells. J. Biol. Chem., 265:18538—18545, 1 990. 2. Bazar, L., Meighen, D., Harris, V., Duncan, R., Levens, D., and Avigan, M. Tar geted melting and binding of a DNA regulatory element by a transactivator of c— myc • J. Biol. Chem., 270: 8241-8248, 1995. 1. Avigan, M., ι ·, Strober, Β ·, and Levens, D. A far upstream element stimulates c— myc expression in differentiated leukemia cells. J. Biol. Chem., 265: 18538—18545, 1 990. 2. Bazar, L., Meighen, D., Harris, V., Duncan, R., Levens, D., and Avigan, M. Tar geted melting and binding of a DNA regulatory element by a transactivator of c—myc • J. Biol. Chem., 270: 8241-8248, 1995.
3. Duncan, R., Bazar, L., Michelotti, G., Tomonaga, Τ·, Krutzsch, Η·, Avigan, M., and Levens, D. A sequence-specific, single-strand binding protein activates the far upstream element of c— myc and defines a new DNA— binding motif. Genes Dev., 8: 4 65-480, 1994.  3. Duncan, R., Bazar, L., Michelotti, G., Tomonaga, Τ ·, Krutzsch, Η ·, Avigan, M., and Levens, D. A sequence-specific, single-strand binding protein activates the far upstream element of c— myc and defines a new DNA— binding motif. Genes Dev., 8: 4 65-480, 1994.
4. Michelotti, G. A., Michelotti, E. F., Pullner, A., Duncan, R. C., Eick, D., and Le vens, D. Multiple single— stranaed cis elements are associated with activated chroma tin of the human c一 myc gene in vivo. Mol. Cell. Biol., 16: 2656 - 2669, 1996.  4. Michelotti, GA, Michelotti, EF, Pullner, A., Duncan, RC, Eick, D., and Le vens, D. Multiple single— stranaed cis elements are associated with activated chroma tin of the human c one myc gene in vivo. Mol. Cell. Biol., 16: 2656-2669, 1996.
5. Liu, J., He, L., Collins, I., Ge, Η·, Libutti, D., Li, J., Egly, J., and Levens, D. T he FBP Interacting repressor targets TFIIH to inhibit activated transcription. Mol. Cell, 5: 331-341, 2000.  5. Liu, J., He, L., Collins, I., Ge, Η, Libutti, D., Li, J., Egly, J., and Levens, D. The FBP Interacting repressor targets TFIIH to inhibit activated transcription. Mol. Cell, 5: 331-341, 2000.
6. Liu, J., Akoulitchev, S., Weber, A., Ge, Η·, Chuikov, S., Libutti, D., Wang, X. W., Conaway, J. W., Harris, C. C., Conaway, R. C., Reinberg, D., and Levens, D. Defective interplay of activators and repressors with TFIIH in xeroderma pigmentos um. Cell, 104: 353—363, 2001.  6. Liu, J., Akoulitchev, S., Weber, A., Ge, Η, Chuikov, S., Libutti, D., Wang, XW, Conaway, JW, Harris, CC, Conaway, RC, Reinberg, D., and Levens, D. Defective interplay of activators and repressors with TFIIH in xeroderma pigmentos um. Cell, 104: 353—363, 2001.
7. Tomonaga, Τ·, and Levens, D. Heterogeneous nuclear ribonucleoprotein K is a D NA- binding transactivator. J. Biol. Chem., 270: 4875 - 4881, 1995.  7. Tomonaga, Tsuji, and Levens, D. Heterogeneous nuclear ribonucleoprotein K is a D NA-binding transactivator. J. Biol. Chem., 270: 4875-4881, 1995.
8. Pelengaris, S., Khan, M., and Evan, G. I. Suppression of Myc— induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogeni c progression. Cell, 109: 321—334, 2002.  8. Pelengaris, S., Khan, M., and Evan, G. I. Suppression of Myc— induced apoptosis in beta cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell, 109: 321—334, 2002.
9. Leveillard, Τ·, Andera, L., Bissonnette, Ν·, Schaeffer, L., Bracco, L., Egly, J. M. , and Wasylyk, B. Functional interactions between p53 and the TFIIH complex are affected by tumor asiciated mutations. EMBO J., 15: 1615—1624, 1996.  9. Leveillard, Τ ·, Andera, L., Bissonnette, Ν ·, Schaeffer, L., Bracco, L., Egly, JM, and Wasylyk, B. Functional interactions between p53 and the TFIIH complex are affected by tumor asiciated mutations EMBO J., 15: 1615-1624, 1996.
10. Robles, A. I., Wang, X. W., Harris, C. C. Drug-induced apoptosis is delayed a nd reduced in XPD lymphoblastoid cell lines: possible role of TFIIH in p53— mediated apoptotic cell death. Oncogene, 18: 4681-4688, 1999. 11. Bazan, V., Migliavacca, M., Tubiolo, C, et al. Have p53 gene mutations and p rotein expression a different biological significance in colorectal cancer? J. Cell Phy siol, 191: 237-246, 2002. 産業上の利用可能性 10. Robles, AI, Wang, XW, Harris, CC Drug-induced apoptosis is delayed a nd reduced in XPD lymphoblastoid cell lines: possible role of TFIIH in p53— mediated apoptotic cell death. Oncogene, 18: 4681-4688, 1999. 11. Bazan, V., Migliavacca, M., Tubiolo, C, et al. Have p53 gene mutations and protein expression a different biological significance in colorectal cancer? J. Cell Phy siol, 191: 237-246, 2002. Availability on
[0064] 本発明は、癌、特に大腸癌に代表される消化器癌等のより有効で確実な検出 '診断' 治療を提供するに加え、癌の遺伝子診断システムと遺伝子治療臨床研究システムの 開発に利用される各種の生体分子を提供する。 [0064] The present invention provides a more effective and reliable detection 'diagnosis' treatment of cancer, particularly digestive organ cancer represented by colorectal cancer, as well as development of cancer gene diagnosis system and gene therapy clinical research system. Various biomolecules used in the field are provided.
[0065] 更に、 c-myc遺伝子転写抑制因子 FIRのスプライシングバリアントは、その発現を抑制[0065] Furthermore, the splicing variant of c-myc gene transcription repressor FIR represses its expression.
•低下させるような物質のスクリーニング法に使用することが可能であり、このような物 質は、アポトーシスを誘導し、癌を抑制阻害する作用があるものと考えられる。 • It can be used in screening methods for substances that can be reduced, and such substances are thought to have the effect of inducing apoptosis and suppressing and inhibiting cancer.

