WO2008044787A1 - Composition for treating cancer using dref as molecular target - Google Patents
Composition for treating cancer using dref as molecular target Download PDFInfo
- Publication number
- WO2008044787A1 WO2008044787A1 PCT/JP2007/070031 JP2007070031W WO2008044787A1 WO 2008044787 A1 WO2008044787 A1 WO 2008044787A1 JP 2007070031 W JP2007070031 W JP 2007070031W WO 2008044787 A1 WO2008044787 A1 WO 2008044787A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- dref
- seq
- cancer
- gene
- cell
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/30—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/5005—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
- G01N33/5008—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
- G01N33/5011—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
Definitions
- the present invention relates to a composition for cancer treatment that targets the human cell growth regulator DREF or DREF gene as a molecular target. 'Ming
- DREF is a Drosophila transcription factor isolated by Hirose et al., which recognizes and binds to a DNA sequence called DRE-mot if in one sequence of a gene promoter and induces the expression of that gene (non-patent literature). 1).
- Drosophila it is reported that PCNA (prol iferating cell nuclear antigen) transcription is induced in the S phase, and that several gene forces related to cell proliferation are reported to be regulated by DREF. Yes.
- a human homologue of Drosophila DREF was cloned and sequenced by Ohshima et al., And it was reported that human DREF was involved in the induction of histone HI gene (Non-patent Document 2). At this time, the expression of the human DREF gene is reduced by the two siRNAs generated against the DREF CR1 domain, thereby suppressing the histone HI gene expression (Non-patent Document 2).
- the human DREF gene is a gene that was found by Esposito et al. (Non-patent Document 3) from a cDNA library derived from a human non-inducible male teratocarcinoma cell line Tramp (Ac-like transposable element; putative Ac-like transposable element; Chromosome: 'X, Y; Location: Xp22.33; Ypll).
- DREF is one of the factors involved in cell cycle progression by knocking down the human DREF gene, and that expression of liposomal protein (RP) is reduced by knockdown. It was pointed out that it plays a role of stimulating cell proliferation through gene regulation (Non-patent Documents 4 and 5). - So far, human DREF has been suggested to be associated with cell proliferation ability S, but no detailed functional analysis has been performed in relation to cancer therapy.
- Non-Patent Document 4 Sano, Y. et al., Abstracts of the Molecular Biology Society of Japan (issued on November 25, 2005), lecture number 1P-0873, 2005
- Refractory cancers such as lung cancer and esophageal cancer are resistant to general chemotherapy, and effective means that can be expected to cure other than surgery have not been established. For this purpose, it is necessary to develop molecular targeted therapeutics targeting genes involved in cancer onset, propagation and metastasis, and to establish treatments aimed at cure of cancer.
- An object of the present invention is to provide a therapeutic composition for cancer.
- Another object of the present invention is to provide a method of screening for a therapeutic agent for cancer.
- the present inventors have found that human DREF is a target for cancer treatment, and have completed the present invention. .
- DREF was discovered as a transcription factor of Drosophila, and it was later shown that its homolog exists in humans.
- the present inventors have now found that human DREF is not substantially expressed in normal cells, but is significantly expressed in cancer cells, and based on this, DREF is predicted to be a molecular target for cancer treatment. It was demonstrated that suppression of DREF expression actually leads to cancer suppression and cell death. Therefore, the present invention has the following features.
- the present invention provides an RNAi nucleus against DREF, which suppresses the in vivo function of human cell growth control factor DREF or DREF gene in cancer cells or cancer tissues.
- a cancer therapeutic composition comprising an acid, an antisense nucleic acid, or an antibody.
- DREF is an abbreviation for DRE (DNA replication-related element) -binding factor named by Hirose et al., Also known as ZBEDl (zinc finger, BED-type containing 1).
- ZBEDl zinc finger, BED-type containing 1
- DREF human DREF
- DREF is a protein comprising the amino acid sequence shown in SEQ ID NO: 1, or an amino acid sequence comprising a deletion, substitution or addition of one or several amino acids in the amino acid sequence.
- DREF gene the open reading frame (0RF) nucleotide sequence encoded by an amino acid sequence of SEQ ID NO: 1, nucleotide sequence and more specifically shown in position 202 to 2 28 6 of SEQ ID NO: 2 Including. Both amino acid and nucleotide sequences of DREF are registered with GenBank as Accession number NMJ304729.
- DREF in the present invention includes an amino acid sequence having 95% or more, preferably 97% or more, more preferably 98% or more, and most preferably 99% or more identity with the amino acid sequence shown in SEQ ID NO: 1.
- Variants having in vivo functions equivalent to DREF for example, polymorphic variants, splice variants, degenerate variants, mutants, etc.
- the DREF gene in the present invention is SEQ ID NO: nucleotide sequence shown in 2 (position 202 to 228 6th) 95% or more, preferably 97% or more, more preferably 98% or more, including the most favorable 'Mashiku nucleotide sequence having 99% or more identity And variants similar to those described above having an in vivo function equivalent to DREF after translation.
- the DREF gene is used with the intention of encompassing not only exons, but also introns, 3 'untranslated regions and 5' untranslated regions. '
- the suppression of in vivo function is an effect of suppressing cell proliferation, cell cycle arrest and / or cell death in cancer cells.
- in vivo function refers to the action of DREF in cancer cells, such as cell proliferation, cell cycle progression, and the like.
- RNAi nucleic acid or antisense nucleic acid is capable of cleaving DREF raRNA or suppressing its function.
- RNAi is so-called RNA interference, and is a gene having a sequence homologous to DNA or double-stranded RNA (dsRNA) added from the outside. (For example, Chisato Ushida, Protein Nucleic Acid Enzyme Vol. 46 (No. 10), pp. 1381-1386, 2001).
- the target RNA is mRNA or pre-mRNA encoded by the gene, and the target mRNA is degraded or suppressed in expression by RNAi.
- Other RNAi nucleic acids that can be used in the present invention also include miRNA (microRNA). miRNA binds to DREF mRNA 3, the untranslated region and suppresses DREF gene expression. The size of these RNAi nucleic acids is 19-30 nucleotides long, preferably 19-25 nucleotides long, more preferably 19-23 nucleotides long.
- the “antisense nucleic acid” used in the present invention includes an RNA sequence complementary to the whole or a part of DREF mRNA, DNA encoding the RNA sequence, or a vector containing the DNA sequence. .
- vector refers to a regulatory sequence such as a promoter, poly T sequence, replication origin, terminator, etc., in addition to the DNA encoding the target siRNA or shRNA or DNA encoding the antisense RNA. , Selection markers, etc. can be included.
- the siRNA or shRNA, or antisense RNA is produced in the cell by the expression of the DNA.
- the RNAi nucleic acid or antisense nucleic acid is derived from a nucleotide sequence encoding the CR2 domain or CR3 domain of DREF (SEQ ID NOs: 3 and 4, respectively).
- a preferable nucleotide sequence in the present invention is, for example, in the sequence of SEQ ID NO: 2 of the DREF gene, from position 1,300 to 1,500 (from position 1099 when “A” of the start codon ATG of 0RF is the first position) I 2 "position), 1,900 to 2,000 position (positions 1699 to 1799 when 0RF start codon ATG" A "is first position).
- CR refers to a highly conserved region in the sequence comparison between human DREF and Drosophila DREF. Such areas (Amino acid sequence; nucleotide sequence) is CR1 (positions 4 to 140 of SEQ ID NO: 1; positions 21 to 621 of SEQ ID NO: 2), CR2 (positions 377 to 506 of SEQ ID NO: 1; 1330 to 1719 of SEQ ID NO: 2) ) And CR3 (positions 541 to 688 of SEQ ID NO: 1; positions 1822 to 2265 of SEQ ID NO: 2), and the identities are 27.7%, 29.2%, and 21.1%, respectively (Ohshima, N. et al., J. Biol. Chem. 278: 22928-38, 2003).
- RNAi nucleic acids in the present invention include any one of the DNA sequences of SEQ ID NOs: 5 to 15, 19, 22, 25 and 28 or an RNA sequence corresponding to the DNA sequence.
- DNA represents deoxyribonucleic acid
- RNA represents ribonucleic acid.
- a more preferable vector in the present invention includes the DNA sequence of any one of SEQ ID NOs: 20, 21, 23, 24, 26, 27, 29 and 30.
- the composition of the present invention can comprise an antibody against DREF that inhibits the in vivo function of DREF.
- Any antibody can be used in the present invention as long as such a function can be suppressed.
- Preferred antibodies are DREF CR1 domain, CR2 domain or CR3 domain (SEQ ID NO: 138, 133 or 134, respectively), or a functional domain related to DNA binding of DREF in the CR1 or CR3 domain (eg, SEQ ID NO: 140 or 141), It is an antibody that binds to the epitope.
- the term epitope refers to a site to which an antibody on an antigen molecule binds, and usually the number of amino acids in such a site is 8 or more, preferably 10 or more. It may consist of a target sequence or a discontinuous sequence. Therefore, the antibody of the present invention can immunologically bind to an epitope composed of 8 or more, preferably 10 or more amino acids in the amino acid sequence of DREF.
- 'Preferred antibodies against DREF that can be used in the present invention are human antibodies, humanized antibodies or fragments thereof.
- the present invention further includes adding a candidate drug to a medium containing human cultured cancer cells, and screening a drug that suppresses the expression or translation of the DREF gene in vitro.
- a method for screening is provided.
- Cancer cells that can be used in the present invention are cancer cells from cancer cell lines, biopsy. including.
- the latter cancer cells are derived from a patient and can be used to search for an effective drug for the patient.
- the type of cancer cell is not particularly limited, but cancer cells in which DREF is expressed in cancer cells but hardly or not in normal cells are desirable.
- Candidate drugs that can be used in the present invention may be any of small molecules to macromolecules, and include low molecular organic compounds, peptides, proteins, oligosaccharides, polysaccharides, lipids, and the like.
- screening can be performed using as an index the suppression of cancer cell growth, cell cycle arrest and / or cell death induction. Such an effect is not substantially observed in normal tissues or cells because DREF is hardly or not expressed in normal tissues or cells.
- Figure 1 shows the domain structure of the human DREF / ZBED1 gene (Xp22.33 & Ypll), and the RNAi site and sequence.
- Figure 2 shows the results of RT-PCR analysis of the RNAi effect.
- Figure 3 shows the results of Western blot analysis of the RNAi effect.
- Figure 4 shows the cell cycle analysis results.
- Figure 5A shows the cell cycle analysis results.
- Figure 5B shows the cell cycle analysis results.
- Figure 5C shows the cell cycle analysis results.
- FIG. 6 A shows the results of cell proliferation ability analysis.
- B shows the results of cell proliferation ability analysis.
- FIG. 7 shows the results of immunostaining for 7 H2AX (Phospho-H2AX) and Phospho-ATM (Serl981).
- Figure 8 shows the TUNEL analysis results.
- FIG. 9 shows the results of cell death and cell cycle analysis.
- FIG. 10 shows cell death (mitotic catastroph) in M phase.
- Figure 11 shows the gene expression analysis results of siDREF-, # 4 in ACC-LC-91. 1 2, O showing expression of DREF in lung cancer patient specimens
- Figure 13 shows the DNA sequence of the RPL17 gene (A; SEQ ID NO: 137), which is thought to be associated with cancer growth, among the genes that are suppressed by DREF-RNAifH, and the transcriptional activation of the RPL17 gene by DREF.
- Mechanism (B) is shown.
- the present invention includes an RNAi nucleic acid, an antisense nucleic acid or an antibody against DREF, which suppresses the in vivo function of a human cell growth regulator DREF or DREF gene in cancer cells or cancer tissues.
- a composition is provided.
- DREF is expressed in cancer cells or tissues, but is not substantially (or little or not) expressed in normal cells or tissues.
- the present inventors suppressed cancer cell growth, cell cycle arrest, cell death induction by suppressing (including blocking or inhibiting) DREF gene expression or protein translation in cancer cells or tissues. It was found that such an event occurred. ⁇
- Cancers to be treated by the present invention are not limited to cancers capable of expressing DREF, such as lung cancer, esophageal cancer, knee cancer, stomach cancer, liver cancer, colon cancer, thyroid cancer, prostate cancer, Includes bladder cancer, kidney cancer, skin cancer, gall cancer, brain cancer, breast cancer, ovarian cancer, cervical cancer, testicular cancer, lymphoma, melanoma, sarcoma, osteosarcoma, etc.
- Preferred cancers are lung cancer, esophageal cancer, breast cancer, knee cancer and the like. ',
- the amino acid sequence of DREF which is the target of the present invention, and the nucleotide sequence of the DREF gene are known, for example, as SEQ ID NO: 1 and SEQ ID NO: 2 (open reading frame (0RF): positions 202 to 228 ⁇ ), respectively. Both DREF amino acid and nucleotide sequences are registered in GenBank as Accession number ⁇ _004729.
- the present invention also includes suppressing the in vivo function of a DREF or a mutant of the DREF gene, for example, a mutant based on mutation, polymorphism, alternative splicing, degeneracy and the like. ⁇
- DREF in the present invention is a protein comprising the amino acid sequence shown in SEQ ID NO: 1, or an amino acid sequence containing one or several amino acid deletions, substitutions or additions in the amino acid sequence.
- DREF in the present invention is In vivo function equivalent to DREF, comprising an amino acid sequence having 95% or more, preferably 97% or more, more preferably 98% or more, most preferably 99% or more identity with the amino acid sequence shown in SEQ ID NO: 1. The variant which has is also included.
- the DREF gene in the present invention corresponds to the nucleotide sequence encoded by the amino acid sequence of SEQ ID NO: 1, more specifically the nucleotide sequence shown in SEQ ID NO: 2, particularly DREF 0RF at positions 202 to 2286. Contains nucleotide sequence.
- the DREF gene in the present invention is a DNA further comprising a nucleotide sequence containing a deletion, substitution or addition of one or several nucleotides in the nucleotide sequence shown at positions 202 to 2286 of SEQ ID NO: 2.
- the DREF gene in the present invention has 95% or more, preferably 97% or more, more preferably 98% or more, and most preferably 99% or more identity with the nucleotide sequence shown in positions 202 to 2286 of SEQ ID NO: 2. And a variant having an in vivo function equivalent to DREF after translation. '
- severe means an integer of about 10 or less, such as 9, 8, 7, 6, 5, 4, 3, 2.
- DREF has in vivo functions such as cell proliferation and cell cycle progression, but the present invention makes it possible to suppress the in vivo functions of DREF in cancer and lead to cancer degeneration and cell death.
- examples of the agent that suppresses the expression or protein translation of the target DREF gene include RNAi nucleic acid, antisense nucleic acid and antibody against DREF. Each of these drugs will be described more specifically below.
- RNAi nucleic acid usable in the present invention is a continuous 19 to 30 nucleotide sequence derived from the nucleotide sequence of the DREF gene (consisting of exons, introns, 3 ′ untranslated region and 5 ′ untranslated region) or the nucleotide sequence of DREF mRNA. It is a DNA or RNA containing nucleotides, preferably 19-25 nucleotides, more preferably 19-23 nucleotides.
- RNAi methods that can be used in the present invention include 1) direct introduction of short double-stranded RNA (siRNA) into cells, or 2) incorporation of short-hairpin RNA (shRNA) into various expression vectors, Introduce the vector into the cell, or 3) Insert a short double-stranded DNA corresponding to siRNA between the promoters into a vector with two promoters aligned in the opposite direction to express s iRNA. This includes methods such as preparation and introduction into cells.
- siRNA short double-stranded RNA
- shRNA short-hairpin RNA
- the RNAi nucleic acid includes s iRNA, shRNA or miRNA that enables cleavage of DREF mRNA or suppression of its function.
- the RNAi nucleic acid may also be a DNA encoding these RNAs or a vector containing the DNA, which, when used in vivo, makes it possible to produce siRNA, shRNA or miRNA.
- RNAi nucleic acid is a continuous 19 to 30 nucleotides, preferably 19 to 25 nucleotides derived from nucleotide sequences (SEQ ID NOs: 3 and 4, respectively) encoding the CR2 domain or CR3 domain of DREF.
- Further preferred CR2 domain or CR3 domain target sites are, for example, positions 1,300 to 1,500 in the sequence of SEQ ID NO: 2 of the DREF gene (0RF open-start codon ATG “A” is the first Position 1099 to 1299), 1,900 to 2,000 (0RF start codon ATG
- RNAi nucleic acid that can be used in the present invention is, for example, the DNA sequence of any of the following SEQ ID NOs: 5 to 15, 19, 22, 25, and 28, or the RNA sequence corresponding to the DM sequence (that is, the DNA sequence) Array with T in U replaced by U).
- the number after the sequence is the position of the start site on 0RF of DREF (however, the start codon of 0RF
- CAACCACCACCTCATGCTGGAG 1139 (SEQ ID NO: 7)
- Particularly preferred sequences are SEQ ID NO: 19 (RNAi # 3 (positions 1136 to 1156)), SEQ ID NO: 22 (RNAi # 4 (positions 1718 to 1738) :), SEQ ID NO: 25 (RNAi # 5 (positions 1245 to 1265)) ), And SEQ ID NO: 28 (RNAi # 6 (positions 1737 to 1757)), or an RNA sequence corresponding to the DNA sequence (that is, a sequence in which T in the DNA sequence is replaced with U).
- the number range shown in parentheses together with the SEQ ID No. is when the base number “A” (position 2 ) of the start codon ATG of DREF 0RF is numbered as the first in the nucleotide sequence of SEQ ID No. 2. Represents the position of. ⁇
- RNAi nucleic acid for DREF is the DNA sequence of SEQ ID NO: 33-132 below, or the RNA sequence corresponding to that DNA sequence (ie, a sequence in which T in the DNA sequence is replaced with U).
- CAAGAGCAAGGTGTGGAAGTATT 63 (SEQ ID NO: 33)
- CAGATCGCCTACTCCGGAA 157 (SEQ ID NO: 40)
- AGATCGCCTACTCCGGAAACA 158 (SEQ ID NO: 41)
- GTCCTACCACCTGGAGAAGAAC 188 (SEQ ID NO: 2)
- CTGCGAGTTCGTCAAGAGCAAC 224 (SEQ ID NO: 44) GCGAGTTCGTCAAGAGCAA 227 (SEQ ID NO: 45) CGTGCTGGGCCTCATCTGCGAG 386 (SEQ ID NO: 46) CCCAGCCTCCATCGTGGACGAG 416 (SEQ ID NO: 47) CAGCCTCCATCGTGGACGA 419 (SEQ ID NO: 48) CACCTTCAAGGTGCTGCTGAAG 440 (SEQ ID NO: 49) CCTTCAAGGTGCTGGA 491 (SEQ ID NO: 52) CTCTACCAAGGCCATCCCTGAG 503 (SEQ ID NO: 53) CGTCCGGGAGGTGATCCTGAAG 536 (SEQ ID NO: 54) TCCGGGAGGTGATCCTGAA 538 (SEQ ID NO: 55) CCGGGAGGTGATCCTGAAGGAG 539 (SEQ ID NO: 56) GGGAGGTGATCCTGAAGGA GT GG (GT ACC GG (SEQ ID NO:
- GCTCTATGAGGTCTTCATCGAG 749 (SEQ ID NO: 68) TTCGGGGCCACCACCAACTATGG 793 (SEQ ID NO: 69) CACCAACTATGGCAAGGACATC 803 (SEQ ID NO: 70) CCAACTATGGCAAGGACAT 806 (SEQ ID NO: 71) AGGACATCGTGAAGGCGTGCT 818 (SEQ ID NO: 72) CTGCCGCAAACTGGTGGAGTAC 941 (SEQ ID NO: 73) GCCGCAAACTGGTGGAGTACT 944 (SEQ ID NO: 74)
- CAAGGAGCTTTCCAAGACCTA 1356 (SEQ ID NO: 94)
- ACGCCCGAGATCGACATGTTTCT 1384 (SEQ ID NO: 96)
- CAGAAGTACTGGTGCGTGA 1816 (SEQ ID NO: 117
- GGACGAGCAGGTGTTTCTGTAT 1922 (SEQ ID NO: 119
- the RNAi nucleic acid is a continuous 19-30 nucleotides, preferably 19-25, from the sequence of the mRNA or variant thereof encoded by the DRSF gene comprising the nucleotide sequence of SEQ ID NO: 2.
- An siRNA comprising a nucleotide, more preferably a 19-23 nucleotide sense strand sequence and its complementary sequence, the antisense strand sequence.
- the siRNA can suppress the expression of the DREF gene or mRNA in vivo or in vitro.
- a preferred example of the corresponding DNA sequence comprising the siRNA is a contiguous 19-30 nucleotide, preferably 19-25 nucleotides derived from the nucleotide sequence encoding the CR2 domain or CR3 domain of DREF (SEQ ID NOs: 3 and 4, respectively).
- the DNA sequence comprising 19 to 23 nucleotides more preferably, the nucleotide sequence of SEQ ID NO: 2, positions 1,300 to 1,500 (0RF starting codon ATG “A” is the first position 1999 to 1299), 1,900 to 2,000 (from 1699 to 1799 if the 0RF start codon ATG “A” is the first), 19-30 DNA sequences comprising nucleotides, preferably 19-25 nucleotides, more preferably 19-23 nucleotides, more preferred examples are shown by SEQ ID NOs: 5-15, 19, .22, 25, 28, 33-132 DNA sequence.
- the sequence of the DREF RNAi nucleic acid is not limited to the above sequence, and for example, in the nucleotide sequence of SEQ ID NO: 2 as long as it suppresses the in vivo function of the DREF gene, the corresponding mRNA, or a variant thereof.
- the sequence may be different from the sequence of, for example, 1 to 3 bases. '
- the siRNA of the present invention can be synthesized using a well-known chemical synthesis technique based on the DREF sequence that is the target of siRNA as exemplified above.
- it can be synthesized chemically using an automated DNA (/ RNA) synthesizer using DNA synthesis technology such as solid phase phosphoamidite method, or a siRNA-related contract synthesis company (for example, Funakoshi Corporation) It is also possible to synthesize by outsourcing to companies such as Dharmacon and Ambion.
- the siRNA of the present invention is a duplex that is a precursor thereof. It can also be derived from RNA (shRNA) via processing by the intracellular RNase Dicer.
- shRNA is a double-stranded RNA having a loop between the sense strand sequence and the antisense strand sequence of siRNA, preferably from 1 to 6, preferably 2 to 4 poly U at its 3 ′ end. Including overhangs.
- shRNA is processed into siRNA by a dicer belonging to the RNase III family, and then siRNA is made into a single strand, and its antisense strand RNA is complexed with a molecule having RNA cleavage activity, etc.
- RISC RNA- Induced Silencing Complex
- both the above siRNA of the present invention and its precursor shRNA can be used as an active ingredient of the composition of the present invention.
- the selected RNAi nucleic acid does not exhibit a so-called off-target action in clinical use.
- Off-target action refers to the action of suppressing the expression of another gene that is partially homologous to the RNAi nucleic acid used in addition to the target m gene.
- the candidate RNAi nucleic acid has the ability to confirm that there is no cross-reaction using a gene chip in advance, or there is no sequence identity using a database such as GenBank (NCBI) Or it is necessary to confirm that there is little.
- GenBank NCBI
- human DREF is not substantially expressed in normal cells in humans, but is significantly expressed in cancer cells, so it is expected that the problem of aftergetting can be avoided.
- RNAi nucleic acid of the present invention When introducing the RNAi nucleic acid of the present invention into a patient's body, it is preferable to use a vector capable of directly injecting the nucleic acid into the affected area or capable of expressing the nucleic acid.
- siRNA or vectors can be complexed with ribosomes such as lipofectamine, lipofectin, selfectin, other positively charged ribosomes (eg, positively charged cholesterol), or microcapsules, and this complex can be used.
- ribosomes such as lipofectamine, lipofectin, selfectin, other positively charged ribosomes (eg, positively charged cholesterol), or microcapsules
- the cell membrane of mammals is negatively charged, positively charged ribosomes are preferably used.
- the positively charged ribosome-nucleic acid complex is taken up into the cell by engineered cytosis.
- the nucleic acid can be further transferred to the cytoplasm or nucleus.
- the therapeutic RNAi nucleic acid of the present invention can be encapsulated in nanoparticles having a particle size of about 500 nm or less.
- nanoparticles include hollow nanoparticles formed from, for example, hepatitis B virus envelope L particles, and nucleic acids are encapsulated in the particles by electroporation. This nucleic acid-encapsulated particle can be delivered to the liver when administered into the blood (T. Yamada et al., Nature Biotech 21 (8): 885-890, 2003) and may be useful in the treatment of liver cancer.
- the nucleic acid when encapsulating the therapeutic RNAi nucleic acid of the present invention in a liposome, the nucleic acid is treated with protamine sulfate to cause condensation to form a nucleic acid-protein complex, which is then encapsulated in a positively charged lipid or polymer micelle.
- protamine sulfate when encapsulating the therapeutic RNAi nucleic acid of the present invention in a liposome, the nucleic acid is treated with protamine sulfate to cause condensation to form a nucleic acid-protein complex, which is then encapsulated in a positively charged lipid or polymer micelle.
- Liposome-nucleic acid complexes can be obtained, for example, by the reverse phase evaporation method (F: Szoka et al., Biochim. Biophys. Acta, 601: 559, 1980), Bonoretex shaking method, Calcium fusion-EDTA chelate method (Yasufumi Kaneda, Experimental Medicine 22 ⁇ No. 14 (extra number), pages 14-152, 2004).
- F reverse phase evaporation method
- Bonoretex shaking method Bonoretex shaking method
- Calcium fusion-EDTA chelate method Yasufumi Kaneda, Experimental Medicine 22 ⁇ No. 14 (extra number), pages 14-152, 2004.
- the microinjection method is a method in which a nucleic acid is directly microinjected into a cell with a fine injection needle.
- the viral vector method is a method of introducing a nucleic acid into a cell using a viral vector, in which a recombinant virus incorporating an expression unit that expresses siRNA, shRNA or miRNA is prepared, and the cell is infected. It is a method of expressing in cells. Examples of viruses include adenovirus, adeno-associated virus, retrovirus and the like.
- a nucleic acid that can be used in the present invention includes an expression vector comprising a DNA sequence encoding an RNAi nucleic acid such as siRNA or shRNA under the control of a promoter.
- an expression vector is a hairpin vector.
- This vector includes DNA encoding a hairpin RNA in which the sense strand RNA sequence and the antisense strand RNA sequence are covalently bonded via a single-stranded loop sequence, wherein the DNA is intracellularly contained.
- the hairpin RNA is formed by transcription and processed by a dicer.
- a vector that forms the siRNA is formed by transcription and processed by a dicer.
- Short hairpin RNA as a siRNA precursor transcribed from vector DNA should have an overhang consisting of 3 to 4 U at the end of its antisense strand.
- sense strand RNA and antisense strand RNA can be more stable against nuclease degradation.
- dicer There is one endogenous dicer in humans, which has the role of converting long dsRNA and precursor miRNA into siRNA and mature miRNA, respectively.
- loop sequence according to the present invention examples include 5'-UUCAAGAGA-3 '(SEQ ID NO: 135), 5'-CUUCCUGUCA-3' (SEQ ID NO: 136), 5'-UUCCAG-3 '(SEQ ID NO: 137), etc. However, it is not limited to these.
- RNAi # 3, RNAi # 4, RNAi # 5 and RNAi # 6, which are particularly preferred among the RNAi nucleic acids of the present invention, are shown in SEQ ID NOs: 20 and 21, respectively.
- the underline indicates the sequence corresponding to the sense strand and antisense strand of siRNA.
- RNAi # 3 oligo SEQ ID NOS: 20 and 21, respectively: '
- RNAi # 4 oligo SEQ ID NOS: 23 and 24, respectively:
- RNAi # 5 oligo SEQ ID NOS: 26 and 27, respectively:
- si5F 5, -TACCCCACCATCAGCATGGTCTTCCTGTCAACCATGCTGATGGTGGGGTACTTTTTG-3 '
- RNAi # 6 oligo SEQ ID NO: 29 and 30 respectively:
- si6F 5-( si6R: 3,-The expression vector of the present invention can contain a promoter on the 5th side of the hairpin DNA.
- promoters are pol III promoters, eg, U6 promoter or HI promoter from human or mouse, pol II promoter, or cytomegaloinoles promoter.
- Another example of the expression vector of the present invention is a tandem vector.
- This vector comprises a DNA sequence encoding the sense strand RNA sequence constituting the siRNA and a DNA sequence encoding the antisense strand RNA sequence, and a promoter at the 5 ′ end of each strand, DNA comprising a poly T sequence linked to the 3 'end of each strand, wherein the DNA hybridizes with the sense strand RNA and the antisense strand RNA after transcription in the cell to form the siRNA It is a vector.
- a promoter in a tandem vector is a pol HI promoter, such as a human or mouse-derived U6 promoter or HI promoter, or a site megaloinoles promoter.
- Poly T ⁇ ⁇ ⁇ IJ A poly T sequence consisting of ⁇ 6, preferably 1-5 T, such as 4 or 5 poly T sequences.
- tandem vector of the present invention can be introduced into a cell, transcribed into RNA corresponding to the sense strand and the antisense strand, and hybridized with each other to produce the desired siRNA.
- the hairpin type and tandem type vector is a plasmid vector or a winores vector.
- the plasmid vector may be prepared using the methods described in the Examples below or the methods described in the literature, or commercially available vector systems such as the piGENE TM U6 vector and the piGENE TM Hl vector (Takarabai Bio Inc.). Company) ( ⁇ ⁇ R.
- a plasmid vector is a drug resistance gene (eg, neomycin resistance gene, ampicillin resistance gene, puromycin resistance gene, hygromycin resistance gene) in addition to the DNA sequence and promoter encoding the siRNA of the present invention. It may contain transcription termination sequences, unique restriction sites or multiple cloning sites, and replication origins.
- drug resistance gene eg, neomycin resistance gene, ampicillin resistance gene, puromycin resistance gene, hygromycin resistance gene
- virus vector for example, an adenovirus vector, an adeno-associated virus vector, a lentiwinores betater, a retrowinoles vector (such as a leukemia virus vector), a herpes virus vector, or the like can be used.
- the virus vector is preferably of a type lacking self-replicating ability, for example, so as not to cause disease when used in humans.
- a self-replication ability-deficient adenovirus vector lacking the E1 gene and E3 gene eg, pAdeno- ⁇ from Invitrogen
- Construction of viral base click terpolymer can utilize literature methods (U.S. Patent No. 525 2 4 ⁇ 9 No., and International Publication W094 / 13788). ',
- the plasmid vector of the present invention forms a complex with a positively charged liposome such as lipophectamine, lipofectin, self-actin, or positively charged cholesterol, and is introduced into the patient's body in a force-pellated state.
- a positively charged liposome such as lipophectamine, lipofectin, self-actin, or positively charged cholesterol
- viral vectors can be introduced into cells by introducing them into affected areas and infecting cells (L. Zender et al., Proc. Natl. Acad. Sci. USA (2003), 100: 77797-7802; H. Xia et al., Nature Biotech.
- adenovirus vectors or adeno-associated virus vectors can introduce genes into various cell types with very high efficiency. Since this vector is also not incorporated into the genome, its effect is transient and is considered to be safer than other viral vectors.
- the sense strand sequence and the antisense strand sequence constituting the siRNA are covalently linked via a single strand loop sequence.
- the hairpin RNA is encoded, and the hairpin RNA is formed by transcription in a cell and processed by a dicer to form the siRNA.
- DNA encoding siRNA incorporated into the vector of the present invention comprises a DNA sequence encoding a sense strand sequence constituting siRNA and a DNA sequence encoding an antisense strand sequence, and A promoter is linked to the 5 'end of each strand, and a poly T sequence is linked to the 3' end of each strand, and the sense strand RNA and the antisense strand RNA hybridize after transcription in the cell.
- the siRNA is formed.
- Another nucleic acid as an active ingredient of the composition of the present invention is an antisense nucleic acid.
- This antisense nucleic acid is an RNA sequence containing a sequence complementary to the mRNA sequence corresponding to the DREF gene containing the nucleotide sequence of SEQ ID NO: 2 (positions 202 to 2286), or a partial sequence thereof, or a sequence. Either the nucleotide sequence of number 2 (positions 2 to 2286 ), or a partial sequence thereof, and DNA containing a complementary sequence. ,
- the partial sequence is a sequence consisting of about 30 or more, 50 or more, 70 or more, 100 or more, 150 or more, 200 or more, or 250 or more and a full length of, for example, 50 to 150 nucleotides in the DREF gene or mRNA sequence. Can be included.
- antisense nucleic acid nucleotides In addition to natural nucleotides, antisense nucleic acid nucleotides, antisense nucleic acid nucleotides, antisense nucleic acid nucleotides, antisense nucleic acid nucleotides, antisense nucleic acid nucleotides, antisense nucleic acid nucleotides, antisense nucleic acid nucleotides
- modified nucleotides having groups such as (fluorine, chlorine, bromine or iodine), methyl, carboxymethyl or thio groups.
- Antisense nucleic acids can be synthesized using well-known DNA / RNA synthesis techniques or DNA recombination techniques.
- the vector DNA containing the sequence of SEQ ID NO: 2 is mirrored, and the target sequence is amplified by polymerase chain reaction (PCR) using primers that squeeze the sequence to be amplified. If necessary, it can be cloned into a vector to produce antisense DNA.
- PCR polymerase chain reaction
- the DNA having the amplified target sequence thus obtained is inserted into a vector, the vector is introduced into a eukaryotic or prokaryotic cell, and the transcription system is used for antisense.
- RNA can be obtained.
- the above-described virus vector or plasmid vector can be used.
- the antisense nucleic acid of the present invention is DNA or RNA, it can suppress translation into protein by binding to DREF mRNA.
- the antisense nucleic acid may be encapsulated in a liposome such as a positively charged ribosome as described above, or the antisense nucleic acid may be, for example, a strong pol II or It may be incorporated into a vector (the above-mentioned plasmid or viral vector) so as to be under the control of the pol III promoter. 13
- the present invention further provides a composition for treating cancer comprising an antibody or a fragment thereof that suppresses the in vivo function of a DREF protein comprising the amino acid sequence of SEQ ID NO: 1 or a variant thereof.
- the target protein of the present invention is a protein encoded by the DREF gene, and is a protein involved in human cancer cell proliferation and cell cycle progression. Therefore, inhibition or suppression of the function of the protein expressed in cancer cells can result in suppression of cancer growth and cell death.
- a drug for this purpose is an antibody against the DREF protein or part thereof.
- the antibody against DREF is an antibody against the CR1 domain, CR2 domain or CR3 domain of DREF (SEQ ID NO: 135, 133 or 134, respectively), or a portion thereof.
- An example of a particularly preferred antibody is an antibody against a DRE (DNA replication-related element) binding region on DREF.
- the DNA-binding domain of DREF is called “BED zinc finger” and has a protein structure with one zinc ion. This domain is part of CR1 and contains the 20th to 74th sequence (SEQ ID NO: 140) of the DREF amino acid sequence of SEQ ID NO: 1.
- Antibodies to the domain containing this domain of DREF can inhibit DREF DNA binding and consequently inhibit DREF's in vivo function.
- Zinc finger amino acid sequence (SEQ ID NO: 140):
- Another domain associated with DNA binding is a part of CR3 of DREF, and includes the 571st to 651th sequence (SEQ ID NO: 141) of the DREF amino acid sequence of SEQ ID NO: 1.
- This domain is an activator's family (hAT element superf amily) Called the dimerization domain. Antibodies to the region of DREF containing this domain inhibit DREF dimer formation, resulting in inhibition of DREF DNA binding and inhibition of DREF in vivo function.
- amino acid sequence and nucleotide sequence of the following CR1, CR2, and CR3 domains are used to make (poly) peptides using known peptide synthesis techniques or DNA recombination techniques. Can be synthesized to produce the desired antibody.
- EDSNNHHLMLEASEWATIEGLVELLQPFKQVAEIV EVIAKELSKTYQETPEIDMFLNVATFLDPRYKRLPFLSAFERQQVENRVVEEAKGLLD CR2 nucleotide sequence (SEQ ID NO: 3): GTGGTGGAAGAGGCCAAGGGCCTGCTGGAC
- Such antibodies include polyclonal antibodies, monoclonal antibodies, recombinantly produced antibodies, human antibodies, humanized antibodies, chimeric antibodies, single chain antibodies, Fab, F (ab ') 2 , scFv, Fv, two These include bispecific antibodies and synthetic antibodies.
- the characteristics and structure of the antibody are described in Toru Otsuki et al., Experimental Medicine Vol. 22 No. 14 (Extra) 125-: 130, 2004, Yodosha (Tokyo, Japan). Can be used for.
- Preferred antibodies suitable for use in the present invention are human or humanized antibodies, particularly human or humanized monoclonal antibodies, that cause little or no side effects due to anaphylaxis.
- the antibody class and subclass may be of any type.
- the type of antibody, IgG ⁇ IgM ⁇ IgE ⁇ IgD, IgA include IgG had I g G 2, IgG 3, IgG 4, IgA have IgA 2.
- An antibody may also be derivatized by pegylation, acetylation, glycosylation, amidation, and the like.
- Human antibodies can be generated by, for example, phage display library (pharge display library).
- Human antibody-producing mice contain, for example, human antibody-producing genes in human artificial chromosomes After introducing the human chromosome fragment, using the microcell method, for example, an artificial chromosome is integrated into the genome of a mouse embryonic stem cell, transplanted into the uterus of a foster parent mouse, a chimeric mouse is born, a male or female chimeric mouse is bred, or a chimeric mouse And a wild-type mouse of the same species, for example, by producing a homozygous offspring mouse that contains a human antibody gene and is therefore capable of producing a human antibody (for example, Table 02/092812, International W0 98/24893, International W0 96/34096, etc.).
- This human antibody-producing transgenic mouse is immunized with the target DREF protein of the present invention as an antigen, and then the spleen is removed and fused with this spleen cell and mouse myeloma cell by conventional techniques to form a hybridoma.
- the desired monoclonal antibody can be prepared (G. Kohler and C. Milstein, Nature 256: 495-497, 1975).
- DNA encoding the antibody of interest is screened from a library of immunoglobulin genes obtained directly from untreated human lymphocytes, and phage particles are placed between this DNA and the antibody chain.
- phage particles are placed between this DNA and the antibody chain.
- To establish a physical association which involves enriching affinity-screened phage that present antibodies with affinity for the target.
- an antibody having a binding affinity for a target can be synthesized in a large amount by an ordinary method (for example, JP 20013-527832).
- a humanized antibody can be obtained, for example, by binding a human IgG complementarity determining region (CDR) produced from a mouse immunized with human DREF protein to human IgG.
- CDR complementarity determining region
- Such humanized antibodies can be produced using genetic recombination techniques. Techniques for humanizing antibodies are described, for example, in US Pat. Nos. 6639055 and 5530101. '' In the present invention, DREF protein variants include all naturally occurring variants in human individuals, such as variants based on polymorphisms or mutations, or selective Includes variants by splicing.
- the variant has a sequence in which one or several amino acids are substituted, deleted or added in the amino acid sequence of SEQ ID NO: 1, and has an in vivo function involved in cancer cell growth.
- “several” means 10 or less, 8 ⁇ or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 Means individual.
- the variant comprises a sequence having% identity of 95% or more, 97% or more, 98% or more, or 99% or more with the amino acid sequence of SEQ ID NO: 1.
- '' 0/0 identity may be determined based on the possible known BLAST program introduction of gaps (BL ASTX and BLASTN) In a sequence alignment (e.g. SF Altschul et al, J. Mol. Bio 215: 403-410, 1990, etc.).
- the antibody or fragment thereof of the present invention can be used as a therapeutic agent for cancer alone or in a form to which a drug such as an anticancer drug (chemotherapeutic agent, radioactive substance, etc.) is bound.
- Anti-cancer agents include, but are not limited to, chemotherapeutic agents such as taxol, cytarabine, cisplatin, etoposide, doxonorevicin, daunonorevicin, vinblastine, paclitaxe ⁇ , and radioactive metals such as radioactive indium, technetium, and ytterbium.
- Binding of the anticancer agent to the antibody can be performed by a method including binding of the anticancer agent to any site of the antibody constant region, for example, by covalent bonding through a linker or by coordinate bonding with a metal ion.
- Delivery of the antibody or fragment thereof to the patient is usually appropriate, either alone or in the form of the antibody or fragment thereof encapsulated in liposomes (preferably positively charged liposomes), microcapsules or nanoparticles. It can be administered by a parenteral route (for example, intravenous administration, intraperitoneal administration, intramuscular administration, subcutaneous administration, topical administration, etc.) in combination with an appropriate carrier, excipient or diluent. '1.4 Composition
- compositions of the invention can be used to treat patients with cancer, in particular cancers that express DREF.
- cancers include, but are not limited to, lung cancer, esophageal cancer, sputum cancer, stomach cancer, liver cancer, colon cancer, thyroid cancer, prostate cancer, bladder cancer, kidney cancer, skin cancer, gall cancer, brain tumor Breast cancer, ovarian cancer, cervical cancer, testicular cancer, lymphoma, melanoma, sarcoma, osteosarcoma, etc.
- More preferred cancers include lung cancer, esophageal cancer, breast cancer, vaginal cancer and the like.
- the dose of the nucleic acid in the composition of the present invention is not limited to the following when converted to siRNA molecules or antisense nucleic acid molecules, but is 1 ⁇ 100 to 100 ⁇ ⁇ ⁇ , preferably 10 ⁇ to 50 ⁇ per dosage unit. More preferably, it is 100 ⁇ to 20 °.
- the dose of the antibody or fragment thereof in the composition of the present invention is not limited to the following, but is about 1 to about 100 mg / ml, about 5 to about 70 mg / ml, about 10 to 50 mg per dosage unit. / ml.
- the above dose or dose may vary depending on the patient's condition, age, gender, severity, etc., and the dose or dose should be determined at the discretion of a specialist.
- the composition of the present invention usually contains a pharmaceutically acceptable carrier, excipient or diluent such as sterile water, physiological saline, buffer solution, non-aqueous liquid (eg almond oil, vegetable oil, ethanol, etc.), etc. Can be included.
- the composition further comprises a pharmaceutically acceptable stabilizer (for example, amino acids such as methionine), a preservative (methyl P-hydroxybenzoate, sorbic acid), an isotonic agent (for example, sodium chloride), an emulsifier ( For example, lecithin, gum arabic), a suspending agent (for example, a cellulose derivative) and the like can be contained. .
- a pharmaceutically acceptable carrier for example, amino acids such as methionine
- a preservative methyl P-hydroxybenzoate, sorbic acid
- an isotonic agent for example, sodium chloride
- an emulsifier for example, lecithin, gum arabic
- a suspending agent for example, a
- compositions of the present invention can be administered parenterally, for example intravenously, intraperitoneally, intramuscularly. Administration, subcutaneous administration, local administration, etc. Local administration includes direct injection into the affected area under surgery or endoscopy, etc.
- administration includes direct injection into the affected area under surgery or endoscopy, etc.
- One to several doses of the composition of the present invention to the patient at time intervals such as 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 6 months, 8 months, 1 year or 2 years Can be administered separately.
- RNAi nucleic acid, antisense nucleic acid, or antibody of the present invention When the RNAi nucleic acid, antisense nucleic acid, or antibody of the present invention is applied to cancer, particularly lung cancer, it can induce cell death together with cell cycle arrest strongly in lung cancer cells. In particular, in some lung cancer cells, induction of cell death in M phase called mitotic catastroph was observed. On the other hand, in normal lung-derived cell lines, cell cycle arrest was mild compared to lung cancer.
- the cancer treatment method targeting DREF of the present invention can be used as a tailor-made medicine optimal for cancer patient cases by combining with the screening method for therapeutic agents described below.
- the present invention further suppresses the expression or translation of the DREF gene by adding a candidate drug to a culture medium containing human cultured cancer cells or a human cell line in which the DREF gene has been transfected so that it can be strongly expressed (normal or non-normal).
- a method for screening a drug for treating cancer comprising screening a drug in vitro.
- drug screening is performed by inhibiting cancer cell growth, cell cycle arrest, and / or cell death in cancer cells or mammalian cells that strongly express DREF (eg, humans, mice, etc., preferably humans). Can be used as an index.
- DREF eg, humans, mice, etc., preferably humans.
- the method of the present invention comprises preparing a human cultured cancer cell or a cell line that strongly expresses DREF, culturing the cell in the presence of a candidate drug, and suppressing the expression of the DREF gene or mRNA. Or screening candidate drugs for cell proliferation inhibition, cell cycle arrest and Z or cell death induction of the cell or cell line.
- the human cultured cancer cells include known cancer cell lines, cancer cells derived from biopsies from cancer patients, etc., and can be used in the present invention.
- cancer cell lines are all lung cancer cell lines such as ACC-LC-91 (H. Osada et al., Mol. Carcinog., 2005, 44: 233-241), A549 (ATCC, Rockvill, MD), PC10 (Immuno-Biological Research Institute, Gunma, Japan), Calu6 (ATCC, Rockville, MD).
- a cell line that strongly expresses DREF can be prepared, for example, as follows.
- a lentivirus was prepared by inserting the DREF cDNA into a lentivirus vector (eg CSII-CMV-MCS-IRES2-Bsd vector (obtained from Dr. Hiroyuki Miyoshi of RIKEN (Kodama, Japan)).
- a lentivirus vector eg CSII-CMV-MCS-IRES2-Bsd vector (obtained from Dr. Hiroyuki Miyoshi of RIKEN (Kodama, Japan)
- Blasticidin Invitrogen
- the growth rate can be measured by MTT Atsey etc. using TetraColorOne TM (Seikagaku, Tokyo, Japan) etc.
- DREF strong expression cell lines eg BEAS2B- The DREF strain was observed to increase growth rate about 3 times compared to the control strain after 6 days of culture.
- the degree of suppression of DREF gene or raRNA expression can be determined by a comparison experiment with a control to which no candidate drug is added.
- the expression level is determined by a well-known method (for example, phenol / chloroform / isoform) from cancer cells or DREF strongly expressing cell lines.
- RT-PCR reverse transcriptase-PCR
- For synthesized cDNA, quantitative RT-PCR method, hybridization method using fluorescently or radioactively labeled probe eg Northern hybridization, Southern hybridization, DNA microarray, tissue microarray, etc.
- fluorescently or radioactively labeled probe eg Northern hybridization, Southern hybridization, DNA microarray, tissue microarray, etc.
- the expression level is determined by measuring the intracellular level of the protein encoded by the DREF gene (SEQ ID NO: 1) by immunoassay using an antibody against the protein or a fragment thereof, Western hyperprecipitation method, tissue staining method, etc. It can be determined indirectly by measurement.
- the probe is a nucleotide sequence of SEQ ID NO: 2 (positions 202 to 2286) or a sequence complementary thereto, or a sequence thereof, for example, about 20 or more, about 30 or more, 50 or more, 70 or more, 100 or more, 150 or more, 200 A DNA having a sequence consisting of 250 or more nucleotides.
- the probe is preferably a labeled probe bound with a fluorescent or radioactive label.
- Fluorescent labels include, for example, fluoresamine, rhodamine, their derivatives, Cy3, Cy5, etc.
- Radiolabels include, for example, radioactive phosphorus or thio atoms.
- Hybridization can be performed under low hybridization, medium hybridization or high hybridization conditions, preferably under high hybridization conditions.
- the hybridization temperature can generally be 5 to 10 ° C lower than the melting temperature (Tm).
- Tm melting temperature
- Candidate agents include, but are not limited to, small molecules, peptides, polypeptides, proteins, nucleosides, oligonucleotides, polynucleotides, nucleic acids (DNA or RNA), and the like.
- An immunoassay is an analysis method that uses an antigen-antibody reaction.
- an enzyme-linked antibody method for example, ELISA
- a fluorescent antibody method for example, a solid phase method
- a homogenous method for example, a sandwich method, a piotine / avidin system, etc.
- the solid phase for example, a commercially available plastic plate (for example, a 96-well plate made of polystyrene) can be used.
- the candidate The drug can be identified as a cancer therapeutic.
- Quantitative RT-PCR is performed, for example, by PCR using mRNA or poly A (+) RNA in the vertical form in the presence of thermostable polymerases such as Taq polymerase and using a PREF derived from the DREF gene sequence. Can do.
- the size of the primer is about 15-30 nucleotides, preferably 17-25 nucleotides.
- the expression level of the DREF gene is determined relative to the expression level of the housekeeping gene with no expression fluctuation.
- Candidate drug screening can also be performed by examining cell growth inhibition, cell cycle arrest, or cell death in human cultured cancer cells, or DREF strongly expressing cell lines.
- Cell proliferation can be measured by the following method. On the next day after gene transfer, repopulate the cells and select with puromycin / zg / ml) for 2 days, then continue with puromycin (0.5 ⁇ g / ml) for 10 days. Then, replace with TetraColorOne TM (Seikagaku, Tokyo, Japan) 5% medium and incubate at 37 ° C for 1 hour. The medium is then collected and removed. 0D450nm is measured using a reader (in this case, OD630nm is used as a control), and the number of living cells is determined (colorimetric assay). 0 After that, the cells are fixed with methanol, and then 5% Gierasa (Sigma-Aldrich) Stain with aqueous solution.
- Cell cycle and cell death can be measured by the following methods.
- the cells were observed with a stereomicroscope BHT-323 (Olympus), and the cells were interphase (G9 / G1, S, G2), prophase, metaphase, metaphase, late phase / terminal phase (Anaphase / telophase) ; ⁇ (mi tot ic catastroph ⁇ ), measure by apoptosis.
- RNAi nucleic acid A group of genes whose expression changes when DREF gene expression is suppressed by RNAi nucleic acid was analyzed using a microarray. ⁇
- Human cancer cell line Human lung cancer cell line (ACC-LC- 91 or A549), were cultured in 5% C0 2 presence with 5% fetal calf serum (Invitrogen) added RPMI 1640 medium (Sigma), it was used in the following experiments.
- Human normal airway epithelial cell lines (BEAS2B or HPL1) are 1% fetal bovine serum (Invitrogen;), 5 ⁇ g / ml bovine insulin (Sigma), 5 ⁇ g / ral human transferrin (Sigma), with 0. 1 mu M hydrocortisone (S i gma) ⁇ Pi 0. 2 nM preparative RHO de thyronine (S i gma) was added a Ham F12 medium (Sigma) and cultured in 5% C0 2 the presence of the following Used in the experiment.
- RNAi-vector gene transfer into cells For DREF expression vector and RNAi-vector gene transfer into cells (tra.nsf ect ion), seed 3 x 10 5 cells in a 3.5 cm culture dish and mix DNA and Lipofectamine 2000 (Invitrogen) the next day. And added to the medium. '' When a vector with a puromycin-resistant gene such as RNAi vector is transfected, puromyc in dihydrochlori de (Sigma-Aldrich, St (Louis, M0) 2 g / ml medium was added, and after 2 days of selection, the transfected cells were collected, and RNA and protein samples were prepared or FACS analysis was performed.
- the DREF expression vector was obtained by inserting the 0RF portion of the DREF cDNA obtained by RT-PCR into the expression vector pcDNA3 (Invitrogen). Dr. Fumiko Hirose (Hyogo Prefectural University, Hyogo, Japan) More distributed.
- RNAi sites # 1, # 2 as a control (# 1) and four new RNAi sites (# 3, # 4, # 5 and # 6) ) DNA oligonucleotide was prepared by requesting Greiner Japan (Tokyo, Japan). Its base sequence is as follows (note that the number at the RNAi site represents the position number of the nucleic acid residue when the base “A” of the start codon in DREF 0RF is the first position).
- RNAi # l (SEQ ID NO: 16)
- H1F1 H1R1:
- si3F 5 '-CAACAACCACCACCTCATGCCTTCCTGTCAGCATGAGGTGGTGGTTGTTGCTTTTTG-3'
- RNAi # 4 (SEQ ID NO: 22)
- RNAi # 5 (SEQ ID NO: 25)
- RNAi # 6 (SEQ ID NO: 28)
- Plasmid vector with puromycin resistance gene (pCMV-puro) [Note] This vector The filter was prepared by cutting off the unnecessary part 1. Okb length from Clontech's plasmid pIRESpuro2 (5.2 kb length).
- an RNAi vector containing the mouse U6 gene promoter, a promoter transcribed by RNA polymerase III was prepared (pU6-puro) (Osada., .. et al., Cancer Res. 65: 10680-5, 2005).
- an RNAi betater inserted with the human HI gene promoter was prepared (pHl-RNApuro).
- RNAi # l was inserted after digestion of pHl-RNApuro with Bglll-Hindlll.
- RNAi # 3 to # 6 were inserted after pU6-puro was digested with Apal, treated with T4 DNA polymerase, and digested with EcoRI.
- RNA was recovered from the transfected cells using the RNeasy Mini Kit (Qiagen).
- RT-PCR was performed using the One-step RT-PCR kit (Qiagen) according to the attached instruction manual, and the RT-PCR product was subjected to TBE-agarose gel electrophoresis to examine the expression of the DREF gene ( Figure 2). ).
- DREF F1 5, -TGGCAAGGACATCGTGAAGG (positions 813 to 832 of hDREF 0RF; SEQ ID NO: 135)
- DREF R1 5 '-GACAGCAACAACCACCACCT (1132 to 1151 of hDREF 0RF; SEQ ID NO: 136)'
- DREF protein was detected using an antibody against HA-tag / myc-tag to which DREF-cDNA was added.
- the cells after gene transfer are lysed with 4% SDS solution and heat-treated, and then the protein is electrophoresed on SDS-PAGE and Immobilon using the semi-dry electrotransfer system Ti ⁇ ns-Blot SD cell (Bio-Rad). -Transferred to Millipore.
- the membrane is overlaid with anti-HA-tag antibody (Santa cruz) and anti-myc-tag antibody (9E10, Santa cruz), washed and then overlaid with secondary antibody (Cell Signaling) bound with horseradish peroxidase, and ECL Western DREF protein was detected by Blotting Detection Reagents (GE Healthcare).
- FACS analysis ⁇ - The cells after gene transfer are suspended in IGEPAL (registered trademark) CA-630 (0.5% Sigma-Aldrich), Propidiura iodide (20 ⁇ g / ml, Sigma-Aldrich), PBS solution, and DNA in the naked nucleus is transferred. After staining, the amount of DNA was quantified using FACSCalibur (Becton-Dickinson), and the cell cycle (G0 / G1, S, G2 / M) was analyzed using the cell cycle analysis software ModFit (BD Biosciences).
- the cells were interphase (Go / Gl, S, G2), early cell division (Prophase), metaphase (Metaphase), late phase / terminal (Anaphase / telophase), and dead cells (mitot i (? catastroph and apoptos is)).
- Hiroyuki Miyoshi of RIKEN BioResource Center (Wako City, 'Saitama, Japan) from Lentiviral Systems (pENTR4-Hl, CS-RfA-EG, pCAG-HIVgp, pCMV-VSV-G -Receive distribution of RSV- Rev).
- the U6 prmoter-RNAi-oligo part of the pU6-puro-RNAi vector is inserted into the pENTR4-HI vector and then incorporated into the CS-RfA-EG vector using LR clonase (Invitrogen).
- CDNA is synthesized from 250 ng of the obtained 36 hour RNA sample using MMLV-RT (Agilent Technologies, Palo Alto, Calif.) And an oligo dT primer to which a T7 promoter sequence is added.
- Figure 1 shows the domain structure, RNAi site and sequence of the human DREF / ZBED1 gene.
- the RNAi system was constructed (pCMV-puro-siDREF # l to # 6)
- the DREF-RNAi system was transfected into a human lung cancer cell line, and then the transfected cells were selected by puromyc in treatment for 24-48 hours.
- the lentivirus vector was used for RNAi, and a short hairpin type.ol igo was inserted into the lentivirus vector in the same manner, and the gene was introduced into 293T cells according to a standard method. lentivirus particles expressing short hairpin RNA were collected and added to the cell culture for analysis.
- Figure 2 shows the results of RT-PCR analysis of the RNAi effect. Introduce RNAi vector pCMV-puro-s iDREF # l, # 3, # 4, '# 5, # 6 into lung cancer cell A549, and after puromycin treatment for 48 hours, collect cells, extract RNA, RT- DREF expression was examined by PCR. Compared to the control vector that does not express DREF-siRNA (pCMV-puro_) U6blank, all pCMV-puro-siDREF vectors showed decreased expression due to RNAi.
- pCMV-puro_ DREF-siRNA
- Figure 3 shows the results of Western blot analysis of the RNAi effect.
- a DREF full-length cDNA expression vector and a DREF full-length cDNA expression vector that had undergone a silent mutation in the RNAi sites of # 3 and # 4, respectively, and became RNAi resistant.
- DREF Changes in the expression of DREF protein were examined by Western blot. For detection, an anti-HA polyclonal antibody, which is an antibody against HA-tag inserted at the N-terminus of DREF cDNA, was used. siDREF # 3 In # 4, both wild-type DREF proteins showed a marked decrease. DREF (# 3mut, # 4mut) with # 3 and # 4 silent mutations was resistant to RNAi.
- Figure 4 shows the results of cell cycle analysis (1).
- s iDREF- # 3 and # 5 showed a decrease in the S phase, and a trend toward cell cycle arrest (G1 arrest) at the G1-S transition stage.
- G1 decreased significantly, G2 / M increased, and cell cycle arrest at a very strong G2 / M step was observed.
- a slight increase in G2 / M was also seen in # 3, and # 3 was thought to cause a strong G1 stop and a relatively mild G2 / M stop.
- Figures 5A, 5B, and 5C show the results of cell cycle analysis (2).
- pCMV-puro-siDREF, # 3, # 4 (and U6blank) were introduced into several lung cancer cell lines, and the cell cycle was analyzed.
- ACC-LC-172 as in ACC-LC-91, # 3 causes a strong G1 arrest and a relatively mild G2 / M arrest, while # 4 shows a strong G2 / M cell cycle arrest. It was.
- Calu6 'ACC-LC-319 also showed a tendency to stop G2 / M in # 4.
- A549- # 4 'PC10- # 3 and-# 4 showed a decrease in S and a tendency to stop G1.
- cell cycle arrest was hardly observed in the normal lung-derived cell lines BEAS2B and HPL1.
- 6A and 6B show the results of cell proliferation ability analysis. Puromycin-selection cell number after 10 days was analyzed with MTT assay (using cell proliferation kit). s iDREF- # 3 and # 4 showed a much stronger cell growth inhibitory effect than '4, and # 4 was considered to be stronger. # 1 had only a weak antiproliferative effect.
- FIG. 7 shows the results of staining with yH2AX (Phospho- ⁇ 2 ⁇ ) and Phospho-ATM (Serl981).
- yH2AX Phospho- ⁇ 2 ⁇
- Phospho-ATM Phospho-ATM
- Figure 8 shows the TUNEL analysis results. Induction of apoptosis (cell death) was examined by TUNEL analysis to detect DNA fragmentation. Compared to U6blank, apoptosis induction was seen at # 3 and # 4. Apoptosis frequency was different between # 3 and # 4, and # 3 was almost the same in 4 strains of A549, PC10, ACC-LC-91 and ACC-LC-172, but in # 4, A549 It is almost the same as PC10, and ACC-LC-91 is almost the same as ACC-LC-172, and ACC-LC-91 showed significantly greater apoptosis induction than A549 (some data not shown) ).
- FIG. 10 shows cell death (mitotic catastroph) in M phase.
- Phospho-histon H3 (H3-P) and ⁇ H2AX which are phosphorylated in M phase, were immunostained to investigate the relationship between DNA damage and cell death induction and the cell cycle.
- ACC-LC-91 showed that most of ⁇ 3- ⁇ positive ⁇ phase cells were ⁇ 2 ⁇ -positive, indicating that cell death was induced by DNA damage in the ⁇ phase.
- FIG. 11 shows the results of gene expression analysis of siDREF-# 3 and # 4.
- ACC-LC-91 was infected with siDREF # 3 and siDREF # 4 lentivirus to induce RNAi.
- SAS StatView
- DREF expression was observed, whereas in normal lung, DREF expression was absent.
- Fig. 13 shows the DNA sequence (A) of RPL17 gene, which is considered to be related to cancer growth, among the genes that are suppressed by DREF-RNAi # 4, and the mechanism of transcriptional activation by DREF of RPL17 gene.
- (B) is shown.
- DREF binds to this sequence to induce RPL17 DREF expression, and DREF-RNAi # 4 reduces DREF expression, so it is thought that RPL17 gene expression also decreases.
- RNAi induction 11 are shown in Table 1 (# 4) and Table 2 (# 3). These genes whose expression changes at an early stage of RNAi induction (36 hours) are directly regulated by DREF, and are likely to be cell groups that cause cell cycle arrest and cell death induction. It is considered to be a target.
- SRRMl / SRtnl60 is a co-activator of splicing and induces CD splicing abnormalities in an oncogene RAS-dependent manner (Mol Cell Biol. 2006 Jan; 26 (1): 362-70). It also binds to the oncogene TLS / FUS (Exp Cell Res. 2003 Feb 15; 283 (2): 184-95). The association with cancer development is strongly suggested.
- a gene family with TO40 domains involved in various processes such as signal transduction, apoptosis induction, pre-mRNA maturation, and cytoskeleton formation.
- HBV hepatitis B virus
- Oif icial symbol S ⁇ RS16 and Name: splicing factor, ar'ginine / serine-rich 16 (suppressor-of-white- apricot homolog, Drosophila)
- Su thigh ary Controls splicing before becoming mature mRNA after gene transcription.
- Oxf icial Symbol ZBED1 and Name: zinc finger, BED-type containing 1 Gene aliases: ALTE; TRAMP; KIAA0785
- CTDSP1 Carboxy-terminal domain, RNA polymerase II, polypeptide A
- Gene aliases SCP1; NLIIF (nuclear LIM interac tor-interacting factor) A phosphatase that regulates the activity of RNA polymerase II that transcribes the gene. It controls the expression of a wide range of gene groups.
- RNA helicase with a DEAD box domain that regulates RNA conformation. Involved in the control of a wide range of functions related to RNA, such as the initiation of translation into proteins and splicing.
- This gene family is a gene family with a WD40 domain involved in various processes such as signal transduction, apoptosis induction, pre-mRNA maturation, and cytoskeleton formation.
- TLR Toll-like receptor
- TNF receptor TNF receptor
- TNF-R / TNFRSF1A TNF receptor
- EGRl is a transcriptional regulatory molecule of C2H2_zinc-finger type that regulates the expression of genes involved in differentiation and cell division It is thought to be involved in cancer development.
- ITPKA inositol 1, 4, 5-trisphosphate 3 kinase
- IP3 is an intracellular signal concentration of calcium that is an important signal pathway ITPKA phosphorylates this IP3 to inositol 1, 3, 4, 5-tetrakisphosphate (IP (4)) and suppresses the increase in the strength concentration. It is deeply involved in the regulation of intracellular signals.
- TRAF7 and Name: TNF receptor-associated factor 7 Gene aliases: RFWD1; RNF119; MGC7807; DKFZp586I021
- Oii'icial Symbol DVLl; Name Name: dishevelled, dsh nomolog 1 (Drosophila) Summary: Onset of cancer 'Wnt- -catenin signaling pathway that has been reported to be involved in growth, suppresses -catenin degradation and Wnt --Increase catenin signal. 'Relevant to cancer development is strongly suggested.
- EGR1 is a C2H2-zinc-finger transcriptional regulatory molecule that regulates the expression of genes involved in differentiation and cell division and is thought to be involved in cancer development.
- CtBPl C-terminal binding molecule of adenovirus oncogene E1A.
- This CtBP gene family (CtBPl'CtBP2) is a target gene when the tumor suppressor gene ARF induces apoptosis induction independent of p53. Overexpression of CtBP induces p53-independent apoptosis induction of this ARF. Inhibits (Mol Cell Biol. 2006 Mar; 26 (6): 2360-72 ⁇ ). Therefore, this CtBPl is considered an oncogene.
- SWI / SNF complex that regulates chromatin structure and regulates gene expression. It also functions as a co-activator for nuclear hormone receptors such as androgen receptors that promote prostate cancer growth. It is deeply involved in the regulation of gene expression and essential for prostate cancer growth (Mol Cell Bi. L. 2005 Mar; 25 (6): 2200-15). The association with cancer development is strongly suggested.
- This is a substrate of the representative kinase-type oncogene Src family and functions by binding to PKD2, the gene responsible for polycystic kidney disease. It is suggested to be associated with cancer development.
- One of the biosynthetic enzymes of purine nucleic acid which is a material for DNA, and is related to cell growth.
- the SUV39H family (SUV39H1, SUV39H2) gene induces the methylation of histone H3-lysine9, which induces heterochromatin formation. It binds to RB2 / pl30 of the typical tumor suppressor gene Retinoblastoma (RB) family and promotes transcriptional regulation by RB2. It is suggested to be associated with cancer development. ,
- CEBPD CCAAT / enhancer binding protein
- This gene is upregulated in B-cell malignant lymphoma and is induced by interleukin-4. It is suggested to be related to the onset of tumors such as lymphoma.
- the cancer treatment targeting DREF according to the present invention has a very strong suppression of cell proliferation, induction of cell death in M phase, and G2 / M phase without substantially affecting normal tissues. It has a very strong cell cycle arresting effect in cancer and is very effective for cancer regression and cell death. ,
- the present invention enables cancer treatment using DREF as a molecular target. That is, since the therapeutic agent of the present invention is very effective for cancer regression and cell death, it can be effectively used for the treatment of cancer expressing DREF.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Medicinal Chemistry (AREA)
- Immunology (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Molecular Biology (AREA)
- Biomedical Technology (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Cell Biology (AREA)
- Hematology (AREA)
- Urology & Nephrology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Biochemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Epidemiology (AREA)
- Biotechnology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Toxicology (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Pathology (AREA)
- Microbiology (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Description
DREFを分子標的とする癌治療用組成物 技術分野 Cancer therapeutic composition using DREF as molecular target
本発明は、 ヒト細胞増殖制御因子 DREF又は DREF遺伝子を分子標的とする癌治 療用組成物に関する。' 明 The present invention relates to a composition for cancer treatment that targets the human cell growth regulator DREF or DREF gene as a molecular target. 'Ming
背景技術 Background art
従来、細胞増殖に関連する遺伝子を標的とする癌治療法は種々検討されている。 Conventionally, various cancer treatment methods targeting genes associated with cell proliferation have been studied.
書 book
DREFは Hirose らが単離したショウジョゥバエの転写因子で、 遺伝子プロモータ 一配列中の DRE - mot if と呼ばれる DNA配列を認識してそれに結合し、 その遺伝子 の発現を誘導する'(非特許文献 1 )。 ショ ウジヨ ウバエでは、 S 期に PCNA (prol iferating cel l nuclear antigen)転写を誘導することが報告され、 さ らに細胞増殖に関連するいくつかの遺伝子力 DREFにより発現制御されることが 報告されている。 その後、 ショ ウジヨウバエ DREFのヒ トホモログが、 Ohshimaら によってクローニング、 配列決定され、 ヒ ト DREFがヒストン HI遺伝子の誘導に 係わることが報告された (非特許文献 2 )。 このとき、 ヒ ト DREF遺伝子の発現が DREF CR1 ドメインに対して作製された 2つの siRNAによって低下し、 これによつ てヒス トン HI遺伝子発現が抑制される (非特許文献 2 )。 DREF is a Drosophila transcription factor isolated by Hirose et al., Which recognizes and binds to a DNA sequence called DRE-mot if in one sequence of a gene promoter and induces the expression of that gene (non-patent literature). 1). In Drosophila, it is reported that PCNA (prol iferating cell nuclear antigen) transcription is induced in the S phase, and that several gene forces related to cell proliferation are reported to be regulated by DREF. Yes. Thereafter, a human homologue of Drosophila DREF was cloned and sequenced by Ohshima et al., And it was reported that human DREF was involved in the induction of histone HI gene (Non-patent Document 2). At this time, the expression of the human DREF gene is reduced by the two siRNAs generated against the DREF CR1 domain, thereby suppressing the histone HI gene expression (Non-patent Document 2).
ヒ ト DREF遺伝子は、 Esposito ら (非特許文献 3 ) がヒト非誘導性男性奇形癌 細胞株由来の cDNA ライブラ リ一から見出した遺伝子 Tramp ( Ac-l ike transposable element; putative Ac - l ike transposon ; Chromosome:' X, Y; Location : Xp22. 33 ; Ypll) と同一である。 The human DREF gene is a gene that was found by Esposito et al. (Non-patent Document 3) from a cDNA library derived from a human non-inducible male teratocarcinoma cell line Tramp (Ac-like transposable element; putative Ac-like transposable element; Chromosome: 'X, Y; Location: Xp22.33; Ypll).
Sanoらは、 ヒ ト DREF遺伝子をノックダウンすることによって DREFが細胞周期 進行に関与する因子のひとつであること、 またノックダウンによってリポソーム タンパク質(RP)の発現低下が起こることから、 DREFがタンパク合成遺伝子の調節 を介して細胞増殖を刺激する役割を果たすことを指摘した (非特許文献 4及び非 特許文献 5 )。 . - これまでヒト DREFについて細胞増殖能との関連が示唆された力 S、しかし癌に対 する治療との関連での詳細な機能解析は行われていなかった。 Sano et al. Show that DREF is one of the factors involved in cell cycle progression by knocking down the human DREF gene, and that expression of liposomal protein (RP) is reduced by knockdown. It was pointed out that it plays a role of stimulating cell proliferation through gene regulation (Non-patent Documents 4 and 5). - So far, human DREF has been suggested to be associated with cell proliferation ability S, but no detailed functional analysis has been performed in relation to cancer therapy.
非特許文献 1 Hirose, F.ら, J. Biol. Chera. 271: 3930-3937, 1996 非特許文献 2 Ohshima, N.ら, J. Biol. Chem. 278: 22928 - 38, 2003 非特許文献 3 Espos ito, T.ら, Human Molecular Genet ics, 8 (1) : 61 - 67, Non-patent document 1 Hirose, F. et al., J. Biol. Chera. 271: 3930-3937, 1996 Non-patent document 2 Ohshima, N. et al., J. Biol. Chem. 278: 22928-38, 2003 Non-patent document 3 Espos ito, T., et al., Human Molecular Genetics, 8 (1): 61-67,
1999 1999
非特許文献 4 Sano, Y.ら, 日本分子生物学会要旨集 (平成 17年 11月 25 日発行) ,講演番号 1P - 0873, 2005 Non-Patent Document 4 Sano, Y. et al., Abstracts of the Molecular Biology Society of Japan (issued on November 25, 2005), lecture number 1P-0873, 2005
非特許文献 5 Sano, Y.ら, 第 20 回国際生化学分子生物学会要旨集 (平成 18年 6月 18日発行) ,講演番号 5P - A - 217, 2006 発明の開示 Non-Patent Document 5 Sano, Y. et al., 20th Annual Meeting of the International Biochemical Molecular Biology Society (issued June 18, 2006), Lecture No. 5P-A-217, 2006 Disclosure of Invention
肺癌、 食道癌などの難治性癌は、 一般的な化学療法に対して抵抗性であり外科 手術以外に治癒が期待できる有効な手段が確立されていない。 そのために、 癌の 発症 ·增殖 ·転移に関与する遺伝子を標的とする分子標的治療薬を開発して、 癌 の治癒を目指した治療法を確立することが求められている。 Refractory cancers such as lung cancer and esophageal cancer are resistant to general chemotherapy, and effective means that can be expected to cure other than surgery have not been established. For this purpose, it is necessary to develop molecular targeted therapeutics targeting genes involved in cancer onset, propagation and metastasis, and to establish treatments aimed at cure of cancer.
本発明の目的は、 癌に対する治療用組成物を提供することである。 An object of the present invention is to provide a therapeutic composition for cancer.
本発明の別の目的は、 癌に対する治療用薬剤をスクリーニングする方法を提供 することである。 ' Another object of the present invention is to provide a method of screening for a therapeutic agent for cancer. '
本発明者らは、 ヒト DREFが癌治療の標的になることを見出し、本発明を完成さ せた。 . The present inventors have found that human DREF is a target for cancer treatment, and have completed the present invention. .
DREFは、 背景技術で記載したように、 ショウジョゥバエの転写因子として発見 され、 その後、 ヒトにおいてもそのホモログが存在することが示された。 本発明 者らは今回、 ヒト DREFが正常細胞で実質的に発現されない一方で、癌細胞で有意 に発現されることを見出し、 これに基いて、 DREFが癌治療の分子標的になりうる と予測し、 DREFの発現抑制が実際に癌の抑制と細胞死に導くことを実証した。 したがって、 本発明は、 以下の特徴を有する。 As described in the background art, DREF was discovered as a transcription factor of Drosophila, and it was later shown that its homolog exists in humans. The present inventors have now found that human DREF is not substantially expressed in normal cells, but is significantly expressed in cancer cells, and based on this, DREF is predicted to be a molecular target for cancer treatment. It was demonstrated that suppression of DREF expression actually leads to cancer suppression and cell death. Therefore, the present invention has the following features.
本発明は、 その一の態様において、 癌細胞又は癌組織においてヒ ト細胞増殖制 御因子 DREF又は DREF遺伝子のィンビボ機能を抑制する、 DREFに対する RNAi核 酸、 アンチセンス核酸又は抗体を含むことを特徴とする癌治療用組成物を提供す る。 In one aspect thereof, the present invention provides an RNAi nucleus against DREF, which suppresses the in vivo function of human cell growth control factor DREF or DREF gene in cancer cells or cancer tissues. Provided is a cancer therapeutic composition comprising an acid, an antisense nucleic acid, or an antibody.
DREF は、 Hirose らによって命名された DRE (DNA replication-related element) -binding factor の略称であり、 別名 ZBEDl (zinc finger, BED- type containing 1)とも呼ばれている。 本明細書中では以後、 ヒ ト DREFを単に 「DREF」 と称する。 DREF is an abbreviation for DRE (DNA replication-related element) -binding factor named by Hirose et al., Also known as ZBEDl (zinc finger, BED-type containing 1). Hereinafter, the human DREF is simply referred to as “DREF”.
本発明の実施形態において、 DREFは配列番号 1に示されるアミノ酸配列、 或い は該アミノ酸配列において 1もしくは数個のアミノ酸の欠失、 置換又は付加を含 むアミノ酸配列、 を含むタンパク質である。 また、 DREF遺伝子は、 そのオープン リーディングフレーム(0RF)が配列番号 1のアミノ酸配列によってコードされる ヌクレオチド配列、 より具体的には配列番号 2の 202位〜 2286位に示されるヌク レオチド配列を含む。 DREF のァミノ酸及びヌクレオチド配列はともに、 GenBank に Accession number NMJ304729として登録されている。本発明における DREFは、 配列番号 1に示されるアミノ酸配列と 95%以上、好ましくは 97%以上、 より好ま しくは 98%以上、最も好ましくは 99%以上の同一性を有するアミノ酸配列を含み、 かつ DREFと同等のインビボ機能を有する変異体(例えば、 多型性による変異体、 スプライス変異体、縮重変異体、 突然変異体など も包含する。 また、本発明にお ける DREF遺伝子は、 配列番号 2に示されるヌクレオチド配列(202位〜 2286位) と 95%以上、 好ましくは 97%以上、 より好ましくは 98%以上、 最も好'ましくは 99%以上の同一性を有するヌクレオチド配列を含み、かつ翻訳後に DREFと同等の インビボ機能を有する上記と同様の変異体も包含する。 In an embodiment of the present invention, DREF is a protein comprising the amino acid sequence shown in SEQ ID NO: 1, or an amino acid sequence comprising a deletion, substitution or addition of one or several amino acids in the amino acid sequence. Further, DREF gene the open reading frame (0RF) nucleotide sequence encoded by an amino acid sequence of SEQ ID NO: 1, nucleotide sequence and more specifically shown in position 202 to 2 28 6 of SEQ ID NO: 2 Including. Both amino acid and nucleotide sequences of DREF are registered with GenBank as Accession number NMJ304729. DREF in the present invention includes an amino acid sequence having 95% or more, preferably 97% or more, more preferably 98% or more, and most preferably 99% or more identity with the amino acid sequence shown in SEQ ID NO: 1. Variants having in vivo functions equivalent to DREF (for example, polymorphic variants, splice variants, degenerate variants, mutants, etc. In addition, the DREF gene in the present invention is SEQ ID NO: nucleotide sequence shown in 2 (position 202 to 228 6th) 95% or more, preferably 97% or more, more preferably 98% or more, including the most favorable 'Mashiku nucleotide sequence having 99% or more identity And variants similar to those described above having an in vivo function equivalent to DREF after translation.
本明細書において、 DREF遺伝子は、 ェキソンのみならずイントロン、 3 '非翻 訳領域及び 5 '非翻訳領域も包含することを意図して使用される。 ' In this specification, the DREF gene is used with the intention of encompassing not only exons, but also introns, 3 'untranslated regions and 5' untranslated regions. '
本発明の別の実施形態において、 インビボ機能の抑制は、 癌細胞における、 細 胞増殖抑制、 細胞周期停止及び/又は細胞死誘導作用である。 In another embodiment of the present invention, the suppression of in vivo function is an effect of suppressing cell proliferation, cell cycle arrest and / or cell death in cancer cells.
本明細書で使用される 「インビボ機能」 は、 DREFの癌細胞内での働き、 例えば 細胞増殖、 細胞周期進行などをいう。 As used herein, “in vivo function” refers to the action of DREF in cancer cells, such as cell proliferation, cell cycle progression, and the like.
本発明の別の実施形態において、 RNAi核酸又はアンチセンス核酸は DREF raRNA の切断又はその機能の抑制を可能にするものである。 . 本明細書で使用される 「RNAi」 は、 所謂 RNA干渉(RNA interference)と呼ばれ るものであり、外から加えた DNA又は二本鎖 RNA (dsRNA)により、 それらと相同な 配列をもつ遺伝子の発現が抑制される現象をいう (例えば、 牛田千里、 蛋白質核 酸酵素 Vol. 46 (No. 10)、 pp. 1381- 1386、 2001)。 標的となる RNAは該遺伝子によつ てコードされる mRNA又はプレ mRNAであり、 標的 mRNAは RNAiによって分解され るか或いは発現抑制される。 In another embodiment of the invention, the RNAi nucleic acid or antisense nucleic acid is capable of cleaving DREF raRNA or suppressing its function. . As used herein, “RNAi” is so-called RNA interference, and is a gene having a sequence homologous to DNA or double-stranded RNA (dsRNA) added from the outside. (For example, Chisato Ushida, Protein Nucleic Acid Enzyme Vol. 46 (No. 10), pp. 1381-1386, 2001). The target RNA is mRNA or pre-mRNA encoded by the gene, and the target mRNA is degraded or suppressed in expression by RNAi.
具体的には、本発明で使用可能な好適な RNAi核酸は、 siRNA (small interfering RNA)又は shRNA (short hairpin RNA)、 該 siRNA又は s RNAをコードする DNAヽ 或 いは該 DNAを含むベクター DNAである。本発明において使用可能な別の RNAi核酸 には、 miRNA (microRNA)も含まれる。 miRNAは、 DREF mRNAの 3,非翻訳領域に結 合して DREF遺伝子の発現を抑制するものである。これらの RNAi核酸のサイズは、 19-30ヌクレオチド長、 好ましくは 19〜25ヌクレオチド長、 さらに好ましくは 19〜23ヌクレオチド長である。 Specifically, suitable RNAi nucleic acids that can be used in the present invention include siRNA (small interfering RNA) or shRNA (short hairpin RNA), a DNA encoding the siRNA or sRNA, or a vector DNA containing the DNA It is. Other RNAi nucleic acids that can be used in the present invention also include miRNA (microRNA). miRNA binds to DREF mRNA 3, the untranslated region and suppresses DREF gene expression. The size of these RNAi nucleic acids is 19-30 nucleotides long, preferably 19-25 nucleotides long, more preferably 19-23 nucleotides long.
本発明で使用される 「アンチセンス核酸」 は、 DREF mRNA の全体又はその部分 に相捕的な RNA配列、 或いは該 RNA配列をコードする DNA、 或いは該 DNA配列を 含むベクターを含む。 . The “antisense nucleic acid” used in the present invention includes an RNA sequence complementary to the whole or a part of DREF mRNA, DNA encoding the RNA sequence, or a vector containing the DNA sequence. .
本明細書で使用される 「ベクター」 は、 目的の siRNA又は shRNAをコードする DNA或いはアンチセンス RNAをコードする DNAの他に、 プロモーター、 ポリ T配 列、 複製開始点、 ターミネータ一などの調節配列、 選択マーカーなどを含むこと ができる。 本発明のベクターが癌細胞内に導入されると、 該 DNAの発現によって 該 siRNA又は shRNA、 .或いはアンチセンス RNAが細胞内で産生される。 As used herein, “vector” refers to a regulatory sequence such as a promoter, poly T sequence, replication origin, terminator, etc., in addition to the DNA encoding the target siRNA or shRNA or DNA encoding the antisense RNA. , Selection markers, etc. can be included. When the vector of the present invention is introduced into a cancer cell, the siRNA or shRNA, or antisense RNA is produced in the cell by the expression of the DNA.
本発明の実施形態において、 RNAi核酸又はアンチセンス核酸は DREFの CR2 ド メイン又は CR3 ドメインをコードするヌクレオチド配列 (それぞれ配列番号 3及 び 4 ) に由来するものである。 本発明における好ましいヌクレオチド配列は、 例 えば、 DREF遺伝子の配列番号 2の配列において、 1, 300位〜 1, 500位(0RFの開始 コドン ATGの 「A」 を第 1位とした場合 1099位〜 I2"位)、 1, 900〜2, 000位 (0RF の開始コドン ATGの「A」を第 1位とした場合 1699位〜 1799位)などに存在する。 本明細書で使用する 「CR」 は、 ヒト DREFとショウジョゥバエ DREFとの配列比 較において、 高度に保存された領域(conserved region)を示す。 そのような領域 (ァミノ酸配列;ヌクレオチド配列) は、 CR1 (配列番号 1の 4〜140位;配列番 号 2の 21ト 621位)、 CR2 (配列番号 1の 377〜506位;配列番号 2の 1330〜1719 位) 及び CR3 (配列番号 1の 541〜688位;配列番号 2の 1822〜2265位) からな り、 同一性はそれぞれ 27. 7%、 29. 2%、 21. 1%である (Ohshima, N.ら, J. Biol. Chem. 278: 22928—38, 2003)。 In an embodiment of the invention, the RNAi nucleic acid or antisense nucleic acid is derived from a nucleotide sequence encoding the CR2 domain or CR3 domain of DREF (SEQ ID NOs: 3 and 4, respectively). A preferable nucleotide sequence in the present invention is, for example, in the sequence of SEQ ID NO: 2 of the DREF gene, from position 1,300 to 1,500 (from position 1099 when “A” of the start codon ATG of 0RF is the first position) I 2 "position), 1,900 to 2,000 position (positions 1699 to 1799 when 0RF start codon ATG" A "is first position). As used herein, “CR” refers to a highly conserved region in the sequence comparison between human DREF and Drosophila DREF. Such areas (Amino acid sequence; nucleotide sequence) is CR1 (positions 4 to 140 of SEQ ID NO: 1; positions 21 to 621 of SEQ ID NO: 2), CR2 (positions 377 to 506 of SEQ ID NO: 1; 1330 to 1719 of SEQ ID NO: 2) ) And CR3 (positions 541 to 688 of SEQ ID NO: 1; positions 1822 to 2265 of SEQ ID NO: 2), and the identities are 27.7%, 29.2%, and 21.1%, respectively (Ohshima, N. et al., J. Biol. Chem. 278: 22928-38, 2003).
本発明でより好ましい RNAi核酸は、 配列番号 5〜15、 19、 22、 25及び 28のい ずれか 1つの DNA配列又は該 DNA配列に対応する RNA配列を含む。 本明細書中で 使用する、 DNAなる用語は、 デォキシリボ核酸を表わし、 RNAなる用語は、 リボ核 酸を表わす。 More preferred RNAi nucleic acids in the present invention include any one of the DNA sequences of SEQ ID NOs: 5 to 15, 19, 22, 25 and 28 or an RNA sequence corresponding to the DNA sequence. As used herein, the term DNA represents deoxyribonucleic acid and the term RNA represents ribonucleic acid.
また、 本発明でより好ましいベクターは、 配列番号 20、 21、 23、 24、 26、 27、 29及び 30のいずれか 1つの DNA配列を含む。 Further, a more preferable vector in the present invention includes the DNA sequence of any one of SEQ ID NOs: 20, 21, 23, 24, 26, 27, 29 and 30.
本発明によれば、 本発明の組成物は DREFのインビボ機能を抑制する、 DREFに 対する抗体を含むことができる。 このような機能抑制が可能であれば、 いかなる 抗体も本発明において使用可能である。 好ましい抗体は DREF の CR1 ドメイン、 CR2 ドメイン又は CR3 ドメイン (それぞれ配列番号 138, 133又は 134)、或いは該 CR1又は CR3 ドメイン中の DREFの DNA結合に関連する機能ドメイン(例えば配列 番号 140又は 141)、 のェピトープと結合する抗体である。 本明細書で使用するェ ピトープなる用語は、 抗原分子上の抗体が結合する部位を意味し、 通常、 そのよ うな部位のアミノ酸数が 8以上、好ましくは 10以上であり、アミノ酸は^ ^続的配 列からなっていてもよいし又は不連続的配列からなっていてもよい。 従って、 本 発明の抗体は、上記 DREFのアミノ酸配列中の連続する又は不連続の 8以上、好ま しくは 10 以上のアミノ酸からなるェピトープと免疫学的に結合することができ る。 ' 本発明で使用可能な好ましい DREFに対する抗体は、 ヒト抗体、 ヒ ト化抗体又は そのフラグメントである。 According to the present invention, the composition of the present invention can comprise an antibody against DREF that inhibits the in vivo function of DREF. Any antibody can be used in the present invention as long as such a function can be suppressed. Preferred antibodies are DREF CR1 domain, CR2 domain or CR3 domain (SEQ ID NO: 138, 133 or 134, respectively), or a functional domain related to DNA binding of DREF in the CR1 or CR3 domain (eg, SEQ ID NO: 140 or 141), It is an antibody that binds to the epitope. As used herein, the term epitope refers to a site to which an antibody on an antigen molecule binds, and usually the number of amino acids in such a site is 8 or more, preferably 10 or more. It may consist of a target sequence or a discontinuous sequence. Therefore, the antibody of the present invention can immunologically bind to an epitope composed of 8 or more, preferably 10 or more amino acids in the amino acid sequence of DREF. 'Preferred antibodies against DREF that can be used in the present invention are human antibodies, humanized antibodies or fragments thereof.
本発明はさらに、 別の態様において、 ヒ ト培養癌細胞を含む培地に候補薬剤を 加えて、 DREF遺伝子の発現又は翻訳を抑制する薬剤をインビトロでスクリーニン グすることを含む、 癌治療用薬剤をスクリーニングする方法を提供する。 In another embodiment, the present invention further includes adding a candidate drug to a medium containing human cultured cancer cells, and screening a drug that suppresses the expression or translation of the DREF gene in vitro. A method for screening is provided.
本発明で使用可能な癌細胞は、 癌細胞株、 バイオプシー (生検) からの癌細胞 を含む。 特に後者の癌細胞は、 患者由来のものであり、 該患者に有効な 剤の探 索のために使用しうる。 本発明において、 癌細胞の種類は特に限定されないが、 DREF が癌細胞で発現されるが正常細胞でほとんど又は全く発現されない癌細胞 が望ましい。 Cancer cells that can be used in the present invention are cancer cells from cancer cell lines, biopsy. including. In particular, the latter cancer cells are derived from a patient and can be used to search for an effective drug for the patient. In the present invention, the type of cancer cell is not particularly limited, but cancer cells in which DREF is expressed in cancer cells but hardly or not in normal cells are desirable.
本発明で使用可能な候補薬剤は、 小分子から高分子のいずれでもよく、 低分子 有機化合物、 ペプチド、 タンパク質、 オリゴ糖類、 多糖類、 脂質などを含む。 本発明において、 スクリーニングは、 癌細胞の細胞増殖抑制、 細胞周期停止及 び /又は細胞死誘導作用を指標として行うことができる。 このような作用は、 正 常組織又は細胞では実質的に認められないが、 これは、 DREFが正常組織又は細胞 でほとんど又は全く発現されないからである。 Candidate drugs that can be used in the present invention may be any of small molecules to macromolecules, and include low molecular organic compounds, peptides, proteins, oligosaccharides, polysaccharides, lipids, and the like. In the present invention, screening can be performed using as an index the suppression of cancer cell growth, cell cycle arrest and / or cell death induction. Such an effect is not substantially observed in normal tissues or cells because DREF is hardly or not expressed in normal tissues or cells.
.本明細書は本願の優先権の基礎である日本国特許出願 2006-275695号の明細書 および /または図面に記載される内容を包含する。 図面の簡単な説明 '、 This specification includes the contents described in the specification and / or drawings of Japanese Patent Application No. 2006-275695, which is the basis of the priority of the present application. Brief description of the drawings',
図 1は、 ヒ ト DREF/ZBED1遺伝子(Xp22. 33 & Ypll)のドメイン構造と、 RNAi部 位及び配列を示す . ' ' 図 2は、 RNAi効果の RT- PCR解析結果を示す。 Figure 1 shows the domain structure of the human DREF / ZBED1 gene (Xp22.33 & Ypll), and the RNAi site and sequence. '' Figure 2 shows the results of RT-PCR analysis of the RNAi effect.
図 3は、 RNAi効果の Western blot解析結果を示す。 Figure 3 shows the results of Western blot analysis of the RNAi effect.
図 4は、 細胞周期解析結果を示す。 Figure 4 shows the cell cycle analysis results.
図 5 Aは、 細胞周期解析結果を示す。 Figure 5A shows the cell cycle analysis results.
図 5 Bは、 細胞周期解析結果を示す。 Figure 5B shows the cell cycle analysis results.
図 5 Cは、 細胞周期解析結果を示す。 Figure 5C shows the cell cycle analysis results.
図 6は、 Aは細胞増殖能解析結果を示す。 Bは細胞増殖能解析結果を示す。 図 7は、 7 H2AX (Phospho- H2AX)、 Phospho- ATM (Serl981)免疫染色結果を示す。 図 8は、 TUNEL解析結果を示す。 In FIG. 6, A shows the results of cell proliferation ability analysis. B shows the results of cell proliferation ability analysis. FIG. 7 shows the results of immunostaining for 7 H2AX (Phospho-H2AX) and Phospho-ATM (Serl981). Figure 8 shows the TUNEL analysis results.
図 9は、'細胞死及び細胞周期解析結果を示す。 FIG. 9 shows the results of cell death and cell cycle analysis.
図 1 0は、 M期での細胞死 (mitotic catastroph) を示す。 FIG. 10 shows cell death (mitotic catastroph) in M phase.
図 1 1は、 ACC- LC- 91における s iDREF- , #4の遺伝子発現解析結果を示す。 図 1 2は、 肺癌患者検体における DREFの発現を示す ώ 図 1 3は、 DREF- RNAifH によって発現抑制される遺伝子の中で、 癌の増殖に関 連すると考えられる RPL17遺伝子の DNA配列(A;配列番号 137)と、RPL17遺伝子の DREFによる転写活性化の機序(B)を示す。 発明を実施するための最良の形態 Figure 11 shows the gene expression analysis results of siDREF-, # 4 in ACC-LC-91. 1 2, O showing expression of DREF in lung cancer patient specimens Figure 13 shows the DNA sequence of the RPL17 gene (A; SEQ ID NO: 137), which is thought to be associated with cancer growth, among the genes that are suppressed by DREF-RNAifH, and the transcriptional activation of the RPL17 gene by DREF. Mechanism (B) is shown. BEST MODE FOR CARRYING OUT THE INVENTION
1 . 癌治療用組成物 1. Cancer treatment composition
本発明は、 癌細胞又は癌組織においてヒ ト細胞増殖制御因子 DREF又は DREF遺 伝子のインビボ機能を抑制する、 DREFに対する RNAi核酸、 アンチセンス核酸又 は抗体を含むことを特徴とする癌治療用組成物を提供する。 The present invention includes an RNAi nucleic acid, an antisense nucleic acid or an antibody against DREF, which suppresses the in vivo function of a human cell growth regulator DREF or DREF gene in cancer cells or cancer tissues. A composition is provided.
DREFは、癌細胞又は癌組織で発現されるが、正常細胞又は正常組織では実質的 に (或いは、 ほとんど又は全く) 発現されない。 本発明者らは、 この知見に加え て、 癌細胞又は組織において DREF遺伝子の発現又はタンパク質への翻訳を抑制 (阻止又は阻害を含む) すると、 癌の細胞増殖抑制、 細胞周期停止、 細胞死誘導 などの事象が起こることを見出した。 ^ DREF is expressed in cancer cells or tissues, but is not substantially (or little or not) expressed in normal cells or tissues. In addition to this finding, the present inventors suppressed cancer cell growth, cell cycle arrest, cell death induction by suppressing (including blocking or inhibiting) DREF gene expression or protein translation in cancer cells or tissues. It was found that such an event occurred. ^
本発明による治療の対象となる癌は、 DREFの発現が可能である癌、 以下のもの に限定されないが、 例えば肺癌、 食道癌、 膝癌、 胃癌、肝癌、 大腸癌、 甲状腺癌、 前立腺癌、 膀胱癌、 腎癌、 皮膚癌、 胆癌、 脳腫瘍、 乳癌、 卵巣癌、 子宮頸癌、 精 巣癌、 リンパ腫、 メラノーマ、 肉腫、 骨肉種などを含む。 好ましい癌は、 肺癌、 食道癌、 乳癌、 膝癌などである。 ' , Cancers to be treated by the present invention are not limited to cancers capable of expressing DREF, such as lung cancer, esophageal cancer, knee cancer, stomach cancer, liver cancer, colon cancer, thyroid cancer, prostate cancer, Includes bladder cancer, kidney cancer, skin cancer, gall cancer, brain cancer, breast cancer, ovarian cancer, cervical cancer, testicular cancer, lymphoma, melanoma, sarcoma, osteosarcoma, etc. Preferred cancers are lung cancer, esophageal cancer, breast cancer, knee cancer and the like. ',
本発明の標的である DREFのアミノ酸配列及ぴ DREF遺伝子のヌクレオチド配列 はそれぞれ、 例えば配列番号 1、 配列番号 2 (オープンリーディングフレーム (0RF) : 202位〜 228δ位) として公知である。 DREFのアミノ酸及びヌクレオチド配 列はともに、 GenBankに Accession number丽_004729として登録されている。 本 発明においては、 DREF又は DREF遺伝子の変異体、 例えば、 突然変異、 多型、 選 択的スプライシング、 縮重などに基づいた変異体のインビボ機能を抑制すること も含む。 · The amino acid sequence of DREF, which is the target of the present invention, and the nucleotide sequence of the DREF gene are known, for example, as SEQ ID NO: 1 and SEQ ID NO: 2 (open reading frame (0RF): positions 202 to 228δ), respectively. Both DREF amino acid and nucleotide sequences are registered in GenBank as Accession number 丽 _004729. The present invention also includes suppressing the in vivo function of a DREF or a mutant of the DREF gene, for example, a mutant based on mutation, polymorphism, alternative splicing, degeneracy and the like. ·
具体的には、本発明における DREFは、配列番号 1に示されるアミノ酸配列、或 いは該アミノ酸配列において 1もしくは数個のアミノ酸の欠失、 置換又は付加を 含むアミノ酸配列、 を含むタンパク質である。 或いは、.本発明における DREFは、 配列番号 1に示されるアミノ酸配列と 95%以上、好ましくは 97%以上、 より好ま しくは 98%以上、最も好ましくは 99%以上の同一性を有するアミノ酸配列を含み、 かつ DREFと同等のインビボ機能を有する変異体も包含する。 Specifically, DREF in the present invention is a protein comprising the amino acid sequence shown in SEQ ID NO: 1, or an amino acid sequence containing one or several amino acid deletions, substitutions or additions in the amino acid sequence. . Or DREF in the present invention is In vivo function equivalent to DREF, comprising an amino acid sequence having 95% or more, preferably 97% or more, more preferably 98% or more, most preferably 99% or more identity with the amino acid sequence shown in SEQ ID NO: 1. The variant which has is also included.
また、本発明における DREF遺伝子は、配列番号 1のァミノ酸配列によってコー ドされるヌクレオチド配列、 より具体的には配列番号 2に示されるヌクレオチド 配列、 特に 202位〜 2286位の DREF 0RFに対応するヌクレオチド配列を含む。 本 発明における DREF遺伝子はさらに、 配列番号 2の 202位〜 2286位に示されるヌ クレオチド配列において 1もしくは数個のヌクレオチドの欠失、 置換又は付加を 含むヌクレオチド配列を含む DNAである。或いは、本発明における DREF遺伝子は、 配列番号 2の 202位〜 2286位に示されるヌクレオチド配列と 95%以上、好ましく は 97%以上、 より好ましくは 98%以上、 最も好ましくは 99%以上の同一性を有 するヌクレオチド配列を含み、かつ翻訳後に DREFと同等のインビポ機能を有する 変異体も包含する。 ' The DREF gene in the present invention corresponds to the nucleotide sequence encoded by the amino acid sequence of SEQ ID NO: 1, more specifically the nucleotide sequence shown in SEQ ID NO: 2, particularly DREF 0RF at positions 202 to 2286. Contains nucleotide sequence. The DREF gene in the present invention is a DNA further comprising a nucleotide sequence containing a deletion, substitution or addition of one or several nucleotides in the nucleotide sequence shown at positions 202 to 2286 of SEQ ID NO: 2. Alternatively, the DREF gene in the present invention has 95% or more, preferably 97% or more, more preferably 98% or more, and most preferably 99% or more identity with the nucleotide sequence shown in positions 202 to 2286 of SEQ ID NO: 2. And a variant having an in vivo function equivalent to DREF after translation. '
本明細書で使用される 「数個」 なる用語は、約 10以下の整数、例えば 9、 8、 7、 6、 5、 4、 3、 2を意味する。 As used herein, the term “several” means an integer of about 10 or less, such as 9, 8, 7, 6, 5, 4, 3, 2.
DREFは、 細胞増殖や細胞周期進行などのインビボ機能をもつが、 本発明は、 癌 において DREF のインビボ機能を抑制して癌の縮退や細胞死に導くことを可能に する。 DREF has in vivo functions such as cell proliferation and cell cycle progression, but the present invention makes it possible to suppress the in vivo functions of DREF in cancer and lead to cancer degeneration and cell death.
本発明において、標的 DREF遺伝子の発現又はタンパク質翻訳を抑制す.る薬剤と して、 DREFに対する RNAi核酸、 アンチセンス核酸及び抗体を挙げることができ る。 これらの各薬剤について、 以下にさらに具体的に説明する。 In the present invention, examples of the agent that suppresses the expression or protein translation of the target DREF gene include RNAi nucleic acid, antisense nucleic acid and antibody against DREF. Each of these drugs will be described more specifically below.
1. 1 RNAi核酸 1.1 RNAi nucleic acids
本発明で使用可能な RNAi核酸は、 DREF遺伝子 (ェキソン、 イントロン、 3 '非 翻訳領域及び 5 '非翻訳領域からなる) のヌクレオチド配列、 或いは DREF mRNA のヌクレオチド配列、 に由来の連続する 19〜30ヌクレオチド、 好ましくは 19〜 25ヌクレオチド、 より好ましくは 19〜23ヌクレオチドを含む DNA又は RNAであ る。 The RNAi nucleic acid usable in the present invention is a continuous 19 to 30 nucleotide sequence derived from the nucleotide sequence of the DREF gene (consisting of exons, introns, 3 ′ untranslated region and 5 ′ untranslated region) or the nucleotide sequence of DREF mRNA. It is a DNA or RNA containing nucleotides, preferably 19-25 nucleotides, more preferably 19-23 nucleotides.
本発明で使用可能な RNAi法は、 1 ) 短い二重鎖 RNA (siRNA)を細胞内に直接導 入する、或いは 2 ) short - hairpin型 RNA (shRNA)を各種発現ベクターに組み込み、 そのベクターを細胞内に導入する、 或いは 3 ) 対立方向に並ぶ 2個のプロモータ —を持つベクターに、 siRNAに対応する短い二重鎖 DNAをプロモーター間に揷入 して s iRNAを発現させるベクターを作製し、 細胞内に導入する、 などの手法を含 む。 RNAi methods that can be used in the present invention include 1) direct introduction of short double-stranded RNA (siRNA) into cells, or 2) incorporation of short-hairpin RNA (shRNA) into various expression vectors, Introduce the vector into the cell, or 3) Insert a short double-stranded DNA corresponding to siRNA between the promoters into a vector with two promoters aligned in the opposite direction to express s iRNA. This includes methods such as preparation and introduction into cells.
上記の RNAi核酸は、 DREF mRNAの切断又はその機能抑制を可能にする s iRNA、 shRNA又は miRNAを含む。また、 RNAi核酸は、ィンビボで使用したときに、 s iRNA、 shRNA又は miRNAを産生することを可能にする、 これらの RNAをコードする DNA 又は該 DNAを含むベクターであってもよい。 The RNAi nucleic acid includes s iRNA, shRNA or miRNA that enables cleavage of DREF mRNA or suppression of its function. The RNAi nucleic acid may also be a DNA encoding these RNAs or a vector containing the DNA, which, when used in vivo, makes it possible to produce siRNA, shRNA or miRNA.
本発明において、 好ましい RNAi核酸は、 DREFの CR2 ドメイン又は CR3 ドメイ ンをコードするヌクレオチド配列 (それぞれ配列番号 3及び 4) に由来の連続す る 19〜30ヌクレオチド、好ましくは 19〜25ヌクレオチド、 より好ましくは 19〜 23ヌクレオチドを含む DNA、 又は該 DNAに対応する RNAである。 さらに好ましい CR2 ドメイン又は CR3 ドメインの標的部位はそれぞれ、 例えば、 DREF遺伝子の配 列番号 2の配列において、 1, 300位〜 1, 500位 (0RFの開-始コドン ATGの 「A」 を 第 1位とした場合 1099位〜 1299位)、 1, 900〜2, 000位 (0RFの開始コドン ATGの In the present invention, a preferred RNAi nucleic acid is a continuous 19 to 30 nucleotides, preferably 19 to 25 nucleotides derived from nucleotide sequences (SEQ ID NOs: 3 and 4, respectively) encoding the CR2 domain or CR3 domain of DREF. Is DNA containing 19-23 nucleotides, or RNA corresponding to the DNA. Further preferred CR2 domain or CR3 domain target sites are, for example, positions 1,300 to 1,500 in the sequence of SEQ ID NO: 2 of the DREF gene (0RF open-start codon ATG “A” is the first Position 1099 to 1299), 1,900 to 2,000 (0RF start codon ATG
「A」 を第 1位とした場合 169.9位〜 1799位) などに存在する。 If “A” is the first place, it will be 169.9 to 1799).
本発明において使用可能な RNAi核酸は、例えば下記の配列番号 5〜15、 19、 22、 25及び 28のいずれかの DNA配列、 又はその DM配列に対応する RNA配列 (すな わち、 DNA配列の Tを Uに置き換えた配列)、 を含むものである。 なお、 記の配 列の後に記載の数字は、 DREFの 0RF上の開始部位の位置(但し、 0RFの開始コドン The RNAi nucleic acid that can be used in the present invention is, for example, the DNA sequence of any of the following SEQ ID NOs: 5 to 15, 19, 22, 25, and 28, or the RNA sequence corresponding to the DM sequence (that is, the DNA sequence) Array with T in U replaced by U). The number after the sequence is the position of the start site on 0RF of DREF (however, the start codon of 0RF
ATGの塩基 「A」 を第 1番目として番号づけする。 )を表す。 Number ATG base “A” as the first. ).
CTTGGTGGAGGACAGCAACAA 1122 (配列番号 5 ) CTTGGTGGAGGACAGCAACAA 1122 (SEQ ID NO: 5)
AGCAACAACCACCACCTCATG 1135 (配列番号 6 ) AGCAACAACCACCACCTCATG 1135 (SEQ ID NO: 6)
CAACCACCACCTCATGCTGGAG 1139 (配列番号 7 ) CAACCACCACCTCATGCTGGAG 1139 (SEQ ID NO: 7)
TGGCCGAGATCTTCTGCCAGAC 1642 (配列番号 8 ) TGGCCGAGATCTTCTGCCAGAC 1642 (SEQ ID NO: 8)
GGTGGTGGAGGAGCTGAGCAAC 1700 (配列番号 9 ) GGTGGTGGAGGAGCTGAGCAAC 1700 (SEQ ID NO: 9)
GTCCCAGAAGGTGCTTGGCCTC 1727 (配列番号 10) GTCCCAGAAGGTGCTTGGCCTC 1727 (SEQ ID NO: 10)
CCCAGAAGGTGCTTGGCCT 1730 (配列番号 11) CCCAGAAGGTGCTTGGCCT 1730 (SEQ ID NO: 11)
GAAGGTGCTTGGCCTCAACGA .;, . 1734 (配列番号 12) - GGTGCTTGGCCTCAACGAAGAC 1736 (配列番号 13) GAAGGTGCTTGGCCTCAACGA.;,. 1734 (SEQ ID NO: 12)- GGTGCTTGGCCTCAACGAAGAC 1736 (SEQ ID NO: 13)
GGTGCTTGGCCTCAACGAAGA 1737 (配列番号 14) GGTGCTTGGCCTCAACGAAGA 1737 (SEQ ID NO: 14)
TGCTTGGCCTCAACGAAGA 1739 (配列番号 15) TGCTTGGCCTCAACGAAGA 1739 (SEQ ID NO: 15)
GCAACAACCACCACCTCATGC 1136 (配列番号 19) GCAACAACCACCACCTCATGC 1136 (SEQ ID NO: 19)
GCAACTTCAAGTCCCAGAAGG 1718 (配列番号 22) GCAACTTCAAGTCCCAGAAGG 1718 (SEQ ID NO: 22)
GTACCCCACCATCAGCATGGT 1245 (配列番号 25) GTACCCCACCATCAGCATGGT 1245 (SEQ ID NO: 25)
GGTGCTTGGCCTCAACGAAGA 1737 (配列番号 28) GGTGCTTGGCCTCAACGAAGA 1737 (SEQ ID NO: 28)
特に好ましい配列は、配列番号 19 (RNAi #3 ( 1136〜1 156位))、配列番号 22 (RNAi #4 ( 1718〜1738位) :)、 配列番号 25 (RNAi #5 ( 1245〜1265位) ) , 及び配列番号 28 (RNAi #6 ( 1737〜1757位) )の DNA配列、 又はその DNA配列に対応する RNA配 列 (すなわち、 DNA配列の Tを Uに置き換えた配列')、 である。 なお、 配列番号と 共に括弧内に示した数字範囲は、 配列番号 2のヌクレオチド配列において、 DREF 0RFの開始コドン ATGの塩基「A」 (202位) を第 1番目として番号づけし.たときの 位置を表す。 · Particularly preferred sequences are SEQ ID NO: 19 (RNAi # 3 (positions 1136 to 1156)), SEQ ID NO: 22 (RNAi # 4 (positions 1718 to 1738) :), SEQ ID NO: 25 (RNAi # 5 (positions 1245 to 1265)) ), And SEQ ID NO: 28 (RNAi # 6 (positions 1737 to 1757)), or an RNA sequence corresponding to the DNA sequence (that is, a sequence in which T in the DNA sequence is replaced with U). The number range shown in parentheses together with the SEQ ID No. is when the base number “A” (position 2 ) of the start codon ATG of DREF 0RF is numbered as the first in the nucleotide sequence of SEQ ID No. 2. Represents the position of. ·
DREFに対する RNAi核酸の別の例は、 下記の配列番号 33〜132の DNA配列、 又 はその DNA配列に対応する RNA配列 (すなわち、 DNA配列の Tを Uに置き換えた 配列)、 である。 Another example of an RNAi nucleic acid for DREF is the DNA sequence of SEQ ID NO: 33-132 below, or the RNA sequence corresponding to that DNA sequence (ie, a sequence in which T in the DNA sequence is replaced with U).
CAAGAGCAAGGTGTGGAAGTATT 63 (配列番号 33) CAAGAGCAAGGTGTGGAAGTATT 63 (SEQ ID NO: 33)
GTGGAAGTATTTCGGCTTCGA 75 ' (配列番号 34) GTGGAAGTATTTCGGCTTCGA 75 '(SEQ ID NO: 34)
GGGATGCATCCTGCAGTGGAAG 107 (配列番号 35) GGGATGCATCCTGCAGTGGAAG 107 (SEQ ID NO: 35)
GATGCATCCTGCAGTGGAAGA . 110 (配列番号 36) GATGCATCCTGCAGTGGAAGA .110 (SEQ ID NO: 36)
GATGCATCCTGCAGTGGAA 110 (配列番号 37) GATGCATCCTGCAGTGGAA 110 (SEQ ID NO: 37)
CCCAGATCGCCTACTCCGGAAA 154 (配列番号 38) CCCAGATCGCCTACTCCGGAAA 154 (SEQ ID NO: 38)
CCAGATCGCCTACTCCGGAAA 156 (配列番号 39) CCAGATCGCCTACTCCGGAAA 156 (SEQ ID NO: 39)
CAGATCGCCTACTCCGGAA 157 (配列番号 40) CAGATCGCCTACTCCGGAA 157 (SEQ ID NO: 40)
AGATCGCCTACTCCGGAAACA 158 (配列番号 41 ) AGATCGCCTACTCCGGAAACA 158 (SEQ ID NO: 41)
GTCCTACCACCTGGAGAAGAAC 188 (配列番号 2) GTCCTACCACCTGGAGAAGAAC 188 (SEQ ID NO: 2)
CCTACCACCTGGAGAAGAA 191 (配列番号 43) CCTACCACCTGGAGAAGAA 191 (SEQ ID NO: 43)
CTGCGAGTTCGTCAAGAGCAAC 224 (配列番号 44) GCGAGTTCGTCAAGAGCAA 227 (配列番号 45) CGTGCTGGGCCTCATCTGCGAG 386 (配列番号 46) CCCAGCCTCCATCGTGGACGAG 416 (配列番号 47) CAGCCTCCATCGTGGACGA 419 (配列番号 48) CACCTTCAAGGTGCTGCTGAAG 440 (配列番号 49) CCTTCAAGGTGCTGCTGAA 443 (配列番号 50) AGCTGCCCAGCCGGAAGTACAT 481 (配列番号 51) CCGGAAGTACATCTCTACCAAG 491 (配列番号 52) CTCTACCAAGGCCATCCCTGAG 503 (配列番号 53) CGTCCGGGAGGTGATCCTGAAG 536 (配列番号 54) TCCGGGAGGTGATCCTGAA 538 (配列番号 55) CCGGGAGGTGATCCTGAAGGAG 539 (配列番号 56) GGGAGGTGATCCTGAAGGA 542 (配列番号 57) CCGAGGCCACCTGGTGTGGCAT 565 (配列番号 58) GAGGCCACCTGGTGTGGCA 568 (配列番号 59) CACCGACATGTGGAGGAGTGA 591 (配列番号 60) CGACATGTGGAGGAGTGAGAA 594 (配列番号 61) GTGGAGGAGTGAGAATCAGAAC 599 (配列番号 62) ATCAGAACCGCGCCTACGTCAC 613 (配列番号 63) TGGGCTCCCGCTGCCTGAAGAC 682 (配列番号 64) CTCCCGCTGCCTGAAGACCTTC 686 (配列番号 65) CCCGCTGCCTGAAGACCTT 689 (配列番号 66) CATCACGCGAGTGCTCTATGAGG 738 (配列番号 67) GCTCTATGAGGTCTTCATCGAG 749 (配列番号 68) TTCGGGGCCACCACCAACTATGG 793 (配列番号 69) CACCAACTATGGCAAGGACATC 803 (配列番号 70) CCAACTATGGCAAGGACAT 806 (配列番号 71) AGGACATCGTGAAGGCGTGCT 818 (配列番号 72) CTGCCGCAAACTGGTGGAGTAC 941 (配列番号 73) GCCGCAAACTGGTGGAGTACT 944 (配列番号 74)CTGCGAGTTCGTCAAGAGCAAC 224 (SEQ ID NO: 44) GCGAGTTCGTCAAGAGCAA 227 (SEQ ID NO: 45) CGTGCTGGGCCTCATCTGCGAG 386 (SEQ ID NO: 46) CCCAGCCTCCATCGTGGACGAG 416 (SEQ ID NO: 47) CAGCCTCCATCGTGGACGA 419 (SEQ ID NO: 48) CACCTTCAAGGTGCTGCTGAAG 440 (SEQ ID NO: 49) CCTTCAAGGTGCTGGA 491 (SEQ ID NO: 52) CTCTACCAAGGCCATCCCTGAG 503 (SEQ ID NO: 53) CGTCCGGGAGGTGATCCTGAAG 536 (SEQ ID NO: 54) TCCGGGAGGTGATCCTGAA 538 (SEQ ID NO: 55) CCGGGAGGTGATCCTGAAGGAG 539 (SEQ ID NO: 56) GGGAGGTGATCCTGAAGGA GT GG (GT ACC GG (SEQ ID NO: 59) CACCGACATGTGGAGGAGTGA 591 (SEQ ID NO: 60) CGACATGTGGAGGAGTGAGAA 594 (SEQ ID NO: 61) GTGGAGGAGTGAGAATCAGAAC 599 (SEQ ID NO: 62) ATCAGAACCGCGCCTACGTCAC 613 (SEQ ID NO: 63) TGGGCTCCCGCTGCCTGAAGAC 682 (TTC GCGACT CTCG (SEQ ID NO: 66) CATCACGCGAGTGCTCTATGAGG 738 (Allocation No. 67) GCTCTATGAGGTCTTCATCGAG 749 (SEQ ID NO: 68) TTCGGGGCCACCACCAACTATGG 793 (SEQ ID NO: 69) CACCAACTATGGCAAGGACATC 803 (SEQ ID NO: 70) CCAACTATGGCAAGGACAT 806 (SEQ ID NO: 71) AGGACATCGTGAAGGCGTGCT 818 (SEQ ID NO: 72) CTGCCGCAAACTGGTGGAGTAC 941 (SEQ ID NO: 73) GCCGCAAACTGGTGGAGTACT 944 (SEQ ID NO: 74)
GCCGCAAACTGGTGGAGTA 944 (配列番号 75)GCCGCAAACTGGTGGAGTA 944 (SEQ ID NO: 75)
CCGCAAACTGGTGGAGTACTTCC 945 (配列番号 76)CCGCAAACTGGTGGAGTACTTCC 945 (SEQ ID NO: 76)
GTGGAGTACTTCCAGCAGTCT 955 (配列番号 77)GTGGAGTACTTCCAGCAGTCT 955 (SEQ ID NO: 77)
CGTGGCCATGTACATGCTCTA 978 (配列番号 78)CGTGGCCATGTACATGCTCTA 978 (SEQ ID NO: 78)
GTGGCCATGTACATGCTCTAT 979 (配列番号 79)GTGGCCATGTACATGCTCTAT 979 (SEQ ID NO: 79)
GGCCATGTACATGCTCTATGAG 980 (配列番号 80)GGCCATGTACATGCTCTATGAG 980 (SEQ ID NO: 80)
CCATGTACATGCTCTATGA 982 (配列番号 81 )CCATGTACATGCTCTATGA 982 (SEQ ID NO: 81)
CATGTACATGCTCTATGAGAAG 983 (配列番号 82)CATGTACATGCTCTATGAGAAG 983 (SEQ ID NO: 82)
CATGCTCTATGAGAAGCAGAAG 989 (配列番号 83)CATGCTCTATGAGAAGCAGAAG 989 (SEQ ID NO: 83)
TGCTCTATGAGAAGCAGAA 992 (配列番号 84)TGCTCTATGAGAAGCAGAA 992 (SEQ ID NO: 84)
GCTGCAGCGCCTCAAGGAGCAG 1079 (配列番号 85)GCTGCAGCGCCTCAAGGAGCAG 1079 (SEQ ID NO: 85)
CCGAGATGCTGTCGGCCTCCAG 1222 (配列番号 86)CCGAGATGCTGTCGGCCTCCAG 1222 (SEQ ID NO: 86)
GAGATGCTGTCGGCCTCCA 1225 (配列番号 87)GAGATGCTGTCGGCCTCCA 1225 (SEQ ID NO: 87)
GCTGCTGCACATGCTCCTGAAC 1271 (配列番号 88)GCTGCTGCACATGCTCCTGAAC 1271 (SEQ ID NO: 88)
TGCTGCACATGCTCCTGAA 1274 (配列番号 89)TGCTGCACATGCTCCTGAA 1274 (SEQ ID NO: 89)
TGCACATGCTCCTGAACACCAC 1276 (配列番号 90)TGCACATGCTCCTGAACACCAC 1276 (SEQ ID NO: 90)
GGCCAAGGAGGTCATCGCCAAG 1337 (配列番号 91)GGCCAAGGAGGTCATCGCCAAG 1337 (SEQ ID NO: 91)
CCAAGGAGGTCATCGCCAA 1340 (配列番号 92)CCAAGGAGGTCATCGCCAA 1340 (SEQ ID NO: 92)
CATCGCCAAGGAGCTTTCCAAG 1349 (配列番号 93)CATCGCCAAGGAGCTTTCCAAG 1349 (SEQ ID NO: 93)
CAAGGAGCTTTCCAAGACCTA 1356 (配列番号 94)CAAGGAGCTTTCCAAGACCTA 1356 (SEQ ID NO: 94)
GACGCCCGAGATCGACATGTTTC 1383 (配列番号 95)GACGCCCGAGATCGACATGTTTC 1383 (SEQ ID NO: 95)
ACGCCCGAGATCGACATGTTTCT 1384 (配列番号 96)ACGCCCGAGATCGACATGTTTCT 1384 (SEQ ID NO: 96)
GCCCGAGATCGACATGTTTCTCA 1386 (配列番号 97)GCCCGAGATCGACATGTTTCTCA 1386 (SEQ ID NO: 97)
CCGAGATCGACATGTTTCTCAAC 1388 (配列番号 98)CCGAGATCGACATGTTTCTCAAC 1388 (SEQ ID NO: 98)
GGTGGAGAATCGCGTGGTGGAA 1475 (配列番号 99)GGTGGAGAATCGCGTGGTGGAA 1475 (SEQ ID NO: 99)
TGGAGAATCGCGTGGTGGA 1478 (配列番号 100)TGGAGAATCGCGTGGTGGA 1478 (SEQ ID NO: 100)
GGAGAATCGCGTGGTGGAAGA 1479 (配列番号 101)GGAGAATCGCGTGGTGGAAGA 1479 (SEQ ID NO: 101)
GGCCAAGGGCCTGCTGGACAAG 1499 (配列番号 102) CCAAGGGCCTGCTGGACAA 1502 (配列番号 103〕GGCCAAGGGCCTGCTGGACAAG 1499 (SEQ ID NO: 102) CCAAGGGCCTGCTGGACAA 1502 (SEQ ID NO: 103)
GCCCGCCAGCGTCATCAACAAC 1616 (配列番号 104〕GCCCGCCAGCGTCATCAACAAC 1616 (SEQ ID NO: 104)
CGGCCAGCGTCATCAACAACAT 1618 (配列番号 105〕CGGCCAGCGTCATCAACAACAT 1618 (SEQ ID NO: 105)
CCGCCAGCGTCATCAACAA 1619 (配列番号 106:CCGCCAGCGTCATCAACAA 1619 (SEQ ID NO: 106:
CGCCAGCGTCATCAACAACAT 1620 (配列番号 107:CGCCAGCGTCATCAACAACAT 1620 (SEQ ID NO: 107:
CGCCAGCGTCATCAACAACATGC 1620 (配列番号 108〕CGCCAGCGTCATCAACAACATGC 1620 (SEQ ID NO: 108)
CATCAACAACATGCTGGCCGAG 1628 (配列番号 109:CATCAACAACATGCTGGCCGAG 1628 (SEQ ID NO: 109:
ACAACATGCTGGCCGAGATCT 1634 (配列番号 110:ACAACATGCTGGCCGAGATCT 1634 (SEQ ID NO: 110:
GCTGCCCAAGGTGCTGCAGAAG 1799 (配列番号 111:GCTGCCCAAGGTGCTGCAGAAG 1799 (SEQ ID NO: 111:
TGCCCAAGGTGCTGCAGAA 1801 (配列番号 112:TGCCCAAGGTGCTGCAGAA 1801 (SEQ ID NO: 112:
GCCCAAGGTGCTGCAGAAGTAC 1802 (配列番号 113:GCCCAAGGTGCTGCAGAAGTAC 1802 (SEQ ID NO: 113:
CCAAGGTGCTGCAGAAGTA 1805 (配列番号 1 14CCAAGGTGCTGCAGAAGTA 1805 (SEQ ID NO: 1 14
TGCAGAAGTACTGGTGCGTGAC 1813 (配列番号 115TGCAGAAGTACTGGTGCGTGAC 1813 (SEQ ID NO: 115
TGCAGAAGTACTGGTGCGTGA 1814 (配列番号 1 16TGCAGAAGTACTGGTGCGTGA 1814 (SEQ ID NO: 1 16
CAGAAGTACTGGTGCGTGA 1816 (配列番号 117CAGAAGTACTGGTGCGTGA 1816 (SEQ ID NO: 117
CGTGGTCAGCGCCAAGAGGAAC . 1880 (配列番号 118CGTGGTCAGCGCCAAGAGGAAC. 1880 (SEQ ID NO: 118
GGACGAGCAGGTGTTTCTGTAT 1922 (配列番号 119GGACGAGCAGGTGTTTCTGTAT 1922 (SEQ ID NO: 119
GGACGAGCAGGTGTTTCTGTAT 1922 (配列番号 120GGACGAGCAGGTGTTTCTGTAT 1922 (SEQ ID NO: 120
ACGAGCAGGTGTTTCTGTA 1924 (配列番号 121ACGAGCAGGTGTTTCTGTA 1924 (SEQ ID NO: 121
ACGAGCAGGTGTTTCTGTATGAG 1924 (配列番号 122ACGAGCAGGTGTTTCTGTATGAG 1924 (SEQ ID NO: 122
GCAGGTGTTTCTGTATGAGAAC 1928 (配列番号 123GCAGGTGTTTCTGTATGAGAAC 1928 (SEQ ID NO: 123
AGGTGTTTCTGTATGAGAA 1931 (配列番号 124AGGTGTTTCTGTATGAGAA 1931 (SEQ ID NO: 124
TGGCGTCAGCGGCGGTTTCTTTG 2037 (配列番号 125TGGCGTCAGCGGCGGTTTCTTTG 2037 (SEQ ID NO: 125
GGCGTCAGCGGCGGTTTCTTTGG 2038 (配列番号 126GGCGTCAGCGGCGGTTTCTTTGG 2038 (SEQ ID NO: 126
CAGCGGCGGTTTCTTTGGCATT 2042 (配列番号 127CAGCGGCGGTTTCTTTGGCATT 2042 (SEQ ID NO: 127
GCGGCGGTTTCTTTGGCATTAG 2044 (配列番号 128GCGGCGGTTTCTTTGGCATTAG 2044 (SEQ ID NO: 128
GCGGCGGTTTCTTTGGCAT 2045 (配列番号 129GCGGCGGTTTCTTTGGCAT 2045 (SEQ ID NO: 129
CGGTTTCTTTGGCATTAGGGAC 2048 (配列番号 130CGGTTTCTTTGGCATTAGGGAC 2048 (SEQ ID NO: 130
GTTTCTTTGGCATTAGGGA . 2051 (配列番号 131 TAGGGACAGCAGCTTCCTGTAG 2063 (配列番号 132) GTTTCTTTGGCATTAGGGA .2051 (SEQ ID NO: 131 TAGGGACAGCAGCTTCCTGTAG 2063 (SEQ ID NO: 132)
本発明の好適な実施形態において、 RNAi核酸は、 配列番号 2のヌクレオチド配 列を含む DRSF遺伝子によってコードされる mRNA又はその変異体の配列からの連 続する 19〜30ヌクレオチド、 好ましくは 19〜25ヌクレオチド、 より好ましくは 19〜23 ヌクレオチドのセンス鎖配列とその相補的配列であるアンチセンス鎖配 列とを含んでなる siRNAである。 ここで、 該 siRNAは、 インビボ又はインビトロ で DREF遺伝子又は mRNAの発現を抑制することができる。 siRNAを構成する対応 の DNA配列の好ましい例は、 DREFの CR2 ドメイン又は CR3 ドメインをコードする ヌクレオチド配列 (それぞれ配列番号 3及び 4) に由来の連続する 19〜30ヌクレ ォチド、 好ましくは 19〜25ヌクレオチド、 より好ましくは 19〜23ヌクレオチド を含む DNA配列、 より好ましい例は、 配列番号 2のヌクレオチド配列において、 1, 300位〜 1, 500位 (0RFの開始コ ドン ATGの 「A」 を第 1位とした場合 1099位〜 1299位)、 1,900〜2,000位(0RFの開始コドン ATGの「A」を第 1位とした場合 1699 位〜 1799位) の配列からの連続する 19〜30ヌクレオチド、 好ましくは 19〜25ヌ クレオチド、 より好ましくは 19〜23ヌクレオチドを含む DNA配列、 さらに好まし い例は、 配列番号 5〜15、 19、 .22、 25、 28、 33〜132によって示される DNA配列で ある。 In a preferred embodiment of the invention, the RNAi nucleic acid is a continuous 19-30 nucleotides, preferably 19-25, from the sequence of the mRNA or variant thereof encoded by the DRSF gene comprising the nucleotide sequence of SEQ ID NO: 2. An siRNA comprising a nucleotide, more preferably a 19-23 nucleotide sense strand sequence and its complementary sequence, the antisense strand sequence. Here, the siRNA can suppress the expression of the DREF gene or mRNA in vivo or in vitro. A preferred example of the corresponding DNA sequence comprising the siRNA is a contiguous 19-30 nucleotide, preferably 19-25 nucleotides derived from the nucleotide sequence encoding the CR2 domain or CR3 domain of DREF (SEQ ID NOs: 3 and 4, respectively). More preferably, the DNA sequence comprising 19 to 23 nucleotides, more preferably, the nucleotide sequence of SEQ ID NO: 2, positions 1,300 to 1,500 (0RF starting codon ATG "A" is the first position 1999 to 1299), 1,900 to 2,000 (from 1699 to 1799 if the 0RF start codon ATG “A” is the first), 19-30 DNA sequences comprising nucleotides, preferably 19-25 nucleotides, more preferably 19-23 nucleotides, more preferred examples are shown by SEQ ID NOs: 5-15, 19, .22, 25, 28, 33-132 DNA sequence.
本発明において、 DREF RNAi核酸の配列は、 上記の配列に限定されず、 DREF遺 伝子、対応する mRNA又はその変異体のィンビボにおける機能を抑制する限り、例 えば、 配列番号 2のヌクレオチド配列において上記の配列番号 5〜15、 19、 22、 25、 28、 33〜132の备配列を 5'方向又は 3'方向に 1〜5塩基シフ トした配列も包 含されるし、或いは上記の特定の配列と異なる、例えば 1〜 3個の塩基が異なる、 配列であってもよい。 ' In the present invention, the sequence of the DREF RNAi nucleic acid is not limited to the above sequence, and for example, in the nucleotide sequence of SEQ ID NO: 2 as long as it suppresses the in vivo function of the DREF gene, the corresponding mRNA, or a variant thereof. This includes sequences prepared by shifting 1 to 5 bases in the 5 'direction or 3' direction of the sequences of SEQ ID NOs: 5 to 15, 19, 22, 25, 28, 33 to 132, or the above-mentioned specific The sequence may be different from the sequence of, for example, 1 to 3 bases. '
本発明の siRNAは、上に例示したような s iRNAの標的となる DREF配列に基づい て、 周知の化学合成技術を用いて合成することができる。 例えば、 固相ホスホア ミダイ ト法などの DNA合成技術を利用した DNA (/RNA)自動合成装置を使用して化' 学的に合成するか、 或いは、 s iRNA 関連の受託合成会社 (例えばフナコシ株式会 社、 Dharmacon社、 Ambion社など) に委託して合成することも可能である。 The siRNA of the present invention can be synthesized using a well-known chemical synthesis technique based on the DREF sequence that is the target of siRNA as exemplified above. For example, it can be synthesized chemically using an automated DNA (/ RNA) synthesizer using DNA synthesis technology such as solid phase phosphoamidite method, or a siRNA-related contract synthesis company (for example, Funakoshi Corporation) It is also possible to synthesize by outsourcing to companies such as Dharmacon and Ambion.
本発明の実施形態によれば、 本発明の. siRNA は、 その前駆体である二本鎖 RNA (shRNA)から、 細胞内 RNァーゼであるダイサー(Dicer)によるプロセシングを 介して誘導されてもよレ、。 According to an embodiment of the present invention, the siRNA of the present invention is a duplex that is a precursor thereof. It can also be derived from RNA (shRNA) via processing by the intracellular RNase Dicer.
shRNAは、 siRNAのセンス鎖配列とアンチセンス鎖配列との間にループを有する 二本鎖 RNAであり、好ましくはその 3 '末端に 1〜 6個、好ましくは 2〜4個のポ リ Uからなるオーバーハングを含む。 shRNAは、 RNァーゼ IIIファミリーに属す るダイサ一によつて siRNAにプロセシングされたのち、 siRNAがー本鎖化され、 そのアンチセンス鎖 RNAが RNA切断活性をもつ分子等と複合体 RISC (RNA- Induced Silencing Complex)を形成し、 これによつて siRNA配列に相補的な配列を持つ標 的 mRNAが切断され、 その結果、 DREF遺伝子の発現が抑制される。 shRNA is a double-stranded RNA having a loop between the sense strand sequence and the antisense strand sequence of siRNA, preferably from 1 to 6, preferably 2 to 4 poly U at its 3 ′ end. Including overhangs. shRNA is processed into siRNA by a dicer belonging to the RNase III family, and then siRNA is made into a single strand, and its antisense strand RNA is complexed with a molecule having RNA cleavage activity, etc. RISC (RNA- Induced Silencing Complex), which cleaves the target mRNA having a sequence complementary to the siRNA sequence, thereby suppressing the expression of the DREF gene.
したがって、 本発明の上記 siRNA及びその前駆体 shRNAはいずれも、 本発明の 組成物の有効成分として使用することができる。 · Therefore, both the above siRNA of the present invention and its precursor shRNA can be used as an active ingredient of the composition of the present invention. ·
選択された RNAi核酸は、臨床使用の際にいわゆるオフターゲット(off- target) 作用を示さないことが望ましい。 オフターゲット作用とは、 標的 m伝子以外に、 使用した RNAi 核酸に部分的にホモロジ一のある別の遺伝子の発現を抑制する作 用をいう。 オフターゲット作用を避けるために、 候補 RNAi核酸について、 予めジ ーンチップなどを利用して交差反応がないことを確認する力、或いは GenBank (米 国 NCBI)などのデータベースを利用して配列同一性のない又はほとんどないこと を確認することが必要である。 しかし幸いにも、 ヒ ト DREFは、 ヒ トにおいて、 正 常細胞で実質的に発現されない一方で、 癌細胞で有意に発現されるため、 ォフタ ーゲッ ト作用の問題は回避できると予想される。 It is desirable that the selected RNAi nucleic acid does not exhibit a so-called off-target action in clinical use. Off-target action refers to the action of suppressing the expression of another gene that is partially homologous to the RNAi nucleic acid used in addition to the target m gene. To avoid the off-target effect, the candidate RNAi nucleic acid has the ability to confirm that there is no cross-reaction using a gene chip in advance, or there is no sequence identity using a database such as GenBank (NCBI) Or it is necessary to confirm that there is little. Fortunately, however, human DREF is not substantially expressed in normal cells in humans, but is significantly expressed in cancer cells, so it is expected that the problem of aftergetting can be avoided.
本発明の RNAi核酸を患者の体内に導入するときには、核酸を患部に直接注入す る力、又は核酸の発現が可能なベクターを使用することが好ましい。或いは、 siRNA 又はベクターを、 リボソーム、 例えばリポフエクタミン、 リポフエクチン、 セル フエクチン、 他の正電荷リボソーム (例えば、 正電荷コレステロール)、 又はマイ グロカプセル、 と複合体形成し、 この複合体を使用することもできる(例えば、 中 西守ら, 蛋白質 核酸 酵素, 44卷 11号, 48〜54頁, 1999年; Clinical Cancer research 59 : 4325-4333, 1999; Wu ら, J. Biol. Chem. 262 : 4429, 1987)。 哺孚し 動物細胞の細胞膜は負電荷を帯びているため、 正電荷リボソームが好ましく使用 される。正電荷リボソーム-核酸複合体は、工ンドサイ トーシスにより細胞内に取 り込まれたのち、 核酸はさらに細胞質又は核へ移行することができる。 或いは、本発明の治療用 RNAi核酸を、粒径約 500nm以下のナノ粒子中に封入す ることもできる。 ナノ粒子として、 例えば B型肝炎ウィルスエンベロープ L粒子 から形成されるホロ一ナノ粒子(hollow nanoparticles)が例示され、 核酸は、 ェ レク トロポレーシヨンによってこの粒子内に封入される。 この核酸封入粒子は、 血中に投与されるとき肝臓に送達されうる (T. Yamada ら, Nature Biotech 21 (8) : 885-890, 2003) ため、 肝癌の治療に有用であろう。 When introducing the RNAi nucleic acid of the present invention into a patient's body, it is preferable to use a vector capable of directly injecting the nucleic acid into the affected area or capable of expressing the nucleic acid. Alternatively, siRNA or vectors can be complexed with ribosomes such as lipofectamine, lipofectin, selfectin, other positively charged ribosomes (eg, positively charged cholesterol), or microcapsules, and this complex can be used. (For example, Nakanishi Mamoru et al., Protein Nucleic Acid Enzyme, 44-11, 48-54, 1999; Clinical Cancer research 59: 4325-4333, 1999; Wu et al., J. Biol. Chem. 262: 4429, 1987) . Since the cell membrane of mammals is negatively charged, positively charged ribosomes are preferably used. The positively charged ribosome-nucleic acid complex is taken up into the cell by engineered cytosis. After being inserted, the nucleic acid can be further transferred to the cytoplasm or nucleus. Alternatively, the therapeutic RNAi nucleic acid of the present invention can be encapsulated in nanoparticles having a particle size of about 500 nm or less. Examples of nanoparticles include hollow nanoparticles formed from, for example, hepatitis B virus envelope L particles, and nucleic acids are encapsulated in the particles by electroporation. This nucleic acid-encapsulated particle can be delivered to the liver when administered into the blood (T. Yamada et al., Nature Biotech 21 (8): 885-890, 2003) and may be useful in the treatment of liver cancer.
或いは、本発明の治療用 RNAi核酸をリポソ一ム中に封入するときには、核酸を 硫酸プロタミンで処理して凝縮を起こし核酸一タンパク質複合体としたのち、 正 電荷脂質又は高分子ミセル中に封入することもできる。 Alternatively, when encapsulating the therapeutic RNAi nucleic acid of the present invention in a liposome, the nucleic acid is treated with protamine sulfate to cause condensation to form a nucleic acid-protein complex, which is then encapsulated in a positively charged lipid or polymer micelle. You can also
•リポソーム-核酸複合体は、例えば逆相蒸発法 (F: Szokaら, Biochim. Biophys. Acta, 601 : 559, 1980)、 ボノレテックス振とう法、 カルシウム融合- EDTA キレート 法 (金田安史,実験医学 22卷 14号 (増刊) , 14ァ〜152頁, 2004年) などの方法 によって製造することができる。 • Liposome-nucleic acid complexes can be obtained, for example, by the reverse phase evaporation method (F: Szoka et al., Biochim. Biophys. Acta, 601: 559, 1980), Bonoretex shaking method, Calcium fusion-EDTA chelate method (Yasufumi Kaneda, Experimental Medicine 22卷 No. 14 (extra number), pages 14-152, 2004).
本発明の s iRNAやベクターなどの核酸を標的癌細胞の中に導入する別の方法は、 マイクロインジェクション法、 ウィルスベクター法などがある。 Other methods for introducing a nucleic acid such as the siRNA or vector of the present invention into a target cancer cell include a microinjection method and a viral vector method.
マイクロインジヱクション法は、 微細な注入針で核酸を細胞内に直接顕微注入 する方法である。 The microinjection method is a method in which a nucleic acid is directly microinjected into a cell with a fine injection needle.
ウィルスベクター法は、 ウィルスベクターを用いて核酸を細胞内に導入する方 法であって、 siRNA、 shRNA又は miRNAを発現する発現ュニットを組み込んだ組換 えウィルスを作製し、.細胞に感染させて細胞内で発現させる方法である。 ウィル スの例は、 アデノウイルス、 アデノ随伴ウィルス、 レトロウィルスなどを含む。 本発明の実施形態により、本発明で使用可能な核酸として、前記 siRNAや shRNA などの RNAi核酸をコードする DNA配列をプロモーターの調節下に含む発現べクタ 一が含まれる。 The viral vector method is a method of introducing a nucleic acid into a cell using a viral vector, in which a recombinant virus incorporating an expression unit that expresses siRNA, shRNA or miRNA is prepared, and the cell is infected. It is a method of expressing in cells. Examples of viruses include adenovirus, adeno-associated virus, retrovirus and the like. According to an embodiment of the present invention, a nucleic acid that can be used in the present invention includes an expression vector comprising a DNA sequence encoding an RNAi nucleic acid such as siRNA or shRNA under the control of a promoter.
発現ベクターの 1つの例は、 ヘアピン型ベクターである。 このベクターは、 前 記センス鎖 RNA配列と前記ァンチセンス鎖 RNA配列とがー本鎖ループ配列を介し て共有結合されているヘアピン型 RNAをコードする DNAを含み、ここで該 DNAは、 細胞内で転写により該ヘアピン型 RNAを形成し、 ダイサ一によりプロセシングさ れて前記 siRNAを形成するベクターである。 One example of an expression vector is a hairpin vector. This vector includes DNA encoding a hairpin RNA in which the sense strand RNA sequence and the antisense strand RNA sequence are covalently bonded via a single-stranded loop sequence, wherein the DNA is intracellularly contained. The hairpin RNA is formed by transcription and processed by a dicer. A vector that forms the siRNA.
siRNAをコードするヘアピン型 DNAの 3,末端には、転写停止シグナル配列とし て、 '或いはオーバーハングのために、 1〜6個、 好ましくは 1〜 5個の Tからな るポリ T配列、 例えば 4個又は 5個のポリ T配列が連結される。 ベクター DNAか ら転写された siRNA前駆体としてのショートヘアピン RNA (shRNA)は、 そのアンチ センス鎖の 3,末端に 2〜4個の U からなるオーバーハングを有することが望ま しく、 オーバーハングの存在によって、 センス鎖 RNA及ぴアンチセンス鎖 RNAは ヌクレアーゼによる分解に対して安定性を増すことができる。 ヒトには内在性の ダイサ一が 1つ存在し、 これが長鎖 dsRNAや前駆体 miRNAをそれぞれ siRNAと成 熟 miRNAに変換する役割をもつ。 At the end of the hairpin DNA encoding siRNA, a poly-T sequence consisting of 1 to 6, preferably 1 to 5, T for transcription termination signal sequence, or for overhanging, for example, Four or five poly T sequences are linked. Short hairpin RNA (shRNA) as a siRNA precursor transcribed from vector DNA should have an overhang consisting of 3 to 4 U at the end of its antisense strand. By this, sense strand RNA and antisense strand RNA can be more stable against nuclease degradation. There is one endogenous dicer in humans, which has the role of converting long dsRNA and precursor miRNA into siRNA and mature miRNA, respectively.
.本発明における前記ループ配列の例は、 5' - UUCAAGAGA- 3' (配列番号 135)、 5' -CUUCCUGUCA-3' (配列番号 136)、 5' - UUCCAG - 3' (配列番号 137) などであるが、 これらに限定されない。 Examples of the loop sequence according to the present invention include 5'-UUCAAGAGA-3 '(SEQ ID NO: 135), 5'-CUUCCUGUCA-3' (SEQ ID NO: 136), 5'-UUCCAG-3 '(SEQ ID NO: 137), etc. However, it is not limited to these.
. 本発明の RNAi核酸のなかで特に好ましい RNAi#3、 RNAi#4、 RNAi#5及び RNAi#6 を発現するためのベクターに組み込まれる DNA配列の組み合わせはそれぞれ、 下 記の配列番号 20と 21、 配列番号 23と 24、 配列番号 26と 27、 配列番号 29と 30 である。 なお、 下線は、 siRNA のセンス鎖及びアンチセンス鎖に対応する配列を 示す。 The combinations of DNA sequences incorporated into vectors for expressing RNAi # 3, RNAi # 4, RNAi # 5 and RNAi # 6, which are particularly preferred among the RNAi nucleic acids of the present invention, are shown in SEQ ID NOs: 20 and 21, respectively. SEQ ID NO: 23 and 24, SEQ ID NO: 26 and 27, SEQ ID NO: 29 and 30. The underline indicates the sequence corresponding to the sense strand and antisense strand of siRNA.
RNAi#3用 oligo (それぞれ、 配列番号 20及び 21) : ' RNAi # 3 oligo (SEQ ID NOS: 20 and 21, respectively): '
RNAi #4用 oligo (それぞれ、 配列番号 23及び 24) : RNAi # 4 oligo (SEQ ID NOS: 23 and 24, respectively):
RNAi#5用 oligo (それぞれ、 配列番号 26及び 27) : RNAi # 5 oligo (SEQ ID NOS: 26 and 27, respectively):
si5F: 5 , -TACCCCACCATCAGCATGGTCTTCCTGTCAACCATGCTGATGGTGGGGTACTTTTTG-3 ' si5F: 5, -TACCCCACCATCAGCATGGTCTTCCTGTCAACCATGCTGATGGTGGGGTACTTTTTG-3 '
RNAi#6用 oligo (それぞれ、 配列番号 29及ぴ 30) : RNAi # 6 oligo (SEQ ID NO: 29 and 30 respectively):
si6F: 5 -( si6R : 3,- 本発明の上記発現ベクターは、上記ヘアピン型 DNAの 5,側にプロモーターを含 むこ.とができる。 プロモーターの例は、 pol III プロモーター、 例; ίばヒ トもし くはマウス由来の U6プロモーター又は HIプロモーター、 pol IIプロモーター、 或いはサイ トメガロウイノレスプロモーターである。 si6F: 5-( si6R: 3,-The expression vector of the present invention can contain a promoter on the 5th side of the hairpin DNA. Examples of promoters are pol III promoters, eg, U6 promoter or HI promoter from human or mouse, pol II promoter, or cytomegaloinoles promoter.
,本発明の発現ベクターの別の例は、 タンデム型ベクターである。 このベクター は、 前記 s iRNAを構成するセンス鎖 RNA配列をコードする DNA配列とァンチセン ス鎖 RNA配列をコードする DNA配列とを連続して含み、かつ各鎖の 5 '末端にプロ モーターが、 また各鎖の 3 '末端にポリ T配列がそれぞれ連結された DNAを含み、 ここで該 DNAは、 細胞内で転写後に該センス鎖 RNAと該アンチセンス鎖 RNAとが ハイブリダィズして前記 siRNAを形成するベクターである。 Another example of the expression vector of the present invention is a tandem vector. This vector comprises a DNA sequence encoding the sense strand RNA sequence constituting the siRNA and a DNA sequence encoding the antisense strand RNA sequence, and a promoter at the 5 ′ end of each strand, DNA comprising a poly T sequence linked to the 3 'end of each strand, wherein the DNA hybridizes with the sense strand RNA and the antisense strand RNA after transcription in the cell to form the siRNA It is a vector.
タンデム型ベクターにおけるプロモーターの例は、 pol HI プロモーター、 例 えばヒ トもしくはマウス由来の U6プロモーター又は HIプロモーター、 或いはサ イ トメガロウイノレスプロモーターである。 An example of a promoter in a tandem vector is a pol HI promoter, such as a human or mouse-derived U6 promoter or HI promoter, or a site megaloinoles promoter.
また、ポリ T酉己歹 IJは、 :!〜 6個、好ましくは 1〜 5個の Tからなるポリ T配列、 例えば 4個又は 5個のポリ T配列である。 Also, Poly T 酉 己 歹 IJ :! A poly T sequence consisting of ˜6, preferably 1-5 T, such as 4 or 5 poly T sequences.
本発明のタンデム型ベクターは、 細胞内に導入されたのち、 センス鎖とアンチ センス鎖に相当する RNAに転写され、 互いにハイブリダイズして目的の siRNAを 生成することができる。 ' The tandem vector of the present invention can be introduced into a cell, transcribed into RNA corresponding to the sense strand and the antisense strand, and hybridized with each other to produce the desired siRNA. '
本発明の実施形態により、 上記ヘアピン型及ぴタンデム型ベクターは、 プラス ミ ドベクター又はウイノレスベクターである。 According to an embodiment of the present invention, the hairpin type and tandem type vector is a plasmid vector or a winores vector.
プラスミ ドベクターは、 後述の実施例に記載の手法又は文献記載の方法を用い て調製してもよいし、 或いは市販のベクター系、 たとえば piGENE™U6系ベクター 及び piGENE™Hl系べクター(タカラバイォ株式会社)を利用することもできる(Τ· R. The plasmid vector may be prepared using the methods described in the Examples below or the methods described in the literature, or commercially available vector systems such as the piGENE ™ U6 vector and the piGENE ™ Hl vector (Takarabai Bio Inc.). Company) (Τ · R.
Brummelkampら, Science (2002) , 296 : 550 - 553 ; N. S. Lee ら, Nature Biotech.Brummelkamp et al., Science (2002), 296: 550-553; N. S. Lee et al., Nature Biotech.
(2002), 20 : 500— 505 ; Μ· Miyagishi ら, Nat. Biotechnol. (2002) , 20 : 497—500 ; P. J.(2002), 20: 500-505; Tsuji Miyagishi et al., Nat. Biotechnol. (2002), 20: 497-500; P. J.
Paddisonら, Genes & Dev. (2002), 16 : 948-958 ; T. Tusch, Nature Biotech (2002) ,Paddison et al., Genes & Dev. (2002), 16: 948-958; T. Tusch, Nature Biotech (2002),
20 : 446-448; C. P. Paul ら, ature Biotech. (2002) , 20 : 505 - 508 ; 多比良和誠ら 編、 RNAi実験プロ トコル、 羊土社、 2003年)。 プラスミ ドベクターは一般に、 本発明の siRNAをコードする DNA配列及びプロ モーターの他に、 薬剤耐性遺伝子 (例えば、 ネオマイシン耐性遺伝子、 アンピシ リン耐性遺伝子、 ピューロマイシン耐性遺伝子、 ハイグロマイシン耐性遺伝子な ど)、 転写停止配列、 ユニーク制限部位もしくはマルチプルクロー-ングサイ ト、 複製開始点などを含むことができる。 20: 446-448; CP Paul et al., Ature Biotech. (2002), 20: 505-508; edited by Yoshikazu Tahira, RNAi Experimental Protocol, Yodosha, 2003). In general, a plasmid vector is a drug resistance gene (eg, neomycin resistance gene, ampicillin resistance gene, puromycin resistance gene, hygromycin resistance gene) in addition to the DNA sequence and promoter encoding the siRNA of the present invention. It may contain transcription termination sequences, unique restriction sites or multiple cloning sites, and replication origins.
ウィルスベクターは、 たとえばアデノウイルスベクター、 アデノ随伴ウィルス ベクター、 レンチウイノレスベタター、 レトロウイノレスベクター (白血病ウィルス ベクターなど)、ヘルぺスウィルスベクターなどを使用することができる。 ウィル スベクターは、 ヒ トに使用する際に疾病を引き起こさないように例えば自己複製 能を欠損したタイプのものが好ましい。 たとえばアデノウィルスベクターの場合 には、 E1遺伝子及び E3遺伝子を欠失した自己複製能欠損型アデノウィルスべク ター (例えば Invitrogen社の pAdeno-Χ)を使用することができる。 ウィルスべク ターの構築は、文献記載の方法を利用することができる(米国特許第 52524ァ9号、 国際公開 W094/13788など)。 '、 As the virus vector, for example, an adenovirus vector, an adeno-associated virus vector, a lentiwinores betater, a retrowinoles vector (such as a leukemia virus vector), a herpes virus vector, or the like can be used. The virus vector is preferably of a type lacking self-replicating ability, for example, so as not to cause disease when used in humans. For example, in the case of an adenovirus vector, a self-replication ability-deficient adenovirus vector lacking the E1 gene and E3 gene (eg, pAdeno-Χ from Invitrogen) can be used. Construction of viral base click terpolymer can utilize literature methods (U.S. Patent No. 525 2 4 § 9 No., and International Publication W094 / 13788). ',
本発明のプラスミ ドベクターは、 上記のとおり、 例えばリポフエクタミン、 リ ポフエクチン、 セルフエクチン、 又は正電荷コレステロールなどの正電荷リポソ ームと複合体を形成し力プセル化された状態で患者の体内に導入することができ る (中西守ら, 上記; Wu ら, 上記)。 また、 ウィルスベクターは患部に導入し細 胞感染させることによって細胞内に遺伝子導入することができる(L. Zender ら, Proc. Natl. Acad. Sc i. USA (2003) , 100 : 77797-7802 ; H. Xia ら, Nature Biotech. (2002), 20 : 1006—101.0 ; X. F. ' Qin ら, Proc. Natl. Acad. Sci. USA (2003), 100 : 183—188 ; G. M. Barton ら, Proc. Natl . Acad., Sc i . USA (2002) , 99 : 14943-14945; J. D. Homme 1 Nature Med. (2003), 9 : 1539—1544)。 特にアデノ ウィルスベクター又はアデノ随伴ウィルスベクターは種々の細胞種に非常に高い 効率で遺伝子導入可能であることが確認されている。 このベクターはまた、 ゲノ ム中に組み込まれることがないため、 その効果は一過性であり安全性も他のウイ ルスベクターと比べて高いと考えられる。 As described above, the plasmid vector of the present invention forms a complex with a positively charged liposome such as lipophectamine, lipofectin, self-actin, or positively charged cholesterol, and is introduced into the patient's body in a force-pellated state. (Mr. Nakanishi et al., Supra; Wu et al., Supra). In addition, viral vectors can be introduced into cells by introducing them into affected areas and infecting cells (L. Zender et al., Proc. Natl. Acad. Sci. USA (2003), 100: 77797-7802; H. Xia et al., Nature Biotech. (2002), 20: 1006-101.0; XF 'Qin et al., Proc. Natl. Acad. Sci. USA (2003), 100: 183-188; GM Barton et al., Proc. Natl. Acad., Sci. USA (2002), 99: 14943-14945; JD Homme 1 Nature Med. (2003), 9: 1539-1544). In particular, it has been confirmed that adenovirus vectors or adeno-associated virus vectors can introduce genes into various cell types with very high efficiency. Since this vector is also not incorporated into the genome, its effect is transient and is considered to be safer than other viral vectors.
本発明のベクターに組み込まれる siRNAをコードする DNAは、 s iRNAを構成す るセンス鎖配列とアンチセンス鎖配列とがー本鎖ループ配列を介して共有結合さ れているヘアピン型 RNAをコードし、 かつ細胞内で転写により該ヘアピン型 RNA を形成し、 ダイサ一によりプロセシングされて該 siRNAを形成するものである。 本発明のベクターに組み込まれる siRNA をコードする DNAの別の例は、 siRNA を構成するセンス鎖配列をコードする DNA配列とアンチセンス鎖配列をコードす る DNA配列とを連続して含み、 かつ各鎖の 5'末端にプロモーターが、 また各鎖の 3'末端にポリ T配列がそれぞれ連結されており、 該 DNAは、 細胞内で転写後に該 センス鎖 RNAと該アンチセンス鎖 RNAとがハイブリダイズして前記 s iRNAを形成 するものである。 In the DNA encoding siRNA incorporated into the vector of the present invention, the sense strand sequence and the antisense strand sequence constituting the siRNA are covalently linked via a single strand loop sequence. The hairpin RNA is encoded, and the hairpin RNA is formed by transcription in a cell and processed by a dicer to form the siRNA. Another example of DNA encoding siRNA incorporated into the vector of the present invention comprises a DNA sequence encoding a sense strand sequence constituting siRNA and a DNA sequence encoding an antisense strand sequence, and A promoter is linked to the 5 'end of each strand, and a poly T sequence is linked to the 3' end of each strand, and the sense strand RNA and the antisense strand RNA hybridize after transcription in the cell. Thus, the siRNA is formed.
1. 2 アンチセンス核酸 1.2 Antisense nucleic acid
本発明の組成物の有効成分としての別の核酸は、 アンチセンス核酸である。 こ のアンチセンス核酸は、 配列番号 2 (202位〜 2286位) のヌクレオチド配列を含 む DREF遺伝子に対応する mRNAの配列、 又はその部分配列、 に相補的な配列を含 む RNA力、 或いは配列番号 2 ( 2位〜2286位) のヌクレオチド配列、 又はその 部分配列、 に相捕的な配列を含む DNAのいずれかである。、 Another nucleic acid as an active ingredient of the composition of the present invention is an antisense nucleic acid. This antisense nucleic acid is an RNA sequence containing a sequence complementary to the mRNA sequence corresponding to the DREF gene containing the nucleotide sequence of SEQ ID NO: 2 (positions 202 to 2286), or a partial sequence thereof, or a sequence. Either the nucleotide sequence of number 2 (positions 2 to 2286 ), or a partial sequence thereof, and DNA containing a complementary sequence. ,
前記部分配列は、 DREF遺伝子又は mRNAの配列において連続する約 30以上、 50 以上、 70以上、 100以上、 150以上、 200以上又は 250以上から全長以下、 例えば 50〜150、 のヌクレオチドからなる配列を含むことができる。 The partial sequence is a sequence consisting of about 30 or more, 50 or more, 70 or more, 100 or more, 150 or more, 200 or more, or 250 or more and a full length of, for example, 50 to 150 nucleotides in the DREF gene or mRNA sequence. Can be included.
アンチセンス核酸のヌクレオチドは、 天然のヌクレオチドに加えて、 ハロゲン In addition to natural nucleotides, antisense nucleic acid nucleotides
(フッ素、 塩素、 臭素又はョゥ素)、 メチル、 カルボキシメチル又はチォ.基などの 基を有する修飾ヌクレオチドを含むことができる。 It can contain modified nucleotides having groups such as (fluorine, chlorine, bromine or iodine), methyl, carboxymethyl or thio groups.
アンチセンス核酸は、 周知の DNA/RNA合成技術又は DNA組換え技術を用いて合 成することができる。 DNA 組換え技術によって合成する場合、 配列番号 2の配列 を含むベクター DNA を鏡型にして、 増幅しょうとする配列を挾み込むプライマー を用いてポリメラーゼ連鎖反応 (PCR) を行って標的配列を増幅し、 必要に応じて ベクター中にクローユングして、 アンチセンス DNAを生成することができる。 或 いは、 このようにして得られた増幅標的配列を有する DNAをベクターに挿入し、 該ベクターを真核又は原核細胞に導入し、 その転写系を利用してアンチセンス Antisense nucleic acids can be synthesized using well-known DNA / RNA synthesis techniques or DNA recombination techniques. When synthesizing by DNA recombination technology, the vector DNA containing the sequence of SEQ ID NO: 2 is mirrored, and the target sequence is amplified by polymerase chain reaction (PCR) using primers that squeeze the sequence to be amplified. If necessary, it can be cloned into a vector to produce antisense DNA. Alternatively, the DNA having the amplified target sequence thus obtained is inserted into a vector, the vector is introduced into a eukaryotic or prokaryotic cell, and the transcription system is used for antisense.
RNA を得ることができる。 ベクターは、 上に記載のウィルスベクター、 プラスミ ドベクターを使用するこ.とができる。 . - 本発明のアンチセンス核酸は、それが DNAであっても RNAであっても、 DREF mRNA に結合することによって、 タンパク質への翻訳を抑制することができる。 RNA can be obtained. As the vector, the above-described virus vector or plasmid vector can be used. - Whether the antisense nucleic acid of the present invention is DNA or RNA, it can suppress translation into protein by binding to DREF mRNA.
本発明のアンチセンス核酸を患者に送達するために、 アンチセンス核酸を、 上 述したような正電荷リボソーム等のリポソームに封入してもよいし、 或いはアン チセンス核酸を、 例えば強力な pol II又は pol IIIプロモーターの調節下にある ようにベクター(上述のプラスミ ド又はウィルスベクター)に組み込んでもよい。 1. 3 In order to deliver the antisense nucleic acid of the present invention to a patient, the antisense nucleic acid may be encapsulated in a liposome such as a positively charged ribosome as described above, or the antisense nucleic acid may be, for example, a strong pol II or It may be incorporated into a vector (the above-mentioned plasmid or viral vector) so as to be under the control of the pol III promoter. 13
本発明はさらに、配列番号 1のアミノ酸配列を含む DREFタンパク質又はその変 異体のインビボ機能を抑制する抗体又はそのフラグメントを含む、 癌治療用組成 物を提供する。 The present invention further provides a composition for treating cancer comprising an antibody or a fragment thereof that suppresses the in vivo function of a DREF protein comprising the amino acid sequence of SEQ ID NO: 1 or a variant thereof.
本発明の前記標的タンパク質は、 DREF遺伝子によってコードされるタンパク質 であり、 ヒ ト癌の細胞增殖や細胞周期進行に関与するタンパク質である。 したが つて、 癌細胞内に発現された該タンパク質の機能を阻害又は抑制することによつ て、 結果として癌の増殖の抑制や細胞死に導くことができる。 この目的のための 薬剤は、 DREFタンパク質又はその部分に対する抗体である。 The target protein of the present invention is a protein encoded by the DREF gene, and is a protein involved in human cancer cell proliferation and cell cycle progression. Therefore, inhibition or suppression of the function of the protein expressed in cancer cells can result in suppression of cancer growth and cell death. A drug for this purpose is an antibody against the DREF protein or part thereof.
好ましくは、 DREFに対する抗体は、 DREFの CR1 ドメィン、 CR2 ドメィン又は CR3 ドメイン (それぞれ配列番号 135、 133又は 134)、 或いはその部分に対する抗体 である。 特に好ましい抗体の例は、 DREF 上の DRE (DNA repl ication-related element)結合領域に対する抗体である。 DREFの DNA結合ドメインは 「BEDジンク フィンガー」 と呼ばれ、 亜鉛イオン (一個) をもつ蛋白構造を有する。 このドメ ィンは CR1内の一部であり、配列番号 1の DREFァミノ酸配列の 20番目〜 74番目 の配列(配列番号 140)を含む。 DREFのこのドメィンを含む領域に対する抗体は、 DREFの DNA結合を阻害し、 その結果 DREFのインビポ機能を阻害することができ る。 Preferably, the antibody against DREF is an antibody against the CR1 domain, CR2 domain or CR3 domain of DREF (SEQ ID NO: 135, 133 or 134, respectively), or a portion thereof. An example of a particularly preferred antibody is an antibody against a DRE (DNA replication-related element) binding region on DREF. The DNA-binding domain of DREF is called “BED zinc finger” and has a protein structure with one zinc ion. This domain is part of CR1 and contains the 20th to 74th sequence (SEQ ID NO: 140) of the DREF amino acid sequence of SEQ ID NO: 1. Antibodies to the domain containing this domain of DREF can inhibit DREF DNA binding and consequently inhibit DREF's in vivo function.
BED .ジンクフィンガーァミノ酸配列 (配列番号 140) : BED. Zinc finger amino acid sequence (SEQ ID NO: 140):
RAKSKVWKYFGFDTNAEGCILQWKKIYCRICMAQIAYSGNTSNLSYHLEK丽 PEE RAKSKVWKYFGFDTNAEGCILQWKKIYCRICMAQIAYSGNTSNLSYHLEK 丽 PEE
また、 別の DNA結合に関連するドメインは、 DREFの CR3内の一部であり、 配列 番号 1の DREFァミノ酸配列の 571番目〜651番目の配列(配列番号 141)を含む。 このドメィンは、ァクチベータース一ハ°ーファミ リ一 (hAT element superf amily) と呼ばれ、 二量体开成ドメイン (dimerisation domain) である。 DREF のこのド メインを含む領域に対する抗体は、 DREFの二量体形成を阻害するため、 結果とし て DREFの DNA結合が阻害され、 DREFのインビボ機能が阻害されうる。 Another domain associated with DNA binding is a part of CR3 of DREF, and includes the 571st to 651th sequence (SEQ ID NO: 141) of the DREF amino acid sequence of SEQ ID NO: 1. This domain is an activator's family (hAT element superf amily) Called the dimerization domain. Antibodies to the region of DREF containing this domain inhibit DREF dimer formation, resulting in inhibition of DREF DNA binding and inhibition of DREF in vivo function.
二 量 体 形 成 ド メ イ ン の ア ミ ノ 酸 配 列 ( 配 列 番 号 141 ) : Amino acid sequence of the dimerized domain (sequence number 141):
EQVFLYENA EQVFLYENA
本発明に有用な抗体の作製については、下記の CR1、 CR2及び CR3 ドメインのァ ミノ酸配列及びヌクレオチド配列を利用して、 公知のぺプチド合成技術又は DNA 組換え技術を用いて (ポリ) ペプチドを合成し目的の抗体を作製することができ る。 For the production of an antibody useful in the present invention, the amino acid sequence and nucleotide sequence of the following CR1, CR2, and CR3 domains are used to make (poly) peptides using known peptide synthesis techniques or DNA recombination techniques. Can be synthesized to produce the desired antibody.
CR1のアミノ酸配列 (配列番号 138) : CR1 amino acid sequence (SEQ ID NO: 138):
CR1のヌクレオチド配列 (配列番号 139) The nucleotide sequence of CR1 (SEQ ID NO: 139)
CAGGAGCTGACGGCCGCCGTGCTGGGCCTCATCTGCGAGGGGCTGTACCCA CR2のアミノ酸配列 (配列番号 133) : CAGGAGCTGACGGCCGCCGTGCTGGGCCTCATCTGCGAGGGGCTGTACCCA CR2 amino acid sequence (SEQ ID NO: 133):
EDSNNHHLMLEASEWATIEGLVELLQPFKQVAEIV EVIAKELSKTYQETPEIDMFLNVATFLDPRYKRLPFLSAFERQQVENRVVEEAKGLLD CR2のヌクレオチド配列 (配列番号 3 ) : GTGGTGGAAGAGGCCAAGGGCCTGCTGGAC EDSNNHHLMLEASEWATIEGLVELLQPFKQVAEIV EVIAKELSKTYQETPEIDMFLNVATFLDPRYKRLPFLSAFERQQVENRVVEEAKGLLD CR2 nucleotide sequence (SEQ ID NO: 3): GTGGTGGAAGAGGCCAAGGGCCTGCTGGAC
CR3のァミノ酸配列(配列番号 134) : CR3 amino acid sequence (SEQ ID NO: 134):
FFGI FFGI
CR3のヌクレオチド配列 (配列番号 4 ) : CR3 nucleotide sequence (SEQ ID NO: 4):
TTCTTTGGCATT TTCTTTGGCATT
このような抗体には、 ポリクローナル抗体、 モノクロ—ナル抗体、 組換え産生 抗体、 ヒ ト抗体、 ヒ ト化抗体、 キメラ抗体、 単鎖抗体、 Fab、 F (ab' ) 2、 scFv、 Fv、 二重特異抗体、 合成抗体などが含まれる。 抗体の特性及び構造については、 小闋 至ら,実験医学 22巻 14号 (増刊) 125〜: 130頁, 2004年、 羊土社 (東京、 日本) に記載されており、 その開示は本発明のために使用できる。 Such antibodies include polyclonal antibodies, monoclonal antibodies, recombinantly produced antibodies, human antibodies, humanized antibodies, chimeric antibodies, single chain antibodies, Fab, F (ab ') 2 , scFv, Fv, two These include bispecific antibodies and synthetic antibodies. The characteristics and structure of the antibody are described in Toru Otsuki et al., Experimental Medicine Vol. 22 No. 14 (Extra) 125-: 130, 2004, Yodosha (Tokyo, Japan). Can be used for.
本発明での使用に適する好ましい抗体は、 アナフィラキシーによる副作用を全 く又はほとんど起こさないヒ ト抗体又はヒ ト化抗体、 特にヒ ト又はヒ ト化モノク 口一ナル抗体である。 .また、 抗体のクラス、 サブクラスは任意のタイプのもので よい。 例えば、 抗体のタイプには、 IgGヽ IgMヽ IgEヽ IgD、 IgA、 IgGい IgG2、 IgG3、 IgG4、 IgAい IgA2が含まれる。 抗体はまた、 ぺグ化、 ァセチル化、 グリコシル化、 アミ ド化などによって誘導体化されていてもよい。 Preferred antibodies suitable for use in the present invention are human or humanized antibodies, particularly human or humanized monoclonal antibodies, that cause little or no side effects due to anaphylaxis. The antibody class and subclass may be of any type. For example, the type of antibody, IgGヽIgMヽIgEヽIgD, IgA, include IgG had I g G 2, IgG 3, IgG 4, IgA have IgA 2. An antibody may also be derivatized by pegylation, acetylation, glycosylation, amidation, and the like.
ヒ ト抗体は、例えばファージジスプレイライプラリ一(pharge display library) Human antibodies can be generated by, for example, phage display library (pharge display library).
¾ (T. C. Thomas ら, Mol. Immunol. 33 : 1389-1401, 1996)又はヒ ト抗体産生動物(例 えばマウス、有蹄類など)を用いる方法(I. Ishidaら, Cloning Stem Cell 4: 91- 102¾ (TC Thomas et al., Mol. Immunol. 33: 1389-1401, 1996) or methods using human antibody-producing animals (eg mice, ungulates, etc.) (I. Ishida et al., Cloning Stem Cell 4: 91- 102
2002)によって製造できる。 2002).
ヒ ト抗体産生マウスは、 例えば、 ヒ ト人工染色体にヒト抗体産生遺伝子を含む ヒト染色体断片を導入したのち、 ミクロセル法を用いて例えばマウス胚性幹細胞 ゲノムに人工染色体を組み込み、 仮親マウスの子宮に移植し、 キメラマウスを出 産し、 雌雄のキメラマウスの交配、 或いはキメラマウスと同種の野生型マウスと の交配によって、 ヒ ト抗体遺伝子を含み、 したがってヒ ト抗体の産生が可能であ る、ホモ型の子孫マウスを作出するなどの方法によって作製することができる(例 えば、 再表 02/092812、 国際公開 W0 98/24893、 国際公開 W0 96/34096など)。 こ のヒト抗体産生トランスジエニックマウスに、本発明の標的 DREFタンパク質を抗 原として免疫したのち、 脾臓を摘出し、 慣用技術によってこの脾臓細胞とマウス ミエローマ細胞とを融合してハイプリ ドーマを形成し、 目的のモノクローナル抗 体を作製することができる(G. Kohler及び C. Milstein, Nature 256 : 495-497, 1975)。 Human antibody-producing mice contain, for example, human antibody-producing genes in human artificial chromosomes After introducing the human chromosome fragment, using the microcell method, for example, an artificial chromosome is integrated into the genome of a mouse embryonic stem cell, transplanted into the uterus of a foster parent mouse, a chimeric mouse is born, a male or female chimeric mouse is bred, or a chimeric mouse And a wild-type mouse of the same species, for example, by producing a homozygous offspring mouse that contains a human antibody gene and is therefore capable of producing a human antibody (for example, Table 02/092812, International W0 98/24893, International W0 96/34096, etc.). This human antibody-producing transgenic mouse is immunized with the target DREF protein of the present invention as an antigen, and then the spleen is removed and fused with this spleen cell and mouse myeloma cell by conventional techniques to form a hybridoma. The desired monoclonal antibody can be prepared (G. Kohler and C. Milstein, Nature 256: 495-497, 1975).
ファージディスプレイ ·ライプラリー法は、 未処置のヒ トリンパ細胞から直接 入手した免疫グロブリン遺伝子のライブラリ一から、 目的の抗体をコードする DNAをスクリーニングし、 この DNAと、 抗体鎖との間に、、ファージ粒子を用いて 物理的会合を確立し、 これによつて、 標的に親和性をもつ抗体を提示するファー ジを親和性スクリーニングによって富化することを含む。 この方法を用いて、 標 的に対する結合親和性をもつ抗体を、 通常の手法によって大量に合成することが できる (例えば、 特表 20013-527832)。 In the phage display library method, DNA encoding the antibody of interest is screened from a library of immunoglobulin genes obtained directly from untreated human lymphocytes, and phage particles are placed between this DNA and the antibody chain. To establish a physical association, which involves enriching affinity-screened phage that present antibodies with affinity for the target. Using this method, an antibody having a binding affinity for a target can be synthesized in a large amount by an ordinary method (for example, JP 20013-527832).
ヒト化抗体は、例えば、 ヒ ト DREFタンパク質を免疫したマウスから作製した該 DREFに対するマウス抗体の相補性決定領域(CDR)を、 ヒ ト IgGに結合することに よって得ることができる。 このようなヒト化抗体は、 遺伝子組換え技術を用いる ことに作製可能である。抗体をヒト化する技術は、例えば米国特許第 6639055号、 同 5530101号などに記載されている。 ' 本発明において、 DREFタンパク質の変異体は、 ヒ ト個体内で自然発生的に生ず るすべての変異体を含み、 例えば多型性や突然変異に基づくような変異体、 或い は選択的スプライシングによる変異体を含む。 変異体は、 配列番号 1のアミノ酸 配列において、 1もしくは数個のアミノ酸が置換、 欠失又は付加された配列を有 し、 かつ癌の細胞増殖に関与するインビボ機能を有する。 本明細書において 「数 個」 とは、 10個以下、 8 ·個以下、 6個以下、 5個以下、 4個以下、 3個以下又は 2 個を意味する。 或いは、 変異体は、 配列番号 1のアミノ酸配列と 95%以上、 97% 以上、 98%以上又は 99%以上の%同一性を有する配列を含むものである。 本明細 書において'「0/0同一性」 は、 配列アラインメントにおいてギャップの導入が可能 な公知の BLASTプログラム(BL ASTX及び BLASTN)に基づいて決定されうる(例えば S. F. Altschul ら, J. Mol. Bio. 215 : 403 - 410, 1990など)。 A humanized antibody can be obtained, for example, by binding a human IgG complementarity determining region (CDR) produced from a mouse immunized with human DREF protein to human IgG. Such humanized antibodies can be produced using genetic recombination techniques. Techniques for humanizing antibodies are described, for example, in US Pat. Nos. 6639055 and 5530101. '' In the present invention, DREF protein variants include all naturally occurring variants in human individuals, such as variants based on polymorphisms or mutations, or selective Includes variants by splicing. The variant has a sequence in which one or several amino acids are substituted, deleted or added in the amino acid sequence of SEQ ID NO: 1, and has an in vivo function involved in cancer cell growth. In this specification, “several” means 10 or less, 8 · or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 Means individual. Alternatively, the variant comprises a sequence having% identity of 95% or more, 97% or more, 98% or more, or 99% or more with the amino acid sequence of SEQ ID NO: 1. As used herein '' 0/0 identity "may be determined based on the possible known BLAST program introduction of gaps (BL ASTX and BLASTN) In a sequence alignment (e.g. SF Altschul et al, J. Mol. Bio 215: 403-410, 1990, etc.).
本発明の抗体又はその断片は、 単独で、 或いは抗癌剤 (化学療法剤、 放射性物 質など) などの薬剤を結合させた形態で、 癌に対する治療剤として用いることが できる。 抗癌剤には、 例えばタキソール、 シタラビン、 シスプラチン、 エトポシ ド、 ドキソノレビシン、 ダウノノレビシン、 ビンブラスチン、 パクリタキセ^^などの 化学療法剤、 放射性インジウム、 テクネチウム、 イッテルビウムなどの放射性金 属などが含まれるが、 これらに限定されない。 抗体への抗癌剤の結合は、 例えば リンカーを介して共有結合によって、 或いは金属イオンとの配位結合によって、 抗体定常領域の任意の部位に抗癌剤を結合することを含む方法によって行うこと ができる。 The antibody or fragment thereof of the present invention can be used as a therapeutic agent for cancer alone or in a form to which a drug such as an anticancer drug (chemotherapeutic agent, radioactive substance, etc.) is bound. Anti-cancer agents include, but are not limited to, chemotherapeutic agents such as taxol, cytarabine, cisplatin, etoposide, doxonorevicin, daunonorevicin, vinblastine, paclitaxe ^^, and radioactive metals such as radioactive indium, technetium, and ytterbium. . Binding of the anticancer agent to the antibody can be performed by a method including binding of the anticancer agent to any site of the antibody constant region, for example, by covalent bonding through a linker or by coordinate bonding with a metal ion.
患者への抗体又はそのフラグメントの送達は、単独か又は例えばリポソーム(好 ましくは、正電荷リポソ一ム).、マイクロカプセル又はナノ粒子中に抗体又はその フラグメントを封入した形態で、 通常は適当な担体、 賦形剤又は希釈剤と組み合 わせて、非経口経路 (例えば、 静脈内投与、腹腔内投与、筋肉内投与、皮下投与、 局所投与など) にて行うことができる。 ' 1. 4組成物 Delivery of the antibody or fragment thereof to the patient is usually appropriate, either alone or in the form of the antibody or fragment thereof encapsulated in liposomes (preferably positively charged liposomes), microcapsules or nanoparticles. It can be administered by a parenteral route (for example, intravenous administration, intraperitoneal administration, intramuscular administration, subcutaneous administration, topical administration, etc.) in combination with an appropriate carrier, excipient or diluent. '1.4 Composition
本発明の組成物は、.癌、特に DREFを発現する癌をもつ患者を治療するために使 用することができる。そのような癌の例は、以下のものに限定されないが、肺癌、 食道癌、 瞵癌、 胃癌、 肝癌、 大腸癌、 甲状腺癌、 前立腺癌、 膀胱癌、 腎癌、 皮膚 癌、 胆癌、 脳腫瘍、 乳癌、 卵巣癌、 子宮頸癌、 精巣癌、 リンパ腫、 メラノーマ、 肉腫、 骨肉種などを含み、 より好ましい癌は、 肺癌、 食道癌、 乳癌、 腌癌などで める。 The compositions of the invention can be used to treat patients with cancer, in particular cancers that express DREF. Examples of such cancers include, but are not limited to, lung cancer, esophageal cancer, sputum cancer, stomach cancer, liver cancer, colon cancer, thyroid cancer, prostate cancer, bladder cancer, kidney cancer, skin cancer, gall cancer, brain tumor Breast cancer, ovarian cancer, cervical cancer, testicular cancer, lymphoma, melanoma, sarcoma, osteosarcoma, etc. More preferred cancers include lung cancer, esophageal cancer, breast cancer, vaginal cancer and the like.
本発明の組成物中の核酸の用量は、 siRNA分子又はアンチセンス核酸分子に換 算すると、 以下のものに限定されないが、 1投与単位あたり、 1ηΜ〜100 μ Μ、 好ま しくは 10ηΜ〜50 Μ、 より好ましくは 100ηΜ〜20 ί Μである。 本発明の組成物中の抗体又はそのフラグメントの用量は、 以下のものに限定さ れないが、 1投与単位あたり、 約 1〜約 100mg/ml、 約 5〜約 70mg/ml、 約 10〜 50mg/mlである。 The dose of the nucleic acid in the composition of the present invention is not limited to the following when converted to siRNA molecules or antisense nucleic acid molecules, but is 1η100 to 100 μ投 与, preferably 10ηΜ to 50Μ per dosage unit. More preferably, it is 100ηΜ to 20 °. The dose of the antibody or fragment thereof in the composition of the present invention is not limited to the following, but is about 1 to about 100 mg / ml, about 5 to about 70 mg / ml, about 10 to 50 mg per dosage unit. / ml.
しかし、 上記の用量又は投与量は、 患者の状態、 年齢、 性別、 重篤度などに応 じて変化しうるものであり、 専門医の判断により用量又は投与量が決定されるべ きである。 However, the above dose or dose may vary depending on the patient's condition, age, gender, severity, etc., and the dose or dose should be determined at the discretion of a specialist.
本発明の組成物は、 通常、 製薬上許容可能な担体、 賦形剤又は希釈剤、 例えば 滅菌水、 生理食塩水、 緩衝液、 非水性液体 (例えば、 アーモンド油、 植物油、 ェ タノールなど) などを含むことができる。 該組成物にはさらに、 製薬上許容可能 な安定剤 (例えばメチォニンなどのアミノ酸類)、保存剤 (P-ヒ ドロキシ安息香酸 メチル、 ソルビン酸)、 等張化剤 (例えば塩化ナトリウム)、 乳化剤 (例えばレシ チン、 アラビアゴム)、懸濁化剤 (例えばセルロース誘導体) などを含有させるこ とができる。 . The composition of the present invention usually contains a pharmaceutically acceptable carrier, excipient or diluent such as sterile water, physiological saline, buffer solution, non-aqueous liquid (eg almond oil, vegetable oil, ethanol, etc.), etc. Can be included. The composition further comprises a pharmaceutically acceptable stabilizer (for example, amino acids such as methionine), a preservative (methyl P-hydroxybenzoate, sorbic acid), an isotonic agent (for example, sodium chloride), an emulsifier ( For example, lecithin, gum arabic), a suspending agent (for example, a cellulose derivative) and the like can be contained. .
' 好ましい医薬製剤は、溶液剤、懸濁液剤、乳剤、 リポソ^ "ム封入剤などである。 本発明の組成物の投与方法は、 非経口投与、 例えば静脈内投与、 腹腔内投与、 筋肉内投与、 皮下投与、 局所投与などを含む。 局所投与には、 外科手術又は内視 鏡下で患部に直接注射する方法などが含まれる。 また、 専門医が決定した治療計 画に基づいて、一定の時間間隔、例えば 1週間、 2週間、 3週間、 1ヶ月、 2ヶ月、 6ヶ月、 8ヶ月、 1年又は 2年などの間隔で、 患者に対して、 本発明の組成物を 1 〜数回に分けて投与することができる。 'Preferred pharmaceutical preparations are solutions, suspensions, emulsions, liposome encapsulants, etc. The composition of the present invention can be administered parenterally, for example intravenously, intraperitoneally, intramuscularly. Administration, subcutaneous administration, local administration, etc. Local administration includes direct injection into the affected area under surgery or endoscopy, etc. In addition, based on a treatment plan determined by a specialist One to several doses of the composition of the present invention to the patient at time intervals such as 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 6 months, 8 months, 1 year or 2 years Can be administered separately.
本発明の RNAi核酸、 アンチセンス核酸、 又は抗体を癌、特に肺癌に適用したと き、 肺癌細胞で強く細胞周期停止とともに細胞死誘導を起こすことができる。 特 に一部の肺癌細胞では、 mitot ic catastrophと呼ばれる、 M期での細胞死誘導が 見られた。 一方、 正常肺由来細胞株では、 肺癌に比して、 細胞周期停止は軽度で あ た。 When the RNAi nucleic acid, antisense nucleic acid, or antibody of the present invention is applied to cancer, particularly lung cancer, it can induce cell death together with cell cycle arrest strongly in lung cancer cells. In particular, in some lung cancer cells, induction of cell death in M phase called mitotic catastroph was observed. On the other hand, in normal lung-derived cell lines, cell cycle arrest was mild compared to lung cancer.
本発明の DREFを標的とする癌治療法は、下記に述べる治療剤のスクリーニング 法と組み合わせることによって、 癌患者の症例に最適なオーダーメイド医療とし て使用できるだろう。 The cancer treatment method targeting DREF of the present invention can be used as a tailor-made medicine optimal for cancer patient cases by combining with the screening method for therapeutic agents described below.
2 . 癌治療薬のスク リ一二ング · 本発明はさらにヒ ト培養癌細胞、或いは強発現可能に DREF遺伝子をトランスフ ェクシヨンした(正常又は非正常) ヒト細胞株、を含む培地に候補薬剤を加えて、 DREF遺伝子の発現又は翻訳を抑制する薬剤をインビトロでスクリ一ユングする ことを含む、 癌治療用薬剤をスクリーニングする方法を提供する。 2. Screening of cancer drugs The present invention further suppresses the expression or translation of the DREF gene by adding a candidate drug to a culture medium containing human cultured cancer cells or a human cell line in which the DREF gene has been transfected so that it can be strongly expressed (normal or non-normal). There is provided a method for screening a drug for treating cancer, comprising screening a drug in vitro.
薬剤のスクリーニングは、 具体的には、 癌細胞、 或いは DREF強発現哺乳類 (例 えばヒ ト、 マウスなど、 好ましくはヒ ト) 細胞株、 の細胞増殖抑制、 細胞周期停 止及び/又は細胞死誘導を指標とすることができる。 Specifically, drug screening is performed by inhibiting cancer cell growth, cell cycle arrest, and / or cell death in cancer cells or mammalian cells that strongly express DREF (eg, humans, mice, etc., preferably humans). Can be used as an index.
具体的には、 本発明の方法は、 ヒ ト培養癌細胞、 又は DREF強発現細胞株、 を準 備し、 該細胞を候補薬剤の存在下で培養し、 DREF遺伝子又は mRNAの発現の抑制 について、 或いは上記細胞又は細胞株の細胞增殖抑制、 細胞周期停止及び Z又は 細胞死誘導について、 候補薬剤をスクリーニングすることを含む。 Specifically, the method of the present invention comprises preparing a human cultured cancer cell or a cell line that strongly expresses DREF, culturing the cell in the presence of a candidate drug, and suppressing the expression of the DREF gene or mRNA. Or screening candidate drugs for cell proliferation inhibition, cell cycle arrest and Z or cell death induction of the cell or cell line.
ヒ ト培養癌細胞には、 公知の癌細胞株、 癌患者からのバイオプシー由来の癌細 胞などが含まれ、 本発明において使用できる。 癌細胞株の例は、 いずれも肺癌細 胞株である、 ACC-LC-91 (H. Osada ら, Mol. Carcinog.、2005, 44: 233- 241)等の ACC- LC、 A549 (ATCC, Rockvill, MD)、 PC10 (免疫生物研究所、 群馬、 日本)、 Calu6 (ATCC, Rockvi ll, MD)などである。 The human cultured cancer cells include known cancer cell lines, cancer cells derived from biopsies from cancer patients, etc., and can be used in the present invention. Examples of cancer cell lines are all lung cancer cell lines such as ACC-LC-91 (H. Osada et al., Mol. Carcinog., 2005, 44: 233-241), A549 (ATCC, Rockvill, MD), PC10 (Immuno-Biological Research Institute, Gunma, Japan), Calu6 (ATCC, Rockville, MD).
DREF強発現細胞株は、 例えば次のようにして作製することができる。 レンチウ ィルスベクター(例えば CSII- CMV- MCS- IRES2- Bsdベクター(理化学研究所(埯玉、 日本) の三好浩之博士から入手) に DREF cDNAを挿入したレンチウィルスを作製 し、 これを、 ヒ ト正常気道上皮由来細胞株 BEAS2B に感染させ、 Blasticidin (Invitrogen)で選択培養することで、 DREFを恒常的に強発現している株を選択す る。培養培地としては、例えば胎児牛血清 1 %添加 F12培地(Sigma)を使用できる。 増殖速度は、 TetraColorOne™ (生化学工業、 東京、 日本) を用いる MTTアツセィ などによって測定できる。 このようにして実際に作製された DREF 強発現細胞株 (例えば BEAS2B-DREF株) は、 6日の培養で対照株と比べて約 3倍の増殖速度の 亢進が観察された。 A cell line that strongly expresses DREF can be prepared, for example, as follows. A lentivirus was prepared by inserting the DREF cDNA into a lentivirus vector (eg CSII-CMV-MCS-IRES2-Bsd vector (obtained from Dr. Hiroyuki Miyoshi of RIKEN (Kodama, Japan)). Infect the respiratory epithelium-derived cell line BEAS2B and selectively culture with Blasticidin (Invitrogen) to select a strain that constantly expresses DREF strongly. Medium (Sigma) can be used The growth rate can be measured by MTT Atsey etc. using TetraColorOne ™ (Seikagaku, Tokyo, Japan) etc. DREF strong expression cell lines (eg BEAS2B- The DREF strain) was observed to increase growth rate about 3 times compared to the control strain after 6 days of culture.
本発明の方法において、 DREF遺伝子又は raRNAの発現の抑制の程度は、 候補薬 剤を添加しない対照との比較実験によって判定できる。 発現レベルは、 癌細胞、 又は DREF強発現細胞株、から周知の方法(例えばフヱノール/クロ口ホルム/イソ ァミルアルコール法やグァニジゥム /CsCl法、オリゴ dTセルロースカラムクロマ トグラフィーなど) で得た全 RNA又は mRNA又はポリ A (+) RNAについて、或いは逆 転写酵素 - PCR (RT-PCR)法によって RNAから合成された cDNAについて、定量 RT- PCR 法、 蛍光又は放射性標識したプローブを用いるハイブリダィゼーシヨン法 (例え ばノーザンハイブリダィゼーシヨン、 サザンハイブリダィゼーシヨン、 DNA マイ クロアレイ、 組織マイクロアレイなど) によって決定することができる (西郷薰 ら訳、 分子生物学実験プロ トコール I, II, III、 1997年、 丸善)。 或いは、 発現レ ベルは、 DREF遺伝子によってコードされるタンパク質 (配列番号 1) の細胞内レ ベルを、 該タンパク質に対する抗体又はそのフラグメントを用いる免疫測定法、 ウェスタンハイプリダイゼーション法、 組織染色法などによって測定することに よって間接的に決定することができる。 In the method of the present invention, the degree of suppression of DREF gene or raRNA expression can be determined by a comparison experiment with a control to which no candidate drug is added. The expression level is determined by a well-known method (for example, phenol / chloroform / isoform) from cancer cells or DREF strongly expressing cell lines. Total RNA or mRNA or poly A (+) RNA obtained by the amyl alcohol method, guanidinium / CsCl method, oligo dT cellulose column chromatography, etc.) or from the RNA by reverse transcriptase-PCR (RT-PCR) method For synthesized cDNA, quantitative RT-PCR method, hybridization method using fluorescently or radioactively labeled probe (eg Northern hybridization, Southern hybridization, DNA microarray, tissue microarray, etc.) (Translated by Atsushi Saigo et al., Molecular Biology Experiment Protocol I, II, III, 1997, Maruzen). Alternatively, the expression level is determined by measuring the intracellular level of the protein encoded by the DREF gene (SEQ ID NO: 1) by immunoassay using an antibody against the protein or a fragment thereof, Western hyperprecipitation method, tissue staining method, etc. It can be determined indirectly by measurement.
前記プローブは、 配列番号 2 (202位〜 2286位)のヌクレオチド配列又はそれと 相補的な配列、 或いはその連続する例えば約 20以上、 約 30 以上、 50以上、 70 以上、 100以上、 150以上、 200以上、 250以上のヌクレオチドからなる配列、 を 有する DNAである。 プローブは、 蛍光又は放射性標識を結合した標識プローブと するのが好ましい。 蛍光性標識には、 例えばフルォレサミン、 ローダミン、 それ らの誘導体、 Cy3、 Cy5などが含まれる、 放射性標識には、 例えば放射性リン又は ィォゥ原子が含まれる。 The probe is a nucleotide sequence of SEQ ID NO: 2 (positions 202 to 2286) or a sequence complementary thereto, or a sequence thereof, for example, about 20 or more, about 30 or more, 50 or more, 70 or more, 100 or more, 150 or more, 200 A DNA having a sequence consisting of 250 or more nucleotides. The probe is preferably a labeled probe bound with a fluorescent or radioactive label. Fluorescent labels include, for example, fluoresamine, rhodamine, their derivatives, Cy3, Cy5, etc. Radiolabels include, for example, radioactive phosphorus or thio atoms.
ハイブリダイゼーションは、 低ハイブリダイゼーション、 中ハイプリダイゼー シヨン又は高ハイブリダィゼーション条件によって、 好ましくは高ハイブリダィ ゼーシヨン条件によって行うことができる。 例えば、 約 45〜50°Cで 2〜6 X SSC Hybridization can be performed under low hybridization, medium hybridization or high hybridization conditions, preferably under high hybridization conditions. For example, 2-6 X SSC at about 45-50 ° C
(1 X SSCは 150mM塩化ナトリ ゥム /15mMタエン酸ナトリゥム) 中でのハイプリダ ィゼーシヨンと、 それに続く、 約 50〜65°Cでの 0. 2〜 2 X SSC/0. 1〜 1 %SDSによ る洗净カ らなる (例えば、 Ausubel ら, Curent Protocols in Molecular Biology,(1 X SSC is 150 mM sodium chloride / 15 mM sodium taenate) followed by a high pre-degradation, followed by 0.2-2 X SSC / 0.1-1% SDS at about 50-65 ° C. (For example, Ausubel et al., Curent Protocols in Molecular Biology,
1995 年, John Wiley and Sons, US ; 西郷薰ら訳、 分子生物学実験プロ トコール1995, John Wiley and Sons, US; translated by Kei Saigo et al., Molecular Biology Experiment Protocol
I, II, III、 1997年、丸善)。 或いは、 60〜65°Cで 6 X SSCヽ Denhardt ' s溶液、 0. 2%I, II, III, 1997, Maruzen). Or 6 X SSC ヽ Denhardt's solution at 60-65 ° C, 0.2%
SDS中でのハイブリダィゼーシヨンののち、 60〜65°Cでの 0· 2 X SSC、 0. 1%SDSに よる洗浄などからなる。 ハイブリダイゼーション温度は、 一般に融解温度(Tm)よ り 5〜 10 °C低い温度を使用する こ とができ る。 Tra は、 例えば、 式 : Tm (°C) =81. 5+16. 6 (log10 [Na+] ) +0. 41 (%G+C) - (600/N) (式中、 Nはハイブリッ ドの塩 基数であり、 [Na+]はハイブリダィゼーシヨンバッファ中の Na+の濃度である。 ) によって求めること力 Sできる Q. Sarabrookら, Molecular Cloning : A Laboratory Mannual 2 Cold Spring Haroor Laboratory, Cold Spring Harbor Laboratory Press, 1989)。 After hybridization in SDS, it consists of cleaning with 0.2X SSC, 0.1% SDS at 60-65 ° C. The hybridization temperature can generally be 5 to 10 ° C lower than the melting temperature (Tm). Tra is, for example, the formula: Tm (° C) = 81.5 + 16.6 (log 10 [Na + ]) +0.41 (% G + C)-(600 / N) (where N is the number of hybrid bases) , [Na + ] is the concentration of Na + in the hybridization buffer.) Q. Sarabrook et al., Molecular Cloning: A Laboratory Mannual 2 Cold Spring Haroor Laboratory, Cold Spring Harbor Laboratory Press, 1989).
,候補薬剤は、 小分子、 ぺプチド、 ポリぺプチド、 タンパク質、 ヌクレオシド、 ォリゴヌクレオチド、 ポリヌクレオチド、 核酸 (DNA又は RNA) などを含むが、 こ れらに限定されない。 Candidate agents include, but are not limited to, small molecules, peptides, polypeptides, proteins, nucleosides, oligonucleotides, polynucleotides, nucleic acids (DNA or RNA), and the like.
免疫測定法は、 抗原一抗体反応を利用する分析法であり、 例えば酵素結合抗体 法 (例えば ELISA)、 蛍光抗体法、 固相法、 均一法、 サンドイッチ法、 ピオチン/ アビジン系などを適宜組み合わせて行うことができる。 固相としては、 例えば巿 販のプラスチック製プレート(例えばポリスチレン製の 96 ゥエルプレートなど) を使用することができる。 これらの方法は、 当業界で周知であり、 その慣用技術 を本発明で使用できる。 An immunoassay is an analysis method that uses an antigen-antibody reaction. For example, an enzyme-linked antibody method (for example, ELISA), a fluorescent antibody method, a solid phase method, a homogenous method, a sandwich method, a piotine / avidin system, etc. It can be carried out. As the solid phase, for example, a commercially available plastic plate (for example, a 96-well plate made of polystyrene) can be used. These methods are well known in the art and their conventional techniques can be used in the present invention.
ヒ ト培養癌細胞、 又は DREF強発現細胞株、 において、 DREF遺伝子又は mRNAの 発現が、 候補薬剤の存在によって、 候補薬剤無添加の対照と比べて、 有意に阻害 又は抑制される場合、 該候補薬剤は癌治療剤として同定しうる。 If the expression of DREF gene or mRNA is significantly inhibited or suppressed in human cultured cancer cells, or DREF strongly expressing cell lines, compared to the control without addition of the candidate drug due to the presence of the candidate drug, the candidate The drug can be identified as a cancer therapeutic.
定量 RT - PCRは、 例えば、 Taqポリメラ一ゼなどの耐熱性ポリメラーゼの存在下 で mRNA又はポリ A (+) RNAを铸型に、 DREF遺伝子の配列由来のプラーマ'一を用い る PCRによって行うことができる。 プライマーのサイズは、約 15〜30ヌクレオチ ド、好ましくは 17〜25ヌクレ: チドである。 このとき発現変動のないハウスキー ビング遺伝子の発現量に対する DREF遺伝子の発現量を決定する。 Quantitative RT-PCR is performed, for example, by PCR using mRNA or poly A (+) RNA in the vertical form in the presence of thermostable polymerases such as Taq polymerase and using a PREF derived from the DREF gene sequence. Can do. The size of the primer is about 15-30 nucleotides, preferably 17-25 nucleotides. At this time, the expression level of the DREF gene is determined relative to the expression level of the housekeeping gene with no expression fluctuation.
候補薬剤のスクリ一ユングはまた、 ヒ ト培養癌細胞、 又は DREF強発現細胞株、 の細胞増殖の抑制、 細胞周期の停止、 又は細胞死を調べることによつても行うこ とができる。 Candidate drug screening can also be performed by examining cell growth inhibition, cell cycle arrest, or cell death in human cultured cancer cells, or DREF strongly expressing cell lines.
細胞増殖は、 次の手法によって測定できる。 遺伝子導入の翌日、 細胞を蒔き直 した後に puromycin /z g/ml)で 2日間選択し、 更に puromycin (0. 5 μ g/ml)で 10 日間選択を続ける。 その後、 TetraColorOne™ (生化学工業、 東京、 日本) 5%入り 培地に置き換え、 37°Cで 1時間反応させる。 その後その培地を回収しプレートリ ーダ一にて 0D450nmを測定し (このとき、 OD630nmを対照とする。)、 生存細胞数 とする (colorimetric assay) 0 又、 その後、 細胞はメタノール固定後に、 5%Gierasa (S igma - Aldrich)水溶液にて染色する。 Cell proliferation can be measured by the following method. On the next day after gene transfer, repopulate the cells and select with puromycin / zg / ml) for 2 days, then continue with puromycin (0.5 μg / ml) for 10 days. Then, replace with TetraColorOne ™ (Seikagaku, Tokyo, Japan) 5% medium and incubate at 37 ° C for 1 hour. The medium is then collected and removed. 0D450nm is measured using a reader (in this case, OD630nm is used as a control), and the number of living cells is determined (colorimetric assay). 0 After that, the cells are fixed with methanol, and then 5% Gierasa (Sigma-Aldrich) Stain with aqueous solution.
細胞周期及び細胞死は、 次の手法によって測定できる。 遺伝子導入細胞を puromycin選択後に回収し、 低張液 (75 mM KC1) に浮遊させる(37°C, 20分)。 そ の後、 攪拌しながら固定液 (メタノール:氷酢酸 = 3 : 1混合液) 約 3容を加え て細胞を固定する。 遠心上清除去後に再度固定液に浮遊させる。 細胞浮遊液をス ライドガラス上に 1、 2滴滴下し風乾する。 その後 5% Giemsa水溶液にて核染色体 を染色する。実体顕微鏡 BHT- 323 (Olympus)にて観察し、細胞を間期(G9/G1, S, G2)、 細胞分裂前期(Prophase)、 細胞分裂中期 (Metaphase)、 細胞分裂後期/終期 (Anaphase/telophase;ヽ (mi tot ic catastroph άΧ)、 apoptosisノ Ιιこ rけ て計測する。 Cell cycle and cell death can be measured by the following methods. The transgenic cells are collected after puromycin selection and suspended in a hypotonic solution (75 mM KC1) (37 ° C, 20 minutes). Then, fix the cells by adding about 3 volumes of fixative (methanol: glacial acetic acid = 3: 1 mixture) with stirring. After removing the supernatant, resuspend in the fixative. Drop 1 or 2 drops of cell suspension on a slide glass and air dry. Then, stain the nuclear chromosome with 5% Giemsa aqueous solution. The cells were observed with a stereomicroscope BHT-323 (Olympus), and the cells were interphase (G9 / G1, S, G2), prophase, metaphase, metaphase, late phase / terminal phase (Anaphase / telophase) ; ヽ (mi tot ic catastroph άΧ), measure by apoptosis.
3 . DREF遺伝子の発現抑制によって影響を受ける遺伝子群 3. Gene groups affected by suppression of DREF gene expression
DREF遺伝子の発現を RNAi核酸によって抑制したときに、 発現が変化する遺伝 子群を、 マイクロアレイを使用して解析した。 · A group of genes whose expression changes when DREF gene expression is suppressed by RNAi nucleic acid was analyzed using a microarray. ·
DREF-RNAi#4による DREF遺.伝子の発現抑制によって、 表 1 (下記参照) に示さ れる遺伝子群の発現に変化が認められた。 Due to the suppression of DREF gene expression by DREF-RNAi # 4, changes in the expression of genes shown in Table 1 (see below) were observed.
DREF- RNAi#3による DREF遺伝子の発現抑制によって、 表 2 (下記参照) に示さ れる遺伝子群の発現に変化が認められた。 ' Due to the suppression of DREF gene expression by DREF-RNAi # 3, changes in the expression of genes shown in Table 2 (see below) were observed. '
遺伝子の多く力 癌の発症、 癌の進展、 癌の転移と関連の高いものであること が判明した。 これらの解析からも、 DREFが種々の癌にとって重要な因子であるこ とが分かる。 それゆえに、 DREFを分子標的とする本発明の方法は、 癌の治療にと つて有効な方法を提供するものであるといえる。 Many genes have been found to be highly related to cancer development, cancer progression, and cancer metastasis. These analyzes also indicate that DREF is an important factor for various cancers. Therefore, it can be said that the method of the present invention using DREF as a molecular target provides an effective method for the treatment of cancer.
以下の実施例によって本発明をさらに具体的に説明するが、 本発明の範囲はこ れらの実施例によつて制限されないものとする。 実施例 The following examples further illustrate the present invention, but the scope of the present invention is not limited by these examples. Example
(実験方法) (experimental method)
1 . ヒ ト癌細胞株 ヒ ト肺癌細胞株 (ACC-LC- 91 又は A549)、 5 %牛胎児血清(Invitrogen)添加 RPMI 1640培地(Sigma)を用いて 5 %C02存在下で培養し、 以下の実験に使用した。 又、ヒ ト正常気道上皮細胞株(BEAS2B又は HPL1)は 1 %牛胎児血清(Invitrogen;)、 5 μ g/ml牛ィンスリン (S igma)、 5 μ g/ral ヒ ト トランスフェリン (S igma)、 0. 1 μ M ハイドロコルチゾン (S i gma) 及ぴ 0. 2nMトリョードサイロニン (S i gma) を添加 した Ham F12培地 (Sigma) を用いて 5 %C02存在下で培養し、 以下の実験に使用 した。 1. Human cancer cell line Human lung cancer cell line (ACC-LC- 91 or A549), were cultured in 5% C0 2 presence with 5% fetal calf serum (Invitrogen) added RPMI 1640 medium (Sigma), it was used in the following experiments. Human normal airway epithelial cell lines (BEAS2B or HPL1) are 1% fetal bovine serum (Invitrogen;), 5 μg / ml bovine insulin (Sigma), 5 μg / ral human transferrin (Sigma), with 0. 1 mu M hydrocortisone (S i gma)及Pi 0. 2 nM preparative RHO de thyronine (S i gma) was added a Ham F12 medium (Sigma) and cultured in 5% C0 2 the presence of the following Used in the experiment.
2 . トランスフエクシヨン及び DREF発現ベクター 2. Transformation and DREF expression vectors
細胞への DREF発現ベクター及び RNAi -ベクターの遺伝子導入 (tra.nsf ect ion) は、 3. 5 cm 培養皿に 3 X 105 の細胞を蒔き、 翌日 DNA と L ipof ectamine2000 (Invi trogen)を混合し、培地中に添加して行った。' RNAi ベクター等の puromycin 耐性遗伝子を持つベクターをトランスフエク トした場合は、 トランスフエクショ ンの翌日より(必要に応じ細胞を蒔き直した後に) puromyc in d ihydrochlori de (Si gma-Aldrich, St. Loui s, M0) 2 g/ml培地に添加し 2日間選択後に遺伝子導 入細胞を回収し、 RNA及び蛋白試料の作成、 或いは FACS解析等を施行した。 DREF 発現ベクターは、 RT-PCRによ.り得られた DREF cDNAの 0RF部分を発現べクターの pcDNA3 (Inv i trogen)に揷入したものを廣瀬富美子博士 (兵庫県立大学、 兵庫、 日 本) より分与された。 For DREF expression vector and RNAi-vector gene transfer into cells (tra.nsf ect ion), seed 3 x 10 5 cells in a 3.5 cm culture dish and mix DNA and Lipofectamine 2000 (Invitrogen) the next day. And added to the medium. '' When a vector with a puromycin-resistant gene such as RNAi vector is transfected, puromyc in dihydrochlori de (Sigma-Aldrich, St (Louis, M0) 2 g / ml medium was added, and after 2 days of selection, the transfected cells were collected, and RNA and protein samples were prepared or FACS analysis was performed. The DREF expression vector was obtained by inserting the 0RF portion of the DREF cDNA obtained by RT-PCR into the expression vector pcDNA3 (Invitrogen). Dr. Fumiko Hirose (Hyogo Prefectural University, Hyogo, Japan) More distributed.
3 . RNAi ォリ ゴヌク レオチドぉよぴ RNAiベクター ' 3. RNAi vector RNAi vector '
図 1に示すように、 対照としての公知の 2つの RNAi部位(#1, #2)のうちの 1箇 所 (#1 ) 及び新規 RNAi 4箇所 (#3, #4, #5及び #6) に対する DNAオリゴヌクレオチ ドを Greiner Japan (東京、 日本) に依頼して作製した。 その塩基配列は以下の とおりである (なお、 RNAiの部位の数字は DREF 0RF中の開始コドンの塩基 「A」 を第 1位としたときの核酸残基の位置番号を表わす)。 As shown in Figure 1, one of the two known RNAi sites (# 1, # 2) as a control (# 1) and four new RNAi sites (# 3, # 4, # 5 and # 6) ) DNA oligonucleotide was prepared by requesting Greiner Japan (Tokyo, Japan). Its base sequence is as follows (note that the number at the RNAi site represents the position number of the nucleic acid residue when the base “A” of the start codon in DREF 0RF is the first position).
RNAi#l (配列番号 16) RNAi # l (SEQ ID NO: 16)
5, - AATTCTGCGAGTTCGTCAAGA-3' (221位〜 241位) 5,-AATTCTGCGAGTTCGTCAAGA-3 '(221st to 241st)
RNAi#l用 ol igo (HI promoter用) (配列番号 17及び 18) Ol igo for RNAi # l (for HI promoter) (SEQ ID NOs: 17 and 18)
H1F1: H1R1 : H1F1: H1R1:
3, 叫 3, shouting
RNAi #3 (ffi列番号 19) RNAi # 3 (ffi column number 19)
5, -GCAACAACCACCACCTCATGC-3' (1136位〜 1156位) 5, -GCAACAACCACCACCTCATGC-3 '(1136th ~ 1156th)
RNAi#3用 ol igo (配列番号 20及ぴ 21) Ol igo for RNAi # 3 (SEQ ID NO: 20 and 21)
si3F: 5 ' -CAACAACCACCACCTCATGCCTTCCTGTCAGCATGAGGTGGTGGTTGTTGCTTTTTG-3 ' si3F: 5 '-CAACAACCACCACCTCATGCCTTCCTGTCAGCATGAGGTGGTGGTTGTTGCTTTTTG-3'
RNAi #4 (配列番号 22) RNAi # 4 (SEQ ID NO: 22)
5, - GCAACTTCAAGTCCCAGAAGG- 3' (1718位〜 1738位) . 5,-GCAACTTCAAGTCCCAGAAGG-3 '(from 1718 to 1738).
RNAi#4用 ol igo (配列番号 23及ぴ 24) Ol igo for RNAi # 4 (SEQ ID NO: 23 and 24)
s i4F s i4F
s i4R s i4R
RNAi #5 (配列番号 25) RNAi # 5 (SEQ ID NO: 25)
5' -GTACCCCACCATCAGCATGGT-3' (1245位〜 1265位) 5 '-GTACCCCACCATCAGCATGGT-3' (1245th ~ 1265th)
RNAi#5用 ol igo (配列番号 26及び 27) Ol igo for RNAi # 5 (SEQ ID NO: 26 and 27)
si5Fsi5F
RNAi #6 (配列番号 28) RNAi # 6 (SEQ ID NO: 28)
5, -GGTGCTTGGCCTCAACGAAGA-3 ' (1737位〜 1757位) 5, -GGTGCTTGGCCTCAACGAAGA-3 '(1737-1757)
RNAi#6用 ol igo (配列番号 29及び 30) Ol igo for RNAi # 6 (SEQ ID NOs: 29 and 30)
s i6F:s i6F:
blank ol igo (対照用)(Apal-EcoRI消化 pU6- puro) (配列番号 31及ぴ 32) ' blankF: 5 ' - CGATATCTTTTTTG-3 ' blank ol igo (for control) (Apal-EcoRI digested pU6-puro) (SEQ ID NO: 31 and 32) 'blankF: 5'-CGATATCTTTTTTG-3 '
blankR: 3 ' -CCGGGCTATAGAAAAAACTTAA-5 ' blankR: 3 '-CCGGGCTATAGAAAAAACTTAA-5'
(注 1 ) RNAi#3〜^の最初の Gはベクター上の Gが読まれるため、 ol igo配列で は RNAi部位の最初の Gが含まれず、 2番目の残基から始まっている。 (Note 1) Since the first G of RNAi # 3 ~ ^ reads the G on the vector, the ol igo sequence does not include the first G of the RNAi site and starts from the second residue.
(注 2 ) 下線部は DREF RNAi部位を示す。 (Note 2) The underlined portion indicates the DREF RNAi site.
puromycin耐性遺伝子を持つプラスミ ドべクタ一(pCMV - puro) 〔(注) このべク ターは、 Clontech社のプラスミ ド pIRESpuro2 (5. 2kb長)から、 不要な部分 1. Okb 長を切り取って作製されたものである。〕 に、 RNA polymerase III によって転写 するプロモーターであるマウス U6遺伝子プロモーターを揷入した RNAiベクター を作製した (pU6- puro) (Osada. , Η.ら, Cancer Res. 65 : 10680-5, 2005)。 同様 にヒ ト HI 遺伝子プロモーターを挿入した RNAi ベタターを作製した (pHl-RNApuro)。 Plasmid vector with puromycin resistance gene (pCMV-puro) [Note] This vector The filter was prepared by cutting off the unnecessary part 1. Okb length from Clontech's plasmid pIRESpuro2 (5.2 kb length). In addition, an RNAi vector containing the mouse U6 gene promoter, a promoter transcribed by RNA polymerase III, was prepared (pU6-puro) (Osada., .. et al., Cancer Res. 65: 10680-5, 2005). Similarly, an RNAi betater inserted with the human HI gene promoter was prepared (pHl-RNApuro).
RNAi べクターの作製のために、 RNAi#l 用 ol igo は、 pHl- RNApuro を Bglll-Hindlll消化後に挿入した。 RNAi #3〜#6は、 pU6- puroを Apal消化、 T4 DNA polymerase処理、 及び EcoRI消化した後に挿入した。 To prepare RNAi vector, ol igo for RNAi # l was inserted after digestion of pHl-RNApuro with Bglll-Hindlll. RNAi # 3 to # 6 were inserted after pU6-puro was digested with Apal, treated with T4 DNA polymerase, and digested with EcoRI.
Blank ol igoは、 pU6-puroを Apal—EcoRI消ィ匕後に挿人した。 Blank ol igo inserted pU6-puro after Apal-EcoRI was erased.
4 . RT- PCR解析 ' 4. RT-PCR analysis ''
遺伝子導入後の細胞から、 RNeasy Mini Kit (Qiagen)にて total RNAを回収し た。 One- step RT- PCR kit (Qiagen)にて、 添付の使用説明書にしたがって RT- PCR を施行し、 RT- PCR産物を TBE- agaroseゲル電気泳動にかけて、 DREF遺伝子の発現 を検討した (図 2 )。 Total RNA was recovered from the transfected cells using the RNeasy Mini Kit (Qiagen). RT-PCR was performed using the One-step RT-PCR kit (Qiagen) according to the attached instruction manual, and the RT-PCR product was subjected to TBE-agarose gel electrophoresis to examine the expression of the DREF gene (Figure 2). ).
PCRプライマー: PCR primers:
DREF F1: 5, -TGGCAAGGACATCGTGAAGG (hDREF 0RFの 813位〜 832位;配列番号 135) DREF R1 : 5' - GACAGCAACAACCACCACCT (hDREF 0RFの 1132位〜 1151位.;配列番号 136) ' DREF F1: 5, -TGGCAAGGACATCGTGAAGG (positions 813 to 832 of hDREF 0RF; SEQ ID NO: 135) DREF R1: 5 '-GACAGCAACAACCACCACCT (1132 to 1151 of hDREF 0RF; SEQ ID NO: 136)'
5 . Western blot解析 5. Western blot analysis
DREFタンパク質は、 DREF - cDNAの付加された HA - tag/myc-tagに対する抗体を用 いて検出された。 遺伝子導入後の細胞を 4%SDS溶液で溶解、 加熱処理した後に、 タンパク質を SDS - PAGEにて電気泳動し、セミ ドライ電気転写システム Ti^ns- Blot SD cel l (Bio - Rad)にて Immobilon- Pメンプレン (Mill ipore) に転写した。 メン プレンに抗 HA- tag抗体 (Santa cruz) 及ぴ抗 myc- tag抗体 (9E10, Santa cruz) を重層し、 洗浄後 horseradish peroxidaseが結合した二次抗体 (Cell Signaling) を再度重層し、 ECL Western Blotting Detection Reagents (GE Healthcare)にて、 DREFタンパク質を検出した。 DREF protein was detected using an antibody against HA-tag / myc-tag to which DREF-cDNA was added. The cells after gene transfer are lysed with 4% SDS solution and heat-treated, and then the protein is electrophoresed on SDS-PAGE and Immobilon using the semi-dry electrotransfer system Ti ^ ns-Blot SD cell (Bio-Rad). -Transferred to Millipore. The membrane is overlaid with anti-HA-tag antibody (Santa cruz) and anti-myc-tag antibody (9E10, Santa cruz), washed and then overlaid with secondary antibody (Cell Signaling) bound with horseradish peroxidase, and ECL Western DREF protein was detected by Blotting Detection Reagents (GE Healthcare).
6 . FACS解析 · - 遺伝子導入後の細胞を、 IGEPAL (登録商標) CA- 630 (0.5% Sigma- Aldrich)、 Propidiura iodide (20 μ g/ml, Sigma- Aldrich)、 PBS溶液で浮遊させて、 裸核中の DNAを染色し、 DNA量を FACSCalibur (Becton- Dickinson)を用いて定量し、 細胞 周期解析ソフト ModFit(BD Biosciences社)で細胞周期 (G0/G1, S, G2/M) を解析 した。 6. FACS analysis ·- The cells after gene transfer are suspended in IGEPAL (registered trademark) CA-630 (0.5% Sigma-Aldrich), Propidiura iodide (20 μg / ml, Sigma-Aldrich), PBS solution, and DNA in the naked nucleus is transferred. After staining, the amount of DNA was quantified using FACSCalibur (Becton-Dickinson), and the cell cycle (G0 / G1, S, G2 / M) was analyzed using the cell cycle analysis software ModFit (BD Biosciences).
7. 細胞増殖解析 7. Cell proliferation analysis
遺伝子導入の翌日、細胞を蒔き直した後に puromycin (2 μ g/ml)で 2 '日間選択し、 更に puromycin (0.5 g/ml)で 10 日間選択を続ける。 その後、 TetraColor One (Seikagaku Kogyo、東京、日本) 5%入り培地に置き換え、 37°Cで 1時間反応させる。 その後その培地を回収しプレートリーダーにて 0D450nm を測定し (このとき、 0D630nmを対照とする。)、生存細胞数とした (colo imetric assay)。又、その後、 細胞はメタノール固定後に、 5% Giemsa (Sigma - Aldrich)水溶液にて染色した。 On the day after gene transfer, repopulate the cells and select with puromycin (2 μg / ml) for 2 'days, and then continue with puromycin (0.5 g / ml) for 10 days. Then, replace with TetraColor One (Seikagaku Kogyo, Tokyo, Japan) 5% medium and incubate at 37 ° C for 1 hour. Thereafter, the medium was collected, and 0D450nm was measured with a plate reader (in this case, 0D630nm was used as a control), and the number of viable cells was determined (coloimetric assay). Thereafter, the cells were fixed with methanol and then stained with 5% Giemsa (Sigma-Aldrich) aqueous solution.
8. 免疫染色 . 8. Immunostaining.
' 遺伝子導入の翌日、 細胞を培養皿に置いたカバーグラス上に蒔き直し、 2日間 puromycin (2 μ g/ml)選択する。その後、 3.7% Formaldehyde (10% Formalin) (Wako, Osaka, Japan) PBS溶液にて細胞を固定し(室温、 8分)、 TBS溶液(150mM NaCl, 50mM Tris (pH7.6))にて洗浄後、 0.5% TritonX (Wako) TBS 溶液にて、 細胞透過処理す る (室温、 8分)。 その後、 各種一次抗体 (抗 T/H2AX (Upstate, Lake Placid, NY), phospho-ATM-Serl981 (Cell Signaling, Danvers, MA), ji phospho-hdstoneH3 (Upstate, Lake Placid, NY)) を添加し、 Alexa Fluor 488 等の蛍光色素結合二 次抗体 (Molecular Probe—Invitrogen)を DAPI (Sigma— Aldrich) と共に添カロした。 洗浄後封入剤 PermaFluor (Thermo Shandon, Pittsburgh, PA) を加えてスライ ド ガラスに載せ、 共焦点レーザー蛍光顕微鏡 Radiance OO (BIO- RAD) にて観察し た。 'The day after gene transfer, seed cells on a cover glass placed in a culture dish and select puromycin (2 µg / ml) for 2 days. Then, cells were fixed with 3.7% Formaldehyde (10% Formalin) (Wako, Osaka, Japan) PBS solution (room temperature, 8 minutes), and washed with TBS solution (150 mM NaCl, 50 mM Tris (pH 7.6)) Apply cell permeabilization with 0.5% TritonX (Wako) TBS solution (room temperature, 8 minutes). After that, various primary antibodies (anti-T / H2AX (Upstate, Lake Placid, NY), phospho-ATM-Serl981 (Cell Signaling, Danvers, MA), ji phospho-hdstoneH3 (Upstate, Lake Placid, NY)) were added, A fluorescent dye-conjugated secondary antibody (Molecular Probe—Invitrogen) such as Alexa Fluor 488 was added together with DAPI (Sigma—Aldrich). After washing, the mounting medium PermaFluor (Thermo Shandon, Pittsburgh, PA) was added, placed on a slide glass, and observed with a confocal laser fluorescence microscope Radiance OO (BIO-RAD).
9. TUNEL解析 9. TUNEL analysis
免疫染色実験と同様に、 遺伝子導入細胞をカバーグラス上に蒔き、 2日間 puromycin選択する。 その後、 3.7% Formaldehyde (10% Formalin) (Wako) PBS溶液 にて細胞を固定する (室温、 10 分)。 その後、 1%過酸化水素含有メタノール溶液 処理 (室温、 2分)、 0.1% TritonX-100含有 PBS溶液処理 (室温、 5分) の後に、 In Situ Cel l Death Detection Kit, Fluorescein (Roche)にて、 細胞内の DNA 切断部位を Fluorescein 結合核酸にて標識する。 その後必要に応じて、 抗 phospho- histoneH3 等で免疫染色し、 洗浄後、 蛍光封入剤 Shandon PermaFluor (Thermo El ectron Corp. , MA, USA)を加えてスライドガラスに載せ、 共焦点レー ザ一蛍光顕微鏡にて観察した ' As in the immunostaining experiment, seed the transgenic cells on a cover glass and select puromycin for 2 days. Then, the cells are fixed with 3.7% Formaldehyde (10% Formalin) (Wako) PBS solution (room temperature, 10 minutes). After treatment with 1% hydrogen peroxide-containing methanol solution (room temperature, 2 minutes) and 0.1% TritonX-100-containing PBS solution (room temperature, 5 minutes), Using the In Situ Cell Death Detection Kit, Fluorescein (Roche), label the intracellular DNA cleavage site with Fluorescein-binding nucleic acid. Then, if necessary, immunostain with anti-phospho-histone H3, etc., and after washing, add fluorescent mounting agent Shandon PermaFluor (Thermo Electron Corp., MA, USA) and place it on a slide glass. Observed at '
1 0 . 細胞死 ·細胞周期解析 1 0. Cell death and cell cycle analysis
遺伝子導入細胞を puromycin選択後に回収し、 低張液 (75 mM KCl) に浮遊させ る(37°C, 20分)。 その後、 攪拌しながら固定液 (メタノール :氷酢酸 = 3 : 1混 合液) 約 3容を加えて細胞を固定する。 遠心上清除去後に再度固定液に浮遊させ る。 細胞浮遊液をスライ ドガラス上に 1、 2滴滴下し風乾する。 その後 5% Giemsa 水溶液にて核染色体を染色した。 実体顕微鏡 BHT- 323 (Olympus)にて観察し、 細 胞を間期(Go/Gl, S, G2),細胞分裂前期 (Prophase) , 細胞分裂中期 (Metaphase) , 細胞分裂後期/終期 (Anaphase/telophase) , 及び死細胞 (mitot i (? catastroph及 ぴ apoptos is)に分けて計測した。 、 The transgenic cells are collected after puromycin selection and suspended in hypotonic solution (75 mM KCl) (37 ° C, 20 minutes). Then, fix the cells by adding about 3 volumes of fixative (methanol: glacial acetic acid = 3: 1 mixture) while stirring. After removing the supernatant, resuspend it in the fixative. Drop 1 or 2 drops of cell suspension on a slide glass and air dry. Thereafter, nuclear chromosomes were stained with 5% Giemsa aqueous solution. Observed with a stereomicroscope BHT-323 (Olympus), the cells were interphase (Go / Gl, S, G2), early cell division (Prophase), metaphase (Metaphase), late phase / terminal (Anaphase / telophase), and dead cells (mitot i (? catastroph and apoptos is)).
1 1 . Microarray角罕 1 1. Microarray
理化学研究所 ·バイオリソースセンター (和光市、'埼玉、 日本) の三好浩之博 士 よ り レ ンチ ウ ィ ルス シス テ ム ( pENTR4-Hl, CS-RfA-EG, pCAG-HIVgp, pCMV - VSV- G - RSV- Rev ) の分与を受ける。 pU6- puro - RNAi ベク ター の U6 prmoter-RNAi-オリ ゴ部分を pENTR4- HI ベク ターに揷入し、 そ'の後 LR clonase (Invitrogen)にて CS-RfA-EG ベタ ターに組み込む。 pCAG-HIVgp, pCMV-VSV-G-RSV-Rev と共に、 293T細胞に transf ectamin2000にてトランスフエク ションする。翌日 Forskol in (S igma - Aldrich) lC M添加培地に置換し 24時間培養 し、 培養上清を回収する。 培養上清を Swing rotor SW28にて 19, 400 rpmで 2時 間超遠心し、 レンチウィルス粒子沈渣を回収し、約 50倍に濃縮するように培地に 溶かし、 ウィルスス トックとする(- 70°C保存)。 ACC- LC- 91細胞にレンチウィルス Hiroyuki Miyoshi of RIKEN BioResource Center (Wako City, 'Saitama, Japan) from Lentiviral Systems (pENTR4-Hl, CS-RfA-EG, pCAG-HIVgp, pCMV-VSV-G -Receive distribution of RSV- Rev). The U6 prmoter-RNAi-oligo part of the pU6-puro-RNAi vector is inserted into the pENTR4-HI vector and then incorporated into the CS-RfA-EG vector using LR clonase (Invitrogen). Transfect 293T cells with transfectamin2000 together with pCAG-HIVgp and pCMV-VSV-G-RSV-Rev. The next day, replace with medium supplemented with Forskol in (Sigma-Aldrich) lCM and incubate for 24 hours. Collect the culture supernatant. The culture supernatant is ultracentrifuged at 19, 400 rpm for 2 hours in a Swing rotor SW28, the lentiviral particle sediment is collected, dissolved in the medium so as to be concentrated about 50 times, and used as a virus stock (-70 ° C save). Lentivirus on ACC-LC-91 cells
50 X濃縮ス トック (1 X 104細胞あたり 1. 5 μ 1)を添加する。 添加後 36, 54, 72時 間後に細胞を回収し、 FACS解析、 RNA試料作製を行う。 得られた 36時間の RNA 試料 250ngから、 MMLV-RT (Agilent Technologies, Palo Alto, CA)及び T7プロ モーター配列を付加したオリゴ dTプライマーを用いて cDNAを合成する。次に T7 RNA polymeraseを含む Low RNA Fluorescent Linear Ampl ification kit (Agi lent Technologies)と、 Cyanine 3 (Cy3) - CTP及び Cyanine 5 (Cy5) - CTP (PerkinElmer, Quebec, Canada)にて、 Cy3/Cy5標識 cRNAを合成する 0 Cy3/Cy5標識 cRNAを Agi lent ol igonucleotide microarray (Whol e human genome 44K)にノヽィプリダイスさせ、 アレースキャナー(Agi lent Technologies)で、 蛍光色素量を定量し解析した。 RNAi#3及び RNAi#4の RNA試料と対照レンチウィルスの RNA試料との組み合わせ で Cy3/Cy5標識を交換 (Color swapping) して 2回行い、 平均値で RNAi後の遺伝 子変化を検討した。 Add 50 X enriched stock (1.5 μ 1 per 1 X 10 4 cells). Collect cells at 36, 54, and 72 hours after addition, and perform FACS analysis and RNA sample preparation. CDNA is synthesized from 250 ng of the obtained 36 hour RNA sample using MMLV-RT (Agilent Technologies, Palo Alto, Calif.) And an oligo dT primer to which a T7 promoter sequence is added. Then T7 Synthesis of Cy3 / Cy5-labeled cRNA using Low RNA Fluorescent Linear Amplification Kit (Agi lent Technologies) including RNA polymerase and Cyanine 3 (Cy3)-CTP and Cyanine 5 (Cy5)-CTP (PerkinElmer, Quebec, Canada) 0 Cy3 / Cy5-labeled cRNA was amplified on an Agilent oligonucleotide microarray (Whole human genome 44K), and the amount of fluorescent dye was quantified and analyzed with an array scanner (Agi lent Technologies). Two combinations of RNAi # 3 and RNAi # 4 RNA samples and control lentiviral RNA samples were exchanged (Color swapping) and performed twice, and the mean change in the genetic change after RNAi was examined.
(結果) (Result)
図 1に、ヒ ト DREF/ZBED1遺伝子のドメィン構造と、 RNAi部位及び配列を示す。 puromycin耐'! "生逾 is子 持つ plasmidべクタ一 ^pCMV-puro)に、 RNA polymerase III が転写するプロモーターであるマウス U6 プロモーターを挿入し、 さらに short-hairpin型 ol igoを挿入して DREF- RNAi系を作製した(pCMV- puro- s iDREF#l 〜#6)。 DREF- RNAi系をヒ ト肺癌細胞株にトランスフエクシヨンし、その後 24〜48 時間 puromyc in処理により遺伝子導入細胞を選択し、 DREF- RNAi作用を解析した。 また、 RNAiのために l entivirusベクターも用いた。 lentivirusベクターに同 様に short hairpin型. ol igoを挿入して、定法にしたがって 293T細胞に遺伝子導 入し、 short hairpin RNAを発現する lentivirus粒子を回収し、 それを細胞培養 液に添加して、 解析した。 ' Figure 1 shows the domain structure, RNAi site and sequence of the human DREF / ZBED1 gene. puromycin resistance '! "Plasmic vector with a ginger is child ^ pCMV-puro) Insert the mouse U6 promoter, which is a promoter transcribed by RNA polymerase III, and then insert a short-hairpin type ol igo to DREF- The RNAi system was constructed (pCMV-puro-siDREF # l to # 6) The DREF-RNAi system was transfected into a human lung cancer cell line, and then the transfected cells were selected by puromyc in treatment for 24-48 hours. In addition, the lentivirus vector was used for RNAi, and a short hairpin type.ol igo was inserted into the lentivirus vector in the same manner, and the gene was introduced into 293T cells according to a standard method. lentivirus particles expressing short hairpin RNA were collected and added to the cell culture for analysis.
図 2は、 RNAi効果の RT-PCR解析結果を示す。 肺癌細胞 A549に、 RNAiベクター pCMV-puro-s iDREF#l, #3, #4,' #5, #6を導入し、 puromycin48時間処理後に、 細 胞を採取し、 RNAを抽出し、 RT- PCRにて DREF発現を検討した。 DREF- siRNAを発 現 しな い対照ベ ク タ ー (pCMV- puro_) U6blank に比 して 、 すべて の pCMV - puro-siDREFベクターで RNAiによる発現低下が見られた。 Figure 2 shows the results of RT-PCR analysis of the RNAi effect. Introduce RNAi vector pCMV-puro-s iDREF # l, # 3, # 4, '# 5, # 6 into lung cancer cell A549, and after puromycin treatment for 48 hours, collect cells, extract RNA, RT- DREF expression was examined by PCR. Compared to the control vector that does not express DREF-siRNA (pCMV-puro_) U6blank, all pCMV-puro-siDREF vectors showed decreased expression due to RNAi.
図 3は、 RNAi効果の Western blot解析結果を示す。 RNAi効果を更に検討する ために、 DREF全長 cDNA発現ベクター、及び、 #3, #4の RNAi部位をそれぞれ s il ent mutationを起こし、 RNAi抵抗性となった DREF全長 cDNA発現ベクターを用い、 Figure 3 shows the results of Western blot analysis of the RNAi effect. To further investigate the RNAi effect, we used a DREF full-length cDNA expression vector and a DREF full-length cDNA expression vector that had undergone a silent mutation in the RNAi sites of # 3 and # 4, respectively, and became RNAi resistant.
Western blotにて DREF蛋白の発現変化を検討した。 検出は DREF cDNAの N末に 揷入した HA- tagに対する抗体である抗 HAポリクローナル抗体を用いた。 siDREF#3 #4でともに野生型 DREF タンパク質の著名な減少がみられた。 #3, #4 の s i l ent mutat ionをもつ DREF (#3mut, #4mut)は RNAiに対して抵抗性であった。 Changes in the expression of DREF protein were examined by Western blot. For detection, an anti-HA polyclonal antibody, which is an antibody against HA-tag inserted at the N-terminus of DREF cDNA, was used. siDREF # 3 In # 4, both wild-type DREF proteins showed a marked decrease. DREF (# 3mut, # 4mut) with # 3 and # 4 silent mutations was resistant to RNAi.
図 4は、 細胞周期解析 ( 1 ) の結果を示す。 肺癌細胞株 ACC- LC- 91 に PCMV-puro-s iDREF#l, #3, #4, #5, #6 (及ぴ対照ベクターの U6bl ank) を導入し、 puromycin選択後に、 PI染色の FACSにて細胞周期解析(DNA含量解析)を行った。 s iDREF-#3, #5で S期の低下が見られ G1-S移行段階での細胞周期停止 (G1停止) 傾向が見られた。又、 #4では G1が著減し G2/Mの著増が見られて、非常に強い G2/M ステップでの細胞周期停止が見られた。 #3でも G2/Mの軽度の増加が見られ、 #3 は強い G1停止と比較的軽度の G2/M停止を起こすと考えられた。 Figure 4 shows the results of cell cycle analysis (1). Lung cancer cell line ACC- LC- 91 to P CMV-puro-s iDREF # l, # 3, # 4, # 5, introduced # 6 (U6bl ank of及Pi control vector), after puromycin selection, the PI staining Cell cycle analysis (DNA content analysis) was performed with FACS. s iDREF- # 3 and # 5 showed a decrease in the S phase, and a trend toward cell cycle arrest (G1 arrest) at the G1-S transition stage. In # 4, G1 decreased significantly, G2 / M increased, and cell cycle arrest at a very strong G2 / M step was observed. A slight increase in G2 / M was also seen in # 3, and # 3 was thought to cause a strong G1 stop and a relatively mild G2 / M stop.
そこで、 以降は強い細胞周期停止作用を示した #3, #4を中心に解析した。 Henceforth, we analyzed mainly # 3 and # 4, which showed strong cell cycle arrest.
.図 5A,5B, 5Cは、 細胞周期解析 (2 ) の結果を示す。 数種の肺癌細胞株に同様に pCMV-puro-s iDREF, #3, #4 (及び U6bl ank ) を導入し細胞周期を解析した。 ACC-LC-172では、 ACC-LC-91と同様に、 #3は強い G1停止と比較的軽度の G2/M停 止を起こし、 #4では強い G2/Mでの細胞周期停止が見られた。 Calu6 ' ACC-LC-319 でも #4で G2/M停止傾向が見られた。 A549- #4 ' PC10-#3及ぴ- #4では、 Sの減少が 見られ、 G1停止傾向であった.。 また、 正常肺由来細胞株の BEAS2B及び HPL1で、 細胞周期停止はほとんど見られなかった。 Figures 5A, 5B, and 5C show the results of cell cycle analysis (2). Similarly, pCMV-puro-siDREF, # 3, # 4 (and U6blank) were introduced into several lung cancer cell lines, and the cell cycle was analyzed. In ACC-LC-172, as in ACC-LC-91, # 3 causes a strong G1 arrest and a relatively mild G2 / M arrest, while # 4 shows a strong G2 / M cell cycle arrest. It was. Calu6 'ACC-LC-319 also showed a tendency to stop G2 / M in # 4. A549- # 4 'PC10- # 3 and-# 4 showed a decrease in S and a tendency to stop G1. In addition, cell cycle arrest was hardly observed in the normal lung-derived cell lines BEAS2B and HPL1.
図 6A, 6Bは、 細胞増殖能解析結果を示す。 puromycin- se l ect ion 10日後の細胞 数を MTTァッセィ (細胞増殖キット使用) で解析した。 s iDREF- #3, #4に'より非常 に強い細胞増殖抑制作用が得られ、 抑制の強さは、 #4 の方が強いと考えられた。 # 1は弱い増殖抑制効果しか られなかった。 6A and 6B show the results of cell proliferation ability analysis. Puromycin-selection cell number after 10 days was analyzed with MTT assay (using cell proliferation kit). s iDREF- # 3 and # 4 showed a much stronger cell growth inhibitory effect than '4, and # 4 was considered to be stronger. # 1 had only a weak antiproliferative effect.
図 7は、 y H2AX (Phospho- Η2ΑΧ)、 Phospho- ATM (Serl981)染色結果を示す。 MTT 解析での増殖抑制の機序として、 DNA damage誘導の可能性を考え、 DNA損傷に反 応し活性化されるキナーゼの ATM (Serl981 リン酸化が活性化の指標)、 およびこ の ATMによりリン酸化されるヒストン Η2ΑΧ ( γ Η2ΑΧ = phospho- Η2ΑΧ)を免疫蛍光 で検討した。 ACC- LC- 91及び ACC-LC- 172でともに、 # 3、 # 4により γ Η2ΑΧシグ ナルの増加がみ られ、 特に #4 では著増 していた。 ACC- LC- 91 の FIG. 7 shows the results of staining with yH2AX (Phospho-Η2ΑΧ) and Phospho-ATM (Serl981). Considering the possibility of DNA damage induction as a mechanism of growth inhibition in the MTT analysis, ATM of the kinase activated in response to DNA damage (Serl981 phosphorylation is an activation index), and phosphorylation by this ATM Oxidized histone Η2ΑΧ (γ Η2ΑΧ = phospho- Η2ΑΧ) was examined by immunofluorescence. In both ACC-LC-91 and ACC-LC-172, γ 2ΑΧ signal increased with # 3 and # 4, especially in # 4. Of ACC-LC-91
Phospho- ATM (Serl981)染色では、 シグナルの著増がみられ、 s iDREF- #3, #4 特にPhospho- ATM (Serl981) staining showed a marked increase in signal, especially s iDREF- # 3, # 4
#4で、 DNA障害が誘導ざれ、 ATM活性化、 Ή2ΑΧ等の ATM.基質のリン酸化、 細胞周 期停止というシグナルが働いていると考えられる。 ACC- LC - 91 と比較して、 A549 · PC10の # 3による γ Η2ΑΧシグナルは、 ほぼ同程度で見られた (データ示さず)。 また、 ACC - LC- 91と比較して、 A549.PC10の # 4による γ Η2ΑΧ.シグナルは、 Α549 (土) く PC10 (+)く ACC- LC - 91 (++)であった (データ示さず)。 In # 4, DNA damage was induced, ATM activation, ATM such as Ή2ΑΧ, substrate phosphorylation, cell periphery It seems that the signal of the phase stop is working. Compared with ACC-LC-91, the γ Η2 に よ る signal from # 3 of A549 · PC10 was almost the same level (data not shown). Compared to ACC-LC-91, the signal of γ Η2ΑΧ by # 4 of A549.PC10 was Α549 (Sat) and PC10 (+) and ACC-LC-91 (++) (data shown) )
図 8は、 TUNEL解析結果を示す。 DNA断片化を検出する TUNEL解析で、 アポトー シス (細胞死) の誘導を検討した。 U6blankと比較して、 #3及び #4でアポトーシ ス誘導が見られた。アポトーシス頻度は #3及ぴ #4で差異があり、#3は A549、PC10、 ACC-LC-91及び ACC- LC- 172の 4株でほぼ同程度であつたが、 #4では、 A549は PC10 とほぼ同程度であり、 また ACC-LC- 91 は ACC - LC- 172 とほぼ同程度であり、 ACC-LC-91は A549 よりかなり大きなアポトーシス誘導を示した(一部データを示 さず)。 Figure 8 shows the TUNEL analysis results. Induction of apoptosis (cell death) was examined by TUNEL analysis to detect DNA fragmentation. Compared to U6blank, apoptosis induction was seen at # 3 and # 4. Apoptosis frequency was different between # 3 and # 4, and # 3 was almost the same in 4 strains of A549, PC10, ACC-LC-91 and ACC-LC-172, but in # 4, A549 It is almost the same as PC10, and ACC-LC-91 is almost the same as ACC-LC-172, and ACC-LC-91 showed significantly greater apoptosis induction than A549 (some data not shown) ).
図 9は、 細胞周期の変化を示す。 FACSでは G2と Mとの区別が容易ではないの で、細胞をギムザ染色して細胞周期(間期(interphase = G1, S, G2) . M期(prophase, metaphase, anaphase-telophase) ) を検討した。 ACC-LC^91 では siDREF #3, #4 で M期の増加が見られた。 また、 #4では強いクロマチン凝集を起 した M期細胞 が増加し M期での細胞死誘導.(mitotic catastroph) が起こっていることが示さ れた。 Figure 9 shows changes in the cell cycle. Since it is not easy to distinguish between G2 and M in FACS, cells are examined by Giemsa staining to examine the cell cycle (interphase (interphase = G1, S, G2). M phase (prophase, metaphase, anaphase-telophase)) did. In ACC-LC ^ 91, siDREF # 3 and # 4 showed an increase in M period. In # 4, the number of M-phase cells that caused strong chromatin aggregation increased, indicating that cell death induction (mitotic catastroph) occurred in M-phase.
図 10は、 M期での細胞死 (mitotic catastroph) を示す。 M期にリン酸化され る phospho - histon H3 (H3-P)と γ H2AXとを免疫染色し、 DNA障害 ·細胞死誘導と 細胞周期との関係を検討した。 ACC- LC - 91 では Η3- Ρ陽性の Μ期細胞の多くが γ Η2ΑΧ陽性であり、Μ期で DNA障害により細胞死が誘導されていることが示された。 図 11は、 siDREF - #3, #4の遺伝子発現解析結果を示す。 ACC-LC-91に s iDREF#3, siDREF#4の lentivirusを感染させ RNAiを誘導した。細胞周期停止(54時間及び 72時間) が起こる前の、 感染後 36時間で RNAを採取し、 対照 lentivirus感染群 の RNAを対照 RNAとして、 siDREF#3, #4によって引き起こされる遺伝子発現の変 化を検討した。この段階で変化する遺伝子群は、 DREFによって直接発現制御され、 細胞周期停止 ·細胞死誘導を引き起こしてくる遺伝子群である可能性が高い。 図 12は、 肺癌患者検体における DREFの発現を示す。 Figure 10 shows cell death (mitotic catastroph) in M phase. Phospho-histon H3 (H3-P) and γH2AX, which are phosphorylated in M phase, were immunostained to investigate the relationship between DNA damage and cell death induction and the cell cycle. ACC-LC-91 showed that most of Η3-Ρ positive Μphase cells were γΗ2ΑΧ-positive, indicating that cell death was induced by DNA damage in the Μ phase. FIG. 11 shows the results of gene expression analysis of siDREF-# 3 and # 4. ACC-LC-91 was infected with siDREF # 3 and siDREF # 4 lentivirus to induce RNAi. Changes in gene expression caused by siDREF # 3 and # 4, with RNA collected 36 hours after infection, before cell cycle arrest (54 hours and 72 hours), and using control lentivirus-infected RNA as control RNA It was investigated. The genes that change at this stage are likely to be genes that are directly regulated by DREF and cause cell cycle arrest and cell death induction. Figure 12 shows the expression of DREF in lung cancer patient specimens.
肺癌患者検体より RNeasy Mini Kit (Qiagen)にて total RNAを回収し、その total RNA 5 g力 ら Moloney Murine Leukemia Virus Reverse Transcriptase (MMLV-RT, Invitrogen)にて、 RTを施行し cDNAを合成した。 その cDNA (RNA 20ng相当)を用 いて、 DREF遺伝子発現量及ぴ内部対照として 18Sリボゾーム RNA発現量を、 図 2 の DREFプライマー及ぴ 18Sプライマーを用いて、 ABI-PRISM 7900 (ABI)にて定量 した。 有意さの検定は、 StatView (SAS)の t検定にて行った。 Total RNA was collected from lung cancer patient samples using the RNeasy Mini Kit (Qiagen). From RNA 5 g, RT was performed using Moloney Murine Leukemia Virus Reverse Transcriptase (MMLV-RT, Invitrogen) to synthesize cDNA. Using the cDNA (equivalent to 20 ng of RNA), the amount of DREF gene expression and the amount of 18S ribosomal RNA expression as an internal control were quantified using ABI-PRISM 7900 (ABI) using the DREF primer and 18S primer shown in Figure 2. did. The significance test was performed by the StatView (SAS) t test.
肺癌では全例 DREF発現が見られたのに対して、 正常肺では DREF発現が無かつ た。又、肺癌の中で組織型間での DREFの発現を比較してみると、組織型毎に DREF 発現が異なり、 特に腺癌と (腺癌、 扁平上皮癌以外の) その他の組織型の肺癌と では有意差(P= 0. 012)があった。 これらの事から、 DREF発現と肺癌発症との関連 が示唆された。 In all lung cancers, DREF expression was observed, whereas in normal lung, DREF expression was absent. In addition, when comparing DREF expression among histological types in lung cancer, DREF expression differs for each histological type, especially adenocarcinoma and lung cancer of other histological types (other than adenocarcinoma and squamous cell carcinoma) There was a significant difference between and (P = 0.012). These facts suggested an association between DREF expression and the development of lung cancer.
.図 13は、 DREF - RNAi#4によって発現抑制される遺伝子の中で、 癌の増殖に関連 すると考えられる RPL17遺伝子の DNA配列(A)と、 RPL17遺伝子の DREFによる転 写活性化の機序(B)を示す。 転写開始点より +7n1:〜 +16ntの位置に DREFタンパク 質の DNA結合モチーフ TGTCGYGAYA (Y=C又は T)にほぼ一致、 (90%)する配列が見出さ れた。 この配列に DREFが結合して RPL17 DREF発現を誘導しており、 DREF - RNAi#4 によって DREF発現が低下するために、 RPL17遺伝子も発現低下すると考えられる。 図 11 のマイクロアレイによる遺伝子発現解析結果を表 1 ( # 4) 及び表 2 ( # 3 )示す。 これらの RNAi誘導の早期(36時間)で発現が変化する遺伝子群は DREF によって直接発現制御され、 細胞周期停止、 細胞死誘導を引き起こして'くる遺伝 子群である可能性が高く、 癌治療の標的となると考えられる。 Fig. 13 shows the DNA sequence (A) of RPL17 gene, which is considered to be related to cancer growth, among the genes that are suppressed by DREF-RNAi # 4, and the mechanism of transcriptional activation by DREF of RPL17 gene. (B) is shown. A sequence almost identical (90%) to the DNA binding motif TGTCGYGAYA (Y = C or T) of the DREF protein was found at + 7n1: ~ + 16nt from the transcription start point. DREF binds to this sequence to induce RPL17 DREF expression, and DREF-RNAi # 4 reduces DREF expression, so it is thought that RPL17 gene expression also decreases. The results of gene expression analysis using the microarray in Fig. 11 are shown in Table 1 (# 4) and Table 2 (# 3). These genes whose expression changes at an early stage of RNAi induction (36 hours) are directly regulated by DREF, and are likely to be cell groups that cause cell cycle arrest and cell death induction. It is considered to be a target.
表 1 table 1
(DREF- RNAi#4によって発現抑制される遺伝子群) ― ― (A group of genes whose expression is suppressed by DREF-RNAi # 4) ― ―
Ofncia丄 Symbol: SRRM1 and Name: serine/arginine: repetitive matrix 1 Gene aliases: 160-KD; P0P101; SRM160; GC39488 . Ofncia 丄 Symbol: SRRM1 and Name: serine / arginine: repetitive matrix 1 Gene aliases: 160-KD; P0P101; SRM160; GC39488.
Summary:細胞表面分子 CM4のスプライシングの異常は癌の悪性化や浸潤能に関連 する。 SRRMl/SRtnl60はスプライシングの co-activatorであり、 癌遺伝子 RAS依存 的にこの CD のスプライシング.の異常を歸導する (Mol Cell Biol. 2006 Jan ;26(1) :362-70)。 又、 癌遺伝子 TLS/FUS と結合する (Exp Cell Res. 2003 Feb 15 ;283 (2) :184 - 95)。 癌発症との関連が強く示唆される。 Summary: Abnormal splicing of the cell surface molecule CM4 is associated with cancer malignancy and invasive potential. SRRMl / SRtnl60 is a co-activator of splicing and induces CD splicing abnormalities in an oncogene RAS-dependent manner (Mol Cell Biol. 2006 Jan; 26 (1): 362-70). It also binds to the oncogene TLS / FUS (Exp Cell Res. 2003 Feb 15; 283 (2): 184-95). The association with cancer development is strongly suggested.
Official Symbol: C0R01C and Name: coronin, actin binding protein, 1C Gene aliases: HCRNN ; coronin - 3 Official Symbol: C0R01C and Name: coronin, actin binding protein, 1C Gene aliases: HCRNN; coronin-3
Summary: シグナル伝達、 apoptosis誘導、 pre-mRNAの成熟、 細胞骨格形成等の多 様な過程に関与する TO40 ドメィンを持つ遺伝子フアミリ一の一つ。 Summary: A gene family with TO40 domains involved in various processes such as signal transduction, apoptosis induction, pre-mRNA maturation, and cytoskeleton formation.
Official Symbol: SON and Name: SON DNA binding protein Official Symbol: SON and Name: SON DNA binding protein
Gene aliases: S0N3; BASS1; DBP-5; 賺 BP; C21orf50; FLJ21099; FLJ3391 KIAA1019 Gene aliases: S0N3; BASS1; DBP-5; 賺 BP; C21orf50; FLJ21099; FLJ3391 KIAA1019
Summary: ヒト B型肝炎ウィルス(HBV)の転写制御因子として同定された。癌遺伝子 MYC family · M0Sと結合する。 Apoptosi sから細胞を保護する機能がある。 癌発症 との関連が強く示唆される。 . Summary: Identified as a transcriptional regulator of human hepatitis B virus (HBV). Oncogene MYC family · Binds to M0S. It has a function to protect cells from Apoptosi s. A strong association with cancer development is strongly suggested. .
Oif icial symbol: S^RS16 and Name: splicing factor, ar'ginine/serine-rich 16 (suppressor - of - white- apricot homolog, Drosophila) Oif icial symbol: S ^ RS16 and Name: splicing factor, ar'ginine / serine-rich 16 (suppressor-of-white- apricot homolog, Drosophila)
Gene aliases: CLASP; SWAP2; FLJ90109 Gene aliases: CLASP; SWAP2; FLJ90109
Su腿 ary:遺伝子転写後に成熟 mRNAとなる前のスプライシングを制御する。 Su thigh ary: Controls splicing before becoming mature mRNA after gene transcription.
Official Symbol: RPL17 and Name: ribosoraal protein L17 Official Symbol: RPL17 and Name: ribosoraal protein L17
Gene aliases: rpL23; GC117162 Gene aliases: rpL23; GC117162
Summary: 蛋白合成機構のリボゾーム構成分子であり 癌では蛋白合成が亢進して おり、 ; ribosomal proteinの発現亢進は癌の予後不良因子となっている。 癌発症と の関連が示唆される。 Summary: It is a ribosome component of the protein synthesis mechanism, and protein synthesis is enhanced in cancer; increased expression of ribosomal protein is a poor prognostic factor for cancer. This suggests an association with cancer development.
Oxf icial Symbol: ZBED1 and Name: zinc finger, BED - type containing 1 Gene aliases: ALTE; TRAMP; KIAA0785 Oxf icial Symbol: ZBED1 and Name: zinc finger, BED-type containing 1 Gene aliases: ALTE; TRAMP; KIAA0785
Summary: DREF遺伝子自体 Summary: DREF gene itself
Official Symbol: CTDSP1 and Name: CTD (carboxy - terminal domain, RNA polymerase II, polypeptide A) small phosphatase 1 Official Symbol: CTDSP1 and Name: CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) small phosphatase 1
Gene aliases: SCP1; NLIIF (nuclear LIM interac tor-interacting factor) 遺伝子を転写する RNA polymerase IIの活性を調節する脱リン酸化酵素。 広範な遺 伝子群の発現を制御する。 Gene aliases: SCP1; NLIIF (nuclear LIM interac tor-interacting factor) A phosphatase that regulates the activity of RNA polymerase II that transcribes the gene. It controls the expression of a wide range of gene groups.
Official Symbol: XRCCl and Name: X-ray repair complementing defective repair in Chinese hamster cells 1 Official Symbol: XRCCl and Name: X-ray repair complementing defective repair in Chinese hamster cells 1
Gene aliases: RCC Gene aliases: RCC
Summary: DNA単鎖切断の修復、 ヌクレオチド除去修復等の過程に必須な遺伝子で、 癌発症との関連が示唆される。 Summary: Genes essential for DNA single strand break repair, nucleotide excision repair, etc. It is suggested to be associated with cancer development.
Official Symbol: WDR81;及び Name: WD repeat domain 81 - Gene aliases: FLJ23776; FLJ33817 Official Symbol: WDR81; and Name: WD repeat domain 81-Gene aliases: FLJ23776; FLJ33817
Summary: シグナル伝達、 apoptosis誘導、 pre- mRNAの成熟、 細胞骨格形成等の多 様な過程に関与する WD40 ドメィンを持つ遣伝子ファミ リ一の一つ。 Summary: One of the gene families with the WD40 domain involved in various processes such as signal transduction, apoptosis induction, pre-mRNA maturation, and cytoskeleton formation.
Official Symbol: DDX21;及ぴ Name: DEAD (Asp - Glu Ala- Asp) box polypeptide 21Official Symbol: DDX21 ; and Name: DEAD (Asp-Glu Ala- Asp) box polypeptide 21
Gene aliases: GUA; GURDB; RH - II/GU; RH-II/GuA; DKFZp686F21172 Gene aliases: GUA; GURDB; RH-II / GU; RH-II / GuA; DKFZp686F21172
Summary: DEAD box ドメインを持つ RNAヘリカーゼで、 RNAの立体構造を制御する。 蛋白への翻訳の開.始、 スプライシング等 RNA に関する広範な機能の制御に関与す る。 Summary: An RNA helicase with a DEAD box domain that regulates RNA conformation. Involved in the control of a wide range of functions related to RNA, such as the initiation of translation into proteins and splicing.
Official Symbol: WDS0F1;及び Name: WD repeats and S0F1 domain containing Gene aliases: Gm83; HSPC064; GC126859; MGC138247; DKFZP56400463 Official Symbol: WDS0F1; and Name: WD repeats and S0F1 domain containing Gene aliases: Gm83; HSPC064; GC126859; MGC138247; DKFZP56400463
Summary: シグナル伝達、 apoptosis誘導、 pre- mRNAの成熟、 細胞骨格形成等の多 様な過程に関与する WD40 ドメインを持つ遺伝子ファミ,リーの一つである。 Summary: This gene family is a gene family with a WD40 domain involved in various processes such as signal transduction, apoptosis induction, pre-mRNA maturation, and cytoskeleton formation.
Official Symbol: CDV3 ;及び Name: CDV3 homo log (mouse) Official Symbol: CDV3; and Name: CDV3 homo log (mouse)
Gene aliases: H41 . Gene aliases: H41.
Summary: 癌遺伝子 erb - B2 を高発現する乳癌で発現 ¾進する遺伝子とレて同定さ れ、 CVD3 系統のマウスで、 心肥大を起こす原因遺伝子としても同定された。 細胞 増殖を促進すると考えられ、 癌発症との関連が強く示唆され 。 Summary: It was identified as a gene that promotes oncogene erb-B2 in breast cancer that highly expresses it, and was also identified as a causative gene that causes cardiac hypertrophy in CVD3 mice. It is thought to promote cell growth and is strongly suggested to be related to cancer development.
Official Symbol "· TIRAP and Name: toll-interleukin 1 receptor (TIR) domain containing adaptor protein Official Symbol "· TIRAP and Name: toll-interleukin 1 receptor (TIR) domain containing adaptor protein
Gene aliases: Mai; wyatt; FLJ42305 Gene aliases: Mai; wyatt; FLJ42305
Summary: Toll-like receptor (TLR)— 2及ぴ TLR— 4に結合し、 TRAF6 と共に NF— κΒ 活性化シグナルを伝達する。 瘙発症との関連が示唆される。 Summary: Binds to Toll-like receptor (TLR) —2 and TLR—4 and transduces NF—κΒ activation signal with TRAF6. It is suggested to be associated with epilepsy.
Official Symbol: TRADD and Name: TNFRSF1A - associated via death domain Gene aliases: Hs.89862; MGC11078 Official Symbol: TRADD and Name: TNFRSF1A-associated via death domain Gene aliases: Hs.89862; MGC11078
Summary: 細胞死を誘導する TNF受容体(TNF-R/TNFRSF1A)に結合し、 apoptosis誘 導と NF- κΒ活性化を制御しており、 Adaptor protein の TRAF2 に結合し、 TRAF2 によるアポトーシス誘導を抑制する。 癌発症との関連が示唆される。 Summary: It binds to TNF receptor (TNF-R / TNFRSF1A) that induces cell death, regulates apoptosis induction and NF-κΒ activation, binds to adapter protein TRAF2, and suppresses apoptosis induction by TRAF2 To do. It is suggested to be associated with cancer development.
Official Symbol: TSG101;及び Name: tumor susceptibility gene 101 Official Symbol: TSG101; and Name: tumor susceptibility gene 101
Gene aliases: TSG10; VPS 23 Gene aliases: TSG10; VPS 23
Summary: microtubuleの重合を制御し癌発症との関連が示唆される stathmin分子 と結合し、細胞の増殖分化を制御する。 乳癌で遺伝子変異が見られる癌抑制遺伝子 である。 Summary: Controls microtubule polymerization and binds to stathmin molecules that are implicated in the onset of cancer, and controls cell proliferation and differentiation. It is a tumor suppressor gene whose gene mutation is found in breast cancer.
Off icia丄. Symbol: EGRl;及び Name: early growth response 1 Off icia 丄. Symbol: EGRl; and Name: early growth response 1
Gene aliases: TIS8; AT225; G0S30; NGFI-A; ZNF225; KROX- 24; ZIF-268 Summary: EGRl は C2H2_zinc-f inger型の転写制御分子で、 分化 ·細胞分裂に関与 する遺伝子群の発現調節を行い、 癌発症に関与すると考えられる。 Gene aliases: TIS8; AT225; G0S30; NGFI-A; ZNF225; KROX-24; ZIF-268 Summary: EGRl is a transcriptional regulatory molecule of C2H2_zinc-finger type that regulates the expression of genes involved in differentiation and cell division It is thought to be involved in cancer development.
Official Symbol: FANCti;及ひ Name: Fanconi anemia, complementation group E Gene aliases: FAE; FACE Official Symbol: FANCti; Oihi Name: Fanconi anemia, complementation group E Gene aliases: FAE; FACE
Summary: Fanconi貧血原因遺伝子群の一つで、 DNA修復機構の構成分子である。 Summary: One of the genes responsible for Fanconi anemia, a component of DNA repair mechanism.
Official Symbol: RAD23A and Name: RAD23 homo log A (S. cerevisiae) Official Symbol: RAD23A and Name: RAD23 homo log A (S. cerevisiae)
Gene aliases: HHR23A; MGC111083 Gene aliases: HHR23A; MGC111083
Summary: DNA障害を認識し、 ヌクレオチド除去修復する機構に関与する。 Summary: Recognize DNA damage and participate in the mechanism of nucleotide excision repair.
Official Symbol: ITPKA;及び Name: inositol 1, 4, 5-trisphosphate 3 kinase A Summary:イノシトール 3燐酸 inositol 1, 4, 5-trisphosphate (IP3)は、 細胞の重 要なシグナル経路であるカルシウム細胞内濃度を上昇させる因子であり、 ITPKAは この IP3 をリン酸化して inositol 1, 3, 4, 5-tetrakisphosphate (IP(4))にして力 ルシゥム濃度の上昇を抑制する。細胞内のシグナルの制御に非常に深く関与してい る。 Official Symbol: ITPKA; and Name: inositol 1, 4, 5-trisphosphate 3 kinase A Summary: Inositol 1, 4, 5-trisphosphate (IP3) is an intracellular signal concentration of calcium that is an important signal pathway ITPKA phosphorylates this IP3 to inositol 1, 3, 4, 5-tetrakisphosphate (IP (4)) and suppresses the increase in the strength concentration. It is deeply involved in the regulation of intracellular signals.
Official Symbol: BIRC3 ·'及ぴ Name: baculoviral IAP repeat-containing 3 Gene aliases: AIP1; API2; MIHC; CIAP2; HAIP1; HIAP1; MALT2; RNF49 Official Symbol: BIRC3 · 'Pai Name: baculoviral IAP repeat-containing 3 Gene aliases: AIP1; API2; MIHC; CIAP2; HAIP1; HIAP1; MALT2; RNF49
Summary: apoptosis を抑制する分子であり、 癌で遺伝子増幅も報告されている。 癌発症との関連が強く示唆される。 Summary: It is a molecule that suppresses apoptosis, and gene amplification has also been reported in cancer. The association with cancer development is strongly suggested.
Official Symbol: P0LD1 ; 及ぴ Name: polymerase (DNA directe.d) , delta 1, catalytic subunit 125kDa Official Symbol : P0LD1 ; and Name: polymerase (DNA directe.d), delta 1, catalytic subunit 125kDa
Gene aliases: POLD Gene aliases: POLD
Summary: 細胞増殖する際に、 DNAを複製する酵素であり、 細胞増殖に必須である。 癌発症との関違が強く示唆される。 Summary: An enzyme that replicates DNA during cell growth and is essential for cell growth. The difference with the onset of cancer is strongly suggested.
Official Symbol: TRAF7;及び Name: TNF receptor-associated factor 7 Gene aliases: RFWD1; RNF119; MGC7807; DKFZp586I021 Official Symbol: TRAF7; and Name: TNF receptor-associated factor 7 Gene aliases: RFWD1; RNF119; MGC7807; DKFZp586I021
Summary: 細胞増殖制御、 細胞死誘導に関与する TNFファミ リ一受容体に結合し、 シグナル伝達に関与する。 癌発症との関違が示唆される。 Summary: It binds to the TNF family receptor involved in cell growth control and cell death induction, and is involved in signal transduction. This suggests a difference from the onset of cancer.
Official Symbol: WDR34;及び Name: WD repeat domain 34 · Official Symbol: WDR34; and Name: WD repeat domain 34
Gene aliases: MGC20486; bA216B9.3; RP11-216B9.5 ' Gene aliases: MGC20486; bA216B9.3; RP11-216B9.5 '
Summary: シグナル 達、 apoptosis誘導、 pre- mRNAの成熟、 細胞骨格形成等の多 様な過程に関与する WD40 ドメィンを持つ遺伝子フアミ リ一の一^ 3である。 Summary: This is a gene family with WD40 domain involved in various processes such as signals, apoptosis induction, pre-mRNA maturation, and cytoskeleton formation.
Oii'icial Symbol: DVLl;及ひ Name: dishevelled, dsh nomolog 1 (Drosophila) Summary: 癌の発症 '増殖の関与が報告されている Wnt- -cateninシグナル経路に あり、 -cateninの分解を抑制し Wnt- - cateninシグナルを増強する。'癌発症と の関連が強く示唆される。 Oii'icial Symbol: DVLl; Name Name: dishevelled, dsh nomolog 1 (Drosophila) Summary: Onset of cancer 'Wnt- -catenin signaling pathway that has been reported to be involved in growth, suppresses -catenin degradation and Wnt --Increase catenin signal. 'Relevant to cancer development is strongly suggested.
表 2 (DREF-RNAi#3によって発現抑制される遺伝子群) _Table 2 (A group of genes whose expression is suppressed by DREF-RNAi # 3) _
Official Symbol: EGR1;及ひ Name: early growth response 1 Official Symbol: EGR1; Oihi Name: early growth response 1
Gene aliases: TIS8; AT225; G0S30; NGFI— A; ZNF225; KROX-24; ZIF-268 Gene aliases: TIS8; AT225; G0S30; NGFI— A; ZNF225; KROX-24; ZIF-268
Summary: EGR1は C2H2- zinc- finger型の転写制御分子で、 分化 .細胞分裂に関与 する遣伝子群の発現調節を行い、 癌発症に関与すると考えられる。 Summary: EGR1 is a C2H2-zinc-finger transcriptional regulatory molecule that regulates the expression of genes involved in differentiation and cell division and is thought to be involved in cancer development.
Official Symbol: CTBP1;及ぴ Name: C - terminal binding protein 1 Official Symbol: CTBP1 ; and Name: C-terminal binding protein 1
Gene aliases: BARS; MGC104684 Gene aliases: BARS; MGC104684
Su脑 ary: アデノウィルスの癌遺伝子 E1Aの C末端結合分子である。この CtBP遣伝 子フアミリー(CtBPl'CtBP2)は、癌抑制遺伝子 ARFが p53非依存性にアポトーシス 誘導を引き起こす際の標的遺伝子であり、 CtBPの過剰発現はこの ARFの p53非依 存性アポトーシス誘導を阻害する(Mol Cell Biol. 2006 Mar ;26 (6) :2360 - 72· )。 し たがって、 この CtBPlは癌遺伝子と考えられる。 Su 脑 ary: C-terminal binding molecule of adenovirus oncogene E1A. This CtBP gene family (CtBPl'CtBP2) is a target gene when the tumor suppressor gene ARF induces apoptosis induction independent of p53. Overexpression of CtBP induces p53-independent apoptosis induction of this ARF. Inhibits (Mol Cell Biol. 2006 Mar; 26 (6): 2360-72 ·). Therefore, this CtBPl is considered an oncogene.
Official Symbol: SMARCE1;及ぴ. Name: SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily e, member 1 Official Symbol: SMARCE1 ; and. Name: SWI / SNF related, matrix associated, actin dependent regulator of chromatin, subfamily e, member 1
Gene aliases: BAF57 Gene aliases: BAF57
Summary: クロマチン構造を制御して遺伝子発現調節する SWI/SNF複合体の構成分 子である。前立腺癌の増殖を促進するアンドロゲン受容体等の核内ホルモン受容体 (nuclear receptor)の co- activatorとしても機能する。 遺伝子発現調節に深く関 与し、 前立腺癌の増殖には必須である (Mol Cell Bi。l. 2005 Mar; 25 (6): 2200-15)。 癌発症との関連が強く示唆される。 Summary: SWI / SNF complex that regulates chromatin structure and regulates gene expression. It also functions as a co-activator for nuclear hormone receptors such as androgen receptors that promote prostate cancer growth. It is deeply involved in the regulation of gene expression and essential for prostate cancer growth (Mol Cell Bi. L. 2005 Mar; 25 (6): 2200-15). The association with cancer development is strongly suggested.
Official Symbol: BAT2D1;及ぴ Name: BAT2 domain containing 1 Official Symbol: BAT2D1 ; and Name: BAT2 domain containing 1
Gene aliases: XTP2; BAT2- iso, KIAA1096 Gene aliases: XTP2; BAT2-iso, KIAA1096
Summary: 泌尿器系腫瘍で遺伝子増幅、 過剰発現が見られる遺伝子で、 癌発症との 関連が示唆される (DNA Cell Biol. 2002 Oct ;21 (10): 707-15)。 Summary: Genes with gene amplification and overexpression in urological tumors, suggesting a relationship with cancer development (DNA Cell Biol. 2002 Oct; 21 (10): 707-15).
Official Symbol: HAX1;及び Name: HCLS1 associated protein X - 1 Official Symbol: HAX1; and Name: HCLS1 associated protein X-1
Gene aliases: HS1BP1; HCLSBP1 . Gene aliases: HS1BP1; HCLSBP1.
Summary: 代表的キナーゼ型癌遺伝子 Srcファミリ一の基質であり、 多嚢胞腎の原 因遺伝子 PKD2と結合して機能する。 癌発症との関連が示唆される。 Summary: This is a substrate of the representative kinase-type oncogene Src family and functions by binding to PKD2, the gene responsible for polycystic kidney disease. It is suggested to be associated with cancer development.
Ofr lcial Symbol: PPAT ; 及 ぴ Name: phosphoribosyl pyrophosphate ami aotransf erase Ofrcial Symbol: PPAT ; and Name: phosphoribosyl pyrophosphate ami aotransf erase
Gene aliases: GPAT; ATASE Gene aliases: GPAT; ATASE
Summary: DNA の材料となるプリ ン核酸の生合成酵素の一つで、 細胞増殖と関違す る。 Summary: One of the biosynthetic enzymes of purine nucleic acid, which is a material for DNA, and is related to cell growth.
Official Symbol: STK40;及ひ Name: serine/threonine kinase 40 Official Symbol: STK40; Oihi Name: serine / threonine kinase 40
Gene aliases: SHIK; SgK495; MGC4796; RP11-268J15.4 Gene aliases: SHIK; SgK495; MGC4796; RP11-268J15.4
Summary: 癌遺伝子 p65RELAや代表的癌抑制遺伝子 p53による転写制御を抑制する (Biochem Biophys Res Cora匪. 2003 Oct 3; 309 (4): 774-8) 0 癌発症との関連が示 唆される。 Summary: Repression of transcriptional regulation by oncogene p65RELA and typical tumor suppressor gene p53 (Biochem Biophys Res Cora 匪. 2003 Oct 3; 309 (4): 774-8) 0 The association with cancer development is suggested.
Official Symbol: SUV39H2;及ぴ Name: suppressor of variegation 3-9 homolog 2 (Drosophila) Official Symbol: SUV39H2 ; and Name: suppressor of variegation 3-9 homolog 2 (Drosophila)
Gene aliases: F1J23414 Gene aliases: F1J23414
Summary: SUV39Hファミリ一(SUV39H1、 SUV39H2)遣伝子で、 ヘテロクロマチン形成 を誘導する histone H3-lysine9 のメチル化を誘導する。 代表的癌抑制遺伝子 Retinoblastoma (RB)フアミリーの RB2/pl30に結合し、 RB2による転写調節を促進 する。 癌発症との関連が示唆される。 , Summary: The SUV39H family (SUV39H1, SUV39H2) gene induces the methylation of histone H3-lysine9, which induces heterochromatin formation. It binds to RB2 / pl30 of the typical tumor suppressor gene Retinoblastoma (RB) family and promotes transcriptional regulation by RB2. It is suggested to be associated with cancer development. ,
Ofr icial Symbol: PTGS2; 及ひ ame: prostaglandin - endoperoxide synthase 2Ofricial Symbol: PTGS2; Oi ame: prostaglandin-endoperoxide synthase 2
^prostaglandin G/H synthase and cyclooxygenase ^ prostaglandin G / H synthase and cyclooxygenase
Gene aliases: C0X2; COX - 2; PHS - 2; PGG/HS; PGHS-2; hCox-2 Gene aliases: C0X2; COX-2; PHS-2; PGG / HS; PGHS-2; hCox-2
Summary: prostaglandin 生合成に関与する分子であるが、 癌での発現宂進が見ら れ、 癌の発症 ·進展,転移との関連が強く示唆されている。 Summary: Although it is a molecule involved in prostaglandin biosynthesis, its expression has been promoted in cancer, and its relation to cancer onset / progress and metastasis has been strongly suggested.
Ofricial Symbol: PARPl;及び Name: poly (ADP-ribose) polymerase family, memoer 1 Ofricial Symbol: PARPl; and Name: poly (ADP-ribose) polymerase family, memoer 1
Gene aliases: PARP; PPOL; ADPRT; ADPRT1; PARP-1; pADPRT-1 Gene aliases: PARP; PPOL; ADPRT; ADPRT1; PARP-1; pADPRT-1
Summary: 細胞増殖 ·分化 ·癌化等のプロセスや、 DNA 損傷反応等で、 核内分子を poly(ADP-ribosyl)化する酵素であり、 癌発症との関連が強く示唆される。 Summary: It is an enzyme that converts nuclear molecules into poly (ADP-ribosyl) by processes such as cell proliferation, differentiation, canceration, and DNA damage reaction, and its relationship with cancer development is strongly suggested.
Official Symbol: CEBPD;及ぴ Name: CCAAT/enhancer binding protein (C/EBP) , delta Official Symbol: CEBPD ; and Name: CCAAT / enhancer binding protein (C / EBP), delta
Gene aliases: CELF; CRP3; C/EBP - delta; NF -] 6- beta Gene aliases: CELF; CRP3; C / EBP-delta; NF-] 6- beta
Summary: 代表的な転写因子で、 免疫反応 ·炎症反応に関連する遺伝子群の発現に 関与する。 Summary: A representative transcription factor that is involved in the expression of genes related to immune and inflammatory responses.
Official Symbol: HK2;及ぴ Name: hexokinase 2 Official Symbol: HK2 ; and Name: hexokinase 2
Gene aliases: HKII; HXK2; DKFZp686 1669 Gene aliases: HKII; HXK2; DKFZp686 1669
Summary: ブドウ糖分解に関与する酵素で、 増殖速度の速い癌細胞でのプドウ糖分 解亢進に関与する。 癌発症との関連が強く示唆される。 Summary: It is an enzyme involved in glucose degradation and is involved in the enhancement of pudosaccharide degradation in cancer cells with a high growth rate. The association with cancer development is strongly suggested.
Official Symbol: C1QDC1;及.び Name: Clq domain containing 1 Official Symbol: C1QDC1 ; and Name: Clq domain containing 1
Gene aliases: EEG1; EEG-1; FLJ11391; FLJ22569; MGC102894 Gene aliases: EEG1; EEG-1; FLJ11391; FLJ22569; MGC102894
細胞が分化する過程で発現誘導される遺伝子で、細胞増殖を抑制する(JBiol Chem.It is a gene whose expression is induced during cell differentiation and suppresses cell proliferation (JBiol Chem.
2004 Jan 16; 279 (3): 1916— 21)。 2004 Jan 16; 279 (3): 1916-21).
Ofticial Symbol: GCET2 j及ひ Name: germinal center expressed transcript 2 Gene aliases: HGAL; GCAT2; MGC40441 Ofticial Symbol: GCET2 jOihi Name: germinal center expressed transcript 2 Gene aliases: HGAL; GCAT2; MGC40441
Summary: B細胞性悪性リンパ腫で発現亢進が見られ、 interleukin-4で発現誘導さ れる遺伝子である。 リ ンパ腫等の腫瘍発症との関連が示唆される。 Summary: This gene is upregulated in B-cell malignant lymphoma and is induced by interleukin-4. It is suggested to be related to the onset of tumors such as lymphoma.
Official Symbol: ACTG1;及ぴ Name: actin, gamma 1 Official Symbol : ACTG1 ; and Name: actin, gamma 1
Gene aliases: ACT; ACTG; DFNA20; DFNA26; beta - actin Gene aliases: ACT; ACTG; DFNA20; DFNA26; beta-actin
Summary:.代表的細胞骨格ァクチン分子の一つで、 細胞の運動性に関与する。 癌の 浸潤転移との関連も示唆される。 Summary: One of the representative cytoskeletal actin molecules involved in cell motility. It is also suggested that it is associated with cancer invasive metastasis.
Official Symbol: PLAGL2;及び Name: pleiomorphic adenoma gene-like 2 Gene aliases: FLJ23283 Official Symbol: PLAGL2; and Name: pleiomorphic adenoma gene-like 2 Gene aliases: FLJ23283
Summary: 腫瘍における染色体転座で活性化される PLAG ファミ リーに属する分子 で、 白血病の発症にも関与する(Blood. 2005 Apr 1; 105 (7): 2900-7) 0 癌発症との 関連が強く示唆される。 Summary: Molecules belonging to the PLAG family activated by chromosomal translocations in tumors In also involved in the development of leukemia (Blood 2005 Apr 1; 105 ( 7):. 2900-7) 0 associated with cancer development is suggested strongly.
Official Symbol : NC0A6;及ぴ Name: nuclear receptor coact ivator 6 Official Symbol: NC0A6 ; and Name: nuclear receptor coact ivator 6
Gene aliases : NRC ; AIB3 ; ASC2 ; PRIP ; TRBP ; RAP250 ; KIAA0181 Gene aliases: NRC; AIB3; ASC2; PRIP; TRBP; RAP250; KIAA0181
Summary : 核内ホルモン受容体に結合し co-activator として機能する。クロマチン 構造を制御するヒス トンァセチル化酵素 ·メチル化酵素とも結合し、 広範な遺伝子 群の転写制御する。 Summary: It binds to a nuclear hormone receptor and functions as a co-activator. It also binds to histone acetylase and methylase that control chromatin structure and regulates transcription of a wide range of genes.
Official Symbol: UHRF1;及ぴ Name: ubiquitin-l ike, containing PHD and RING finger domains, 1 Official Symbol: UHRF1 ; and Name: ubiquitin-l ike, containing PHD and RING finger domains, 1
Gene aliases : Np95 ; ICBP90 ; RNF106 ; huNp95; FLJ21925 ; MGC138707 Gene aliases: Np95; ICBP90; RNF106; huNp95; FLJ21925; MGC138707
Summary :プロモーターの CCAAT box に結合し転写制御し、 細胞増殖に相関して発 現する遣伝子。 癌細胞の増殖の促進 (Mol Biol Cell. 2005 Dec; 16 (12): 5621-9) ゃ抗アポト一シス作用 (Ann N Y Acad Sci. 2004 Dec; 1030: 355-60. ) が報告され ており、 癌発症との関連が強く示唆される。 . Summary: A gene that binds to the promoter's CCAAT box, regulates transcription, and correlates with cell growth. Promotion of cancer cell growth (Mol Biol Cell. 2005 Dec; 16 (12): 5621-9) Anti-apoptotic action (Ann NY Acad Sci. 2004 Dec; 1030: 355-60.) Has been reported. The association with the onset of cancer is strongly suggested. .
Official Symbol : DUSP6;及び Name : dual specificity phosphatase 6 Official Symbol: DUSP6; and Name: dual specificity phosphatase 6
Gene aliases : MKP3 ; PYST1 Gene aliases: MKP3; PYST1
Summary: キナーゼ分子の作用を抑制する脱リン酸化酵素であり、 MAP キナーゼフ ァミ リ ' Summary: A phosphatase that suppresses the action of kinase molecules and is a MAP kinase family.
(MAP /ERK, SAPK/JNK, p38)の機能を抑制し、 癌抑制遺伝子と考えられる。 It suppresses the function of (MAP / ERK, SAPK / JNK, p38) and is considered a tumor suppressor gene.
Official Symbol: SFRS8;及び Name : spl icing factor, arginine/serine-rich 8 (suppressor - of- white-apricot homolog, Drosophila) Official Symbol: SFRS8; and Name: spl icing factor, arginine / serine-rich 8 (suppressor-of- white-apricot homolog, Drosophila)
Gene al iases: SWAP Gene al iases: SWAP
Summary : 遺伝子転写後に成熟 mRNAとなる前のスプライシングを制御し、癌で異常 の見られる fibronecti n、 CD45 等のスプライシングとの関与が報告されている。 癌発症との関連が示唆される。 Summary: It has been reported that splicing of fibronectin, CD45, etc., which is abnormal in cancer, is controlled by controlling splicing before gene transcription after gene transcription. It is suggested to be associated with cancer development.
産業上の利用可能性 . Industrial applicability.
本発明による DREFを標的とする癌治療は、正常組織に実質的に影響を及ぼすこ となく、癌細胞に対して非常に強い細胞増殖抑制、 M期での細胞死誘導、及び G2/M 期での非常に強い細胞周期停止作用を有するため、 癌の退縮と細胞死に非常に有 効である。 , The cancer treatment targeting DREF according to the present invention has a very strong suppression of cell proliferation, induction of cell death in M phase, and G2 / M phase without substantially affecting normal tissues. It has a very strong cell cycle arresting effect in cancer and is very effective for cancer regression and cell death. ,
本発明は、 DREFを分子標的とする癌治療を可能とする。 すなわち、 本発明の治 療剤は、 癌の退縮と細胞死に非常に有効であるため、 DREFを発現する癌の治療に 有効に使用できる。 The present invention enables cancer treatment using DREF as a molecular target. That is, since the therapeutic agent of the present invention is very effective for cancer regression and cell death, it can be effectively used for the treatment of cancer expressing DREF.
本明細書で引用した全ての刊行物、 特許および特許出願をそのまま参考として 本明細書にとり入れるものとする。 All publications, patents and patent applications cited in this specification are used as is for reference. Incorporated herein.
Claims
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2008538780A JP5176035B2 (en) | 2006-10-06 | 2007-10-05 | Composition for cancer treatment using DREF as molecular target |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2006-275695 | 2006-10-06 | ||
| JP2006275695 | 2006-10-06 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2008044787A1 true WO2008044787A1 (en) | 2008-04-17 |
Family
ID=39282971
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/JP2007/070031 Ceased WO2008044787A1 (en) | 2006-10-06 | 2007-10-05 | Composition for treating cancer using dref as molecular target |
Country Status (2)
| Country | Link |
|---|---|
| JP (1) | JP5176035B2 (en) |
| WO (1) | WO2008044787A1 (en) |
Cited By (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2019093502A1 (en) * | 2017-11-09 | 2019-05-16 | 国立大学法人 東京医科歯科大学 | Inhibitor of the expression of cancer-promoting factors, screening method for active ingredient thereof, expression cassette useful in said method, diagnostic drug, and diagnostic method |
| CN109777800A (en) * | 2017-11-15 | 2019-05-21 | 信雅生物科技(苏州)有限公司 | It is a kind of be capable of specificity inhibit ZBED1 gene siRNA construction method and its application |
| US12066429B2 (en) | 2016-05-10 | 2024-08-20 | National University Corporation Tokyo Medical And Dental University | Method for inhibiting the expression of inflammation promoting factors |
| CN118910050A (en) * | 2024-07-19 | 2024-11-08 | 威海市立医院 | ShRNA (short hairpin ribonucleic acid) for targeted silencing of ALIX gene expression, lentiviral expression vector and application |
-
2007
- 2007-10-05 JP JP2008538780A patent/JP5176035B2/en not_active Expired - Fee Related
- 2007-10-05 WO PCT/JP2007/070031 patent/WO2008044787A1/en not_active Ceased
Non-Patent Citations (3)
| Title |
|---|
| OHSHIMA N. ET AL.: "Identification of a human homologue of the DREF transcription factor with a potential role in regulation of the histone H1 gene", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 278, no. 25, 2003, pages 22928 - 22938, XP003021394 * |
| SANO Y. ET AL.: "Tensha Inshi hDREF ha Saibo Shuki Shinko ni Kanyo suru", DAI 28 KAI ANNUAL MEETING OF THE MOLECULAR BIOLOGY SOCIETY OF JAPAN KOEN YOSHISHU, 2005, pages 278, XP003021395 * |
| YAMASHITA D. ET AL.: "hDREF regulates cell proliferation and expression of ribosomal protein genes", MOLECULAR AND CELLULAR BIOLOGY, vol. 27, no. 6, 2007, pages 2003 - 2013, XP003021396 * |
Cited By (11)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US12066429B2 (en) | 2016-05-10 | 2024-08-20 | National University Corporation Tokyo Medical And Dental University | Method for inhibiting the expression of inflammation promoting factors |
| WO2019093502A1 (en) * | 2017-11-09 | 2019-05-16 | 国立大学法人 東京医科歯科大学 | Inhibitor of the expression of cancer-promoting factors, screening method for active ingredient thereof, expression cassette useful in said method, diagnostic drug, and diagnostic method |
| CN111315882A (en) * | 2017-11-09 | 2020-06-19 | 国立大学法人东京医科齿科大学 | Cancer-promoting factor expression inhibitor, method for screening active ingredient thereof, expression cassette useful for the method, diagnostic agent, and diagnostic method |
| JPWO2019093502A1 (en) * | 2017-11-09 | 2021-02-25 | 国立大学法人 東京医科歯科大学 | Cancer promoter expression inhibitor, screening method for its active ingredient, expression cassette, diagnostic agent, and diagnostic method useful for the method. |
| JP7376873B2 (en) | 2017-11-09 | 2023-11-09 | 国立大学法人 東京医科歯科大学 | Cancer-promoting factor expression inhibitor, method for screening its active ingredient, expression cassette useful for the method, diagnostic agent, and diagnostic method |
| IL274456B1 (en) * | 2017-11-09 | 2024-04-01 | Univ Nat Corp Tokyo Medical & Dental | Inhibitor of expression of pro-carcinogenic factors, screening method for an active ingredient thereof, expression cassette used in this method, diagnostic drug and diagnostic method |
| IL274456B2 (en) * | 2017-11-09 | 2024-08-01 | Univ Nat Corp Tokyo Medical & Dental | Inhibitor of expression of pro-carcinogenic factors, screening method for an active ingredient thereof, expression cassette used in this method, diagnostic drug and diagnostic method |
| CN111315882B (en) * | 2017-11-09 | 2024-09-24 | 国立大学法人东京医科齿科大学 | Cancer promoting factor expression inhibitor, screening method for its active ingredient, expression cassette useful for the method, diagnostic drug and diagnostic method |
| US12253515B2 (en) | 2017-11-09 | 2025-03-18 | National University Corporation Tokyo Medical And Dental University | Method for inhibiting the expression of cancer-promoting factors |
| CN109777800A (en) * | 2017-11-15 | 2019-05-21 | 信雅生物科技(苏州)有限公司 | It is a kind of be capable of specificity inhibit ZBED1 gene siRNA construction method and its application |
| CN118910050A (en) * | 2024-07-19 | 2024-11-08 | 威海市立医院 | ShRNA (short hairpin ribonucleic acid) for targeted silencing of ALIX gene expression, lentiviral expression vector and application |
Also Published As
| Publication number | Publication date |
|---|---|
| JPWO2008044787A1 (en) | 2010-02-18 |
| JP5176035B2 (en) | 2013-04-03 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Okino et al. | BRCA1-associated protein 1 (BAP1) deubiquitinase antagonizes the ubiquitin-mediated activation of FoxK2 target genes | |
| Kim et al. | MicroRNA-494 downregulates KIT and inhibits gastrointestinal stromal tumor cell proliferation | |
| Kanehira et al. | Oncogenic role of MPHOSPH1, a cancer-testis antigen specific to human bladder cancer | |
| Hsu et al. | Notch1 pathway-mediated microRNA-151-5p promotes gastric cancer progression | |
| JP7376873B2 (en) | Cancer-promoting factor expression inhibitor, method for screening its active ingredient, expression cassette useful for the method, diagnostic agent, and diagnostic method | |
| Okada et al. | Oncogenic role of NALP7 in testicular seminomas | |
| Hong et al. | Hsa_circ_0074298 promotes pancreatic cancer progression and resistance to gemcitabine by sponging miR-519 to target SMOC | |
| Lee et al. | MZF-1/Elk-1 interaction domain as therapeutic target for protein kinase Cα-based triple-negative breast cancer cells | |
| EP2229946B1 (en) | Use of the growth-stimulating protein KIAA1524 | |
| Lefkimmiatis et al. | p73 and p63 sustain cellular growth by transcriptional activation of cell cycle progression genes | |
| Sun et al. | Cyclin-dependent kinase 4 may be expressed as multiple proteins and have functions that are independent of binding to CCND and RB and occur at the S and G2/M phases of the cell cycle | |
| Nakamura et al. | Overexpression of C16orf74 is involved in aggressive pancreatic cancers | |
| WO2008044787A1 (en) | Composition for treating cancer using dref as molecular target | |
| Nakamura et al. | Novel roles for LIX1L in promoting cancer cell proliferation through ROS1-mediated LIX1L phosphorylation | |
| Song et al. | CBX8 acts as an independent RNA-binding protein to regulate the maturation of miR-378a-3p in colon cancer cells | |
| Wang et al. | Long non-coding RNA UCA1 promotes retinoblastoma progression by modulating the miR-124/c-myc axis | |
| Zhu et al. | LncRNA FAM66C inhibits pancreatic cancer progression by sponging miR-574-3p | |
| Costa et al. | Impact of MUC1 mucin downregulation in the phenotypic characteristics of MKN45 gastric carcinoma cell line | |
| Okamoto et al. | KDM2A-dependent reduction of rRNA transcription on glucose starvation requires HP1 in cells, including triple-negative breast cancer cells | |
| JP5645171B2 (en) | Cell growth inhibitor and screening method thereof | |
| CN101987197A (en) | Method and reagent for inhibiting invasiveness of cancer cells | |
| JP6057408B2 (en) | Cancer preventive and / or therapeutic agent | |
| US20240043557A1 (en) | A platform to obtain monoclonal antibodies directed against processed tumor-specific antigens | |
| Tholanikunnel et al. | Novel mechanisms in the regulation of G protein-coupled receptor trafficking to the plasma membrane | |
| JP4797159B2 (en) | Composition for inhibiting cancer metastasis |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 07829766 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2008538780 Country of ref document: JP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 07829766 Country of ref document: EP Kind code of ref document: A1 |