WO2004048565A1 - アポトーシス関連蛋白質およびその用途 - Google Patents
アポトーシス関連蛋白質およびその用途 Download PDFInfo
- Publication number
- WO2004048565A1 WO2004048565A1 PCT/JP2003/014794 JP0314794W WO2004048565A1 WO 2004048565 A1 WO2004048565 A1 WO 2004048565A1 JP 0314794 W JP0314794 W JP 0314794W WO 2004048565 A1 WO2004048565 A1 WO 2004048565A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- amino acid
- disease
- seq
- acid sequence
- salt
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4747—Apoptosis related proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P11/00—Drugs for disorders of the respiratory system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P19/00—Drugs for skeletal disorders
- A61P19/08—Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- 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
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/06—Immunosuppressants, e.g. drugs for graft rejection
-
- 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
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2500/00—Screening for compounds of potential therapeutic value
Definitions
- the present invention relates to a functional fragment of a protein that binds to and activates ASK1, and various uses of the protein and the fragment, in particular, pharmaceutical uses.
- Apoptosis plays a role in the removal of unnecessary and abnormal cells during development, homeostasis, and a host defense reaction to remove injured cells, and the mechanism at the molecular level is gradually increasing. It is becoming clear. Abnormality of these molecules and disruption of the control mechanism impair the physiological function of apoptosis, and may cause various diseases and exacerbate them. For example, if apoptosis is excessively suppressed, the cells that should be removed will proliferate abnormally and induce neoplastic diseases and autoimmune diseases, etc. Conversely, if apoptosis is abnormally enhanced, the cells that are essentially required It can lead to death and neurodegenerative diseases.
- Mitogen-activated protein (MAP) kinase cascade is a MAP kinase kinase that is activated by physiochemical stress and inflammatory cytokines such as tumor necrosis factor-T (TNF- ⁇ ) and inulin-leukin_1 (IL-1).
- TNF- ⁇ tumor necrosis factor-T
- IL-1 inulin-leukin_1
- MAKKK MAP kinase
- MAPKK MAP kinase
- apoptosis Indicates a phenotype such as C-Jun N-terminal kinase (JNK) and p38 MAP kinase (p38) are known as MAPKs that are part of the signaling pathway that induces apoptosis (for example, Science, 270, 1326 (1995) See). Furthermore, it is involved in inducing the inflammatory response by inducing the production of inflammatory site force-in.
- JNK and p38 are derived from MAPKK MKK4 / 7 and MKK3Z6 Are activated respectively. These MAP KKs are activated by one MAP KKK called Apoptosis signal-regulating kinase 1 (ASK 1) (JP 10-93A, Science, 275, 90-94 (1997)).
- ASK1 Apoptosis signal-regulating kinase 1
- MAPKKK has been reported in large numbers in addition to ASK1, ASK1 is characterized by its ability to induce apoptosis in cells through signaling through activation of JNK and Z or p38.
- ASK 1 activation is involved in cell differentiation such as keratinocyte differentiation and neurite outgrowth of PC12 cells. It is becoming clear that it is playing a role. Furthermore, it has become clear that induction of the production of inflammatory cytokines is involved in inducing an inflammatory response.
- ASK 1 is an important molecule that determines the future fate of cells, it is thought that various factors are involved in its activation and that it is under complex control. So far, it has been reported that activation of ASK 1 requires the formation of homo-oligomers between ASK 1 and subsequent phosphorylation of threonine in the activation loop. Although mainly due to autophosphorylation by ASK1, the existence of another kinase has also been suggested (Journal of Cellular Physiology, 191, 95-104 (2002)).
- rotein phosphatase 5 (PP 5) is bonded directly to AS K 1 under H 2 0 2 stimulation, AS K 1 which is activated by dephosphorylation threonine believed to revert to the inactive state Yes (BO Journal, 20, 6028-6036 (2001)).
- thioredoxin a redox regulator, constantly binds to the N-terminal domain of ASK 1 and acts as an ASK 1 activation inhibitor in the absence of oxidative stress.