Claims

請求の範囲 [1] c— myc遺伝子の転写抑制因子である FIR(FBP Interacting Repressor)のェクソン 2 が欠失したスプライシングバリアントである蛋白質。 [2] 以下のアミノ酸配列を有する蛋白質: Claims [1] c—A protein that is a splicing variant in which exon 2 of FIR (FBP Interacting Repressor), which is a transcription repressor of the myc gene, is deleted. [2] Protein having the following amino acid sequence:
(1)配列番号 1に示されたアミノ酸配列から成る蛋白質、又は  (1) a protein comprising the amino acid sequence shown in SEQ ID NO: 1, or
(2)配列番号 1に示されるアミノ酸配列において、 1個若しく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、又は、配列番号 1に示されるアミノ酸配列と 相同性が 95%以上のアミノ酸配列からなり、且つ、配列番号 1に示されるアミノ酸配 列を有するポリペプチドと実質的に同等の活性を有する蛋白質。  (2) In the amino acid sequence shown in SEQ ID NO: 1, the amino acid sequence in which one or several amino acids are deleted, substituted or added, or homology with the amino acid sequence shown in SEQ ID NO: 1 is 95% or more And a protein having substantially the same activity as a polypeptide having the amino acid sequence shown in SEQ ID NO: 1.
[3] 請求項 1又は 2記載の蛋白質をコードする cDNA又はそれに対応する mRNA。  [3] cDNA encoding the protein according to claim 1 or 2, or mRNA corresponding thereto.
[4] 以下の塩基配列から成る cDNA又はそれに対応する mRNA: [4] cDNA consisting of the following base sequence or mRNA corresponding thereto:
(1)配列番号 2に示された塩基配列、又は  (1) the base sequence shown in SEQ ID NO: 2, or
(2)配列番号 2に示される塩基配列と相補的な塩基配列力 なる DNAとストリンジ ントな条件下でハイブリダィズし、且つ、請求項 1又は 2記載の蛋白質をコードする塩 基配列。  (2) A base sequence that hybridizes under stringent conditions with DNA complementary to the base sequence shown in SEQ ID NO: 2 and encodes the protein according to claim 1 or 2.
[5] FIR遺伝子のイントロン 2における 4塩基繰り返し配列における遺伝子多型を示す塩 基配列を含む連続した塩基配列から成る DNA。  [5] DNA consisting of a continuous base sequence including a base sequence indicating a gene polymorphism in the 4-base repeat sequence in intron 2 of the FIR gene.
[6] 配列番号 3における 118番目〜153番目の塩基配列に見られる 4塩基の 9回繰り返 し配列、又は、その領域における該 4塩基の 5回若しくは 6回繰り返し配列を含む、請 求項 5記載の DNA。 [6] The claim including the 9-repeat sequence of 4 bases found in the 118th to 153rd base sequences of SEQ ID NO: 3, or the 5 or 6 repeat sequences of the 4 bases in the region 5. DNA according to 5.
[7] 4塩基繰り返しの塩基配列が CCCG又は CCTGである、請求項 6記載の DNA。  [7] The DNA according to claim 6, wherein the base sequence of the 4-base repeat is CCCG or CCTG.
[8] 請求項 1若しくは 2記載の蛋白質又はその断片ペプチド、請求項 3又は 4記載の cD NA若しくはそれに対応する mRNA、又はそれらの一部カゝらなるオリゴヌクレオチド、 又は、請求項 5〜7の!ヽずれか一項記載の 4塩基繰り返し配列を含む連続した塩基 配列から成る DNAの存在を測定することから成る、癌の検出方法。  [8] The protein according to claim 1 or 2, or a fragment peptide thereof, the cDNA according to claim 3 or 4, mRNA corresponding thereto, or an oligonucleotide comprising a part thereof, or claims 5-7 A method for detecting cancer, comprising measuring the presence of DNA comprising a continuous base sequence comprising the 4-base repeat sequence according to claim 1.
[9] FIR又はそのスプライシングバリアントに特異的な抗体との抗原抗体反応を利用する 、請求項 8記載の検出方法。  [9] The detection method according to claim 8, wherein an antigen-antibody reaction with an antibody specific for FIR or a splicing variant thereof is used.
[10] ウェスタンプロット法により測定することを特徴とする、請求項 9記載の検出方法。 10. The detection method according to claim 9, wherein the measurement is performed by a Western plot method.
[11] 請求項 3又は 4記載の cDNA若しくは mRNA又はそれらの一部力 なるオリゴヌタレ ォチドを含む核酸分子を増幅する為のプライマーセットを使用する PCRを利用する、 請求項 8記載の検出方法。 [11] The detection method according to claim 8, wherein PCR is performed using a primer set for amplifying a nucleic acid molecule comprising the cDNA or mRNA according to claim 3 or 4, or an oligonucleotide which is a partial force thereof.
[12] RT— PCRである請求項 11記載の検出方法。  [12] The detection method according to claim 11, which is RT—PCR.
[13] リアルタイム定量的 PCRである請求項 11記載の検出方法。  [13] The detection method according to claim 11, which is real-time quantitative PCR.
[14] 請求項 5〜7の 、ずれか一項記載の DNAを含む核酸分子を増幅する為のプライマ 一セットを使用する PCRを利用する、請求項 8記載の検出方法。  [14] The detection method according to claim 8, which utilizes PCR using a set of primers for amplifying a nucleic acid molecule containing the DNA according to any one of claims 5 to 7.
[15] 癌が消ィ匕器癌である、請求項 8〜14のいずれか一項記載の検出方法。 [15] The detection method according to any one of claims 8 to 14, wherein the cancer is a cancer of the extinguisher.
[16] 癌が大腸癌である、請求項 8〜14のいずれか一項記載の検出方法。 [16] The detection method according to any one of [8] to [14], wherein the cancer is colorectal cancer.
[17] 請求項 8〜14のいずれか一項に記載の検出方法に使用する測定キット。 [17] A measurement kit used for the detection method according to any one of claims 8 to 14.
[18] FIR又はそのスプライシングバリアントに特異的な抗体、又は、請求項 3〜7のいずれ か一項記載の核酸分子を増幅する為のプライマーセットを要素として含む、請求項 1 7記載の測定キット。 [18] The measurement kit according to claim 17, comprising an antibody specific for FIR or a splicing variant thereof, or a primer set for amplifying the nucleic acid molecule according to any one of claims 3 to 7 as elements. .
PCT/JP2007/050915 2006-01-27 2007-01-22 METHOD OF DETECTING CANCER USING SPLICING VARIANT OF c-myc GENE TRANSCRIPTIONAL REGULATOR FIR OR FOUR-BASE REPETITIVE SEQUENCE IN INTRON 2 WO2007086342A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2007513551A JP4806776B2 (en) 2006-01-27 2007-01-22 Cancer detection method by splicing variant of c-myc gene transcription repressor FIR or 4-base repeat sequence in intron 2

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2006018341 2006-01-27
JP2006-018341 2006-01-27

Publications (1)

Publication Number Publication Date
WO2007086342A1 true WO2007086342A1 (en) 2007-08-02

Family

ID=38309135

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2007/050915 WO2007086342A1 (en) 2006-01-27 2007-01-22 METHOD OF DETECTING CANCER USING SPLICING VARIANT OF c-myc GENE TRANSCRIPTIONAL REGULATOR FIR OR FOUR-BASE REPETITIVE SEQUENCE IN INTRON 2

Country Status (2)

Country Link
JP (1) JP4806776B2 (en)
WO (1) WO2007086342A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011129427A1 (en) * 2010-04-16 2011-10-20 第一三共株式会社 Diagnostic agent and therapeutic agent for cancer
CN113226318A (en) * 2018-08-13 2021-08-06 北京智康博药肿瘤医学研究有限公司 Biomarkers for cancer therapy
US11753476B2 (en) 2018-04-08 2023-09-12 Cothera Bioscience, Inc. Combination therapy for cancers with BRAF mutation

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004018518A1 (en) * 2002-08-23 2004-03-04 Japan Science And Technology Agency Human solid cancer antigen peptides, polynucleotides encoding the same and utilization thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004018518A1 (en) * 2002-08-23 2004-03-04 Japan Science And Technology Agency Human solid cancer antigen peptides, polynucleotides encoding the same and utilization thereof
WO2004018679A1 (en) * 2002-08-23 2004-03-04 Japan Science And Technology Agency Method and kit for diagnosing cancer

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK [online] OTTENWAELDER B. ET AL., XP003017947, Database accession no. (CR857074) *
DATABASE GENBANK [online] OTTENWAELDER B., XP003017946, Database accession no. (CAH89379) *
KAJIWARA T. ET AL.: "Massho Kecchu deno c-myc Tensha Yokusei Inshi FIR splicing variant no Kenshutsu ni yoru Daichogan Shindan", DAI 65 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 28 August 2006 (2006-08-28), pages 392, P-1005, XP003017952 *
MATSUSHIT K. ET AL.: "An essential role of alternative splicing of c-myc suppressor FUSE-binding protein-interacting repressor in carcinogenesis", CANCER RES., vol. 66, no. 3, 2006, pages 1409 - 1417, XP003017953 *
MATSUSHITA K. ET AL.: "Apoptosis Yudosei c-myc Idenshi Tensha Yokusei Inshi ni yoru Shokaki Gan Idenshi Chiryoho no Kaihatsu", DAI 63 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 25 August 2004 (2004-08-25), pages 516, P-1494, XP003017950 *
MATSUSHITA K. ET AL.: "c-myc Idenshi Tensha Yokusei Inshi FIR no Gan Tokuiteki Screening Splicing Variant o Mochiita Gan Shindan", THE JAPANESE SOCIETY OF GASTROENTEROLOGICAL SURGERY ZASSHI, 1 July 2006 (2006-07-01), pages 449, 1187, XP003017951 *
MATSUSHITA K. ET AL.: "c-myc Idenshi Tensha Yokusei Inshi FIR no Hatsugen to Ganka eno Kanyo", DAI 28 KAI ANNUAL MEETING OF THE MOLECULAR BIOLOGY SOCIETY OF JAPAN KOEN YOSHISHU, 25 November 2005 (2005-11-25), pages 426, 2P-0467, XP003017949 *
MATSUSHITA K. ET AL.: "c-myc Idenshi Tensha Yokusei Inshi FIR no Splicing Variant no Ganka eno Kanyo", DAI 64 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION KIJI, 15 August 2005 (2005-08-15), pages 459, PA3-1042, XP003017948 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011129427A1 (en) * 2010-04-16 2011-10-20 第一三共株式会社 Diagnostic agent and therapeutic agent for cancer
US11753476B2 (en) 2018-04-08 2023-09-12 Cothera Bioscience, Inc. Combination therapy for cancers with BRAF mutation
CN113226318A (en) * 2018-08-13 2021-08-06 北京智康博药肿瘤医学研究有限公司 Biomarkers for cancer therapy

Also Published As

Publication number Publication date
JP4806776B2 (en) 2011-11-02
JPWO2007086342A1 (en) 2009-06-18

Similar Documents

Publication Publication Date Title
Sumegi et al. Recurrent t (2; 2) and t (2; 8) translocations in rhabdomyosarcoma without the canonical PAX‐FOXO1 fuse PAX3 to members of the nuclear receptor transcriptional coactivator family
US20130102542A1 (en) Cancer related isoforms of components of transcription factor complexes as biomarkers and drug targets
Gire et al. The Gly56Arg mutation in NR2E3 accounts for 1–2% of autosomal dominant retinitis pigmentosa
WO2017044801A2 (en) Methods of identifying drug-modulated polypeptide targets for degradation
WO2007086342A1 (en) METHOD OF DETECTING CANCER USING SPLICING VARIANT OF c-myc GENE TRANSCRIPTIONAL REGULATOR FIR OR FOUR-BASE REPETITIVE SEQUENCE IN INTRON 2
Szentirmay et al. The IGF2 receptor is a USF2-specific target in nontumorigenic mammary epithelial cells but not in breast cancer cells
US20030171273A1 (en) Novel transcription factor, BP1
US7666603B2 (en) Breast cancer related protein, gene encoding the same, and method of diagnosing breast cancer using the protein and gene
CA2803215A1 (en) Compositions and methods related to prostate cancer
Xing et al. Characterization of the promoter of 1A6/DRIM, a novel cancer-related gene and identification of its transcriptional activator
Morozov et al. HPV 16 E7 oncoprotein induces expression of a 110 kDa heat shock protein
US20040197786A1 (en) Method of examining steroid resnponsiveness
WO2015194524A1 (en) B-precursor acute lymphoblastic leukemia novel chimeric gene
Kokura et al. Identity between rat htf and human xbp-1 genes: determination of gene structure, target sequence, and transcription promotion function for HTF
JP4617257B2 (en) Human cancer-related genes, products encoded by them and applications
WO2000043522A1 (en) Gene causative of rothmund-thomson syndrome and gene product
JP4098236B2 (en) Nucleic acids with differential expression between hepatoblastoma and normal liver
KR20070027659A (en) A breast cancer related protein, a gene encoding the same, and a method for diagnosing a breast cancer using the protein and gene
CA2491420A1 (en) Rb1 gene induced protein (rb1cc1) and gene
RU2380422C2 (en) Analysis and application of polymorphous for estimating risk of cardiovascular diseases
WO1998006871A1 (en) Materials and methods relating to the diagnosis and prophylactic and therapeutic treatment of papillary renal cell carcinoma
JP4830101B2 (en) Costello syndrome-causing gene mutation and detection or diagnosis method thereof
JP5354484B2 (en) Cancer detection method
WO1997001634A2 (en) Polypeptide for repairing genetic information, nucleotidic sequence which codes for it and process for the preparation thereof (guanine thymine binding protein - gtbp)
JP2008194045A (en) Nucleic acid upregulated in human tumor cells, protein encoded thereby and process for tumor diagnosis

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 2007513551

Country of ref document: JP

DPE2 Request for preliminary examination filed before 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: DE

122 Ep: pct application non-entry in european phase

Ref document number: 07707178

Country of ref document: EP

Kind code of ref document: A1