- ASK 1 activation (EMBO Journal, 17, 2596-2606 (1998)), and TNF receptor-associated activation of ASK 1 causes TNF receptor-associated factor 2 (T RAF 2 ) Binds to the C-terminal domain of ASK 1 to activate ASK 1 (Molecular Cell, 2, 389-395 (1998)); 14-3-3 proteins bind to the C-terminal domain Inhibits ASK 1 activation (Proceedings of National Academy of Sciencies, USA, 96, 8511-8515 (1999)).
- ASK1 is closely related to ER stress-induced apoptosis because treatment of ASK1 knockout mouse cells with ER stress inducers significantly suppresses apoptosis compared to wild-type mouse cells. Therefore, ASK1 inhibitors such as thioredoxin and 14-3-3 protein, ASK1 dominant negative mutants, and ASK1 antisense oligonucleotides described above are neurodegenerative diseases (eg, polyglutamine). Disease has been suggested to be effective in the prevention and treatment of endoplasmic reticulum stress-related diseases (W002 / 38179).
- an object of the present invention is to provide novel knowledge on the activation mechanism of ASK1 and the mechanism of induction of apoptosis-induced Z inflammatory response through it. That is, a first object of the present invention is to identify a novel ASK1 binding protein which has not been known so far and to clarify the mechanism of ASK1 activation control by the protein. Another object of the present invention is to provide a novel preventive and therapeutic means for various diseases involving ASK1, based on the interaction between the protein and ASK1. Disclosure of the invention
- the present inventors considered that human ASK1 full-length cDNA was used as bait, and an expression library derived from human fetal brain was screened by yeast two-hybrid method.
- an expression library derived from human fetal brain was screened by yeast two-hybrid method.
- PGR1 an expression library derived from human fetal brain was screened by yeast two-hybrid method.
- ABP 1 an ASK1-binding protein.
- the present inventors have found that the protein not only binds to ASK1, but also activates ASK1 and its downstream JNK and p38, and induces caspase-dependent apoptosis. .
- the present inventors have conducted further studies based on these findings, and as a result, have completed the present invention.
- a peptide or a salt thereof which contains the same or substantially the same amino acid sequence as a part of the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, and is capable of activating ASK 1;
- a method for producing a peptide or a salt thereof which comprises culturing the transformant according to the above [7] and collecting the peptide or a salt thereof according to the above [1] from the obtained culture.
- [9] contains an amino acid sequence identical or substantially identical to a part of the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, and does not activate ASK 1; Or a peptide or a salt thereof which can be inactivated,
- ASK 1 comprising a protein having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, the peptide according to the above [1], or a salt thereof.
- Activation accelerator
- a medicine comprising the protein having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 or the peptide according to the above [1], or a salt thereof,
- the disease is selected from the group consisting of cancer, autoimmune disease, viral infection, endocrine disease, blood disease, organ hyperplasia, restenosis after angioplasty, and recurrence after cancer resection
- the disease is selected from the group consisting of cancer, autoimmune disease, viral infection, endocrine disease, blood disease, organ hyperplasia, restenosis after angioplasty, and recurrence after cancer resection
- a protein containing the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, or a polypeptide containing the nucleotide sequence encoding the peptide described in [1] above ASK 1 activation promoter comprising nucleotides,
- the disease is selected from the group consisting of cancer, autoimmune disease, viral infection, endocrine disease, blood disease, organ hyperplasia, restenosis after angioplasty, and recurrence after cancer resection [20]
- An appointment comprising a polynucleotide containing a nucleotide sequence encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 or a part thereof Diagnostics for diseases associated with cis or inflammation,
- Diseases include cancer, autoimmune disease, viral infection, endocrine disease, blood disease, organ dysplasia, restenosis after angioplasty, recurrence after cancer resection, transplant rejection, graft-versus-host disease, Immune deficiency, neurodegenerative disease, ischemic heart disease, radiation damage, ultraviolet light disorder, toxic disease, nutritional disorder, inflammatory disease, ischemic neuropathy, diabetic neuropathy, vascular disease, respiratory disease and cartilage disease
- [24] contains a nucleotide sequence complementary to a nucleotide sequence encoding a protein containing the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, or a part thereof
- An ASK 1 activation inhibitor comprising a polynucleotide
- a drug comprising a nucleotide
- Diseases include viral infections, endocrine diseases, blood diseases, organ dysplasia, transplanted organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative diseases, ischemic heart disease, radiation damage, ultraviolet damage, medium Selected from the group consisting of toxic diseases, nutritional disorders, inflammatory diseases, ischemic neuropathies, diabetic neuropathies, vascular diseases, respiratory diseases and cartilage diseases.
- amino acid sequence identical or substantially identical to the amino acid sequence shown in SEQ ID NO: 2 or 4 characterized by being capable of specifically recognizing the amino acid sequence shown in SEQ ID NO: 5 or 6
- a diagnostic agent for an apoptosis or inflammation-related disease comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 or a salt thereof,
- the disease is cancer, autoimmune disease, viral infection, endocrine disease, blood disease, organ dysplasia, restenosis after angioplasty, recurrence after cancer resection, transplant rejection, graft-versus-host disease , Immunodeficiency, neurodegenerative disease, ischemic heart disease, radiation damage, ultraviolet light disorder, toxic disease, nutritional disorder, inflammatory disease, ischemic neuropathy, diabetic neuropathy, vascular disease, respiratory disease and cartilage
- the diagnostic agent of the above-mentioned [30] which is selected from the group consisting of diseases;
- an ASK1 activation inhibitor comprising an antibody against a protein or a salt thereof having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4,
- a medicament comprising an antibody against a protein or a salt thereof having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4,
- Diseases include viral infection, endocrine disease, blood disease, organ dysplasia, transplanted organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative disease, ischemic heart disease, radiation damage, ultraviolet light damage, medium Selected from the group consisting of toxic diseases, nutritional disorders, inflammatory diseases, ischemic neuropathies, diabetic neuropathies, vascular diseases, respiratory diseases and cartilage diseases
- a protein containing the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, or the peptide or the salt thereof according to the above [1], and ASK 1 or N-terminal The method according to the above-mentioned [38], wherein binding to a partial peptide containing an activation control domain or a salt thereof is measured.
- a kit for screening for an ASK 1 activation modulator comprising:
- kit according to [40] further comprising a partial peptide thereof containing ASK 1 or an N-terminal activation control domain, a salt thereof, or a cell producing the same.
- Activation of ASK1 or the partial peptide or a salt thereof in a cell producing ASK1 or a partial peptide thereof containing an N-terminal activation control domain and a kinase domain or a salt thereof is performed by: (1) SEQ ID NO: 2 or SEQ ID NO: (2) SEQ ID NO: 2 or SEQ ID NO: 4 in the presence of a protein containing the same or substantially the same amino acid sequence as the amino acid sequence shown in 4, or the peptide of the above [1] or a salt thereof.
- a polynucleotide comprising a nucleotide sequence encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, or a polynucleotide containing the same, or SEQ ID NO: 2 or
- a polynucleotide comprising a nucleotide sequence encoding a protein having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, or a polynucleotide containing a part thereof, or SEQ ID NO: 2 Or a screening kit for an ASK1 activation regulating substance, comprising an antibody against a protein having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 4 or a salt thereof,
- the disease is selected from the group consisting of cancer, autoimmune disease, viral infection, endocrine disease, blood disease, organ hyperplasia, restenosis after angioplasty, and recurrence after cancer resection [49] The medicine described.
- an apoptotic or inflammatory cytokine comprising a substance having the same or substantially the same amino acid sequence as the SEQ ID NO: 2 or SEQ ID NO: 4 or a substance that reduces the expression or activity of a salt thereof Production inhibitor,
- a medicament comprising a substance that decreases the expression or activity of a protein or a salt thereof having the same or substantially the same amino acid sequence as the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4,
- Diseases include viral infection, endocrine disease, blood disease, organ dysplasia, transplanted organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative disease, ischemic heart disease, radiation damage, ultraviolet light damage, medium
- the drug according to the above [53] which is selected from the group consisting of a toxic disease, a nutritional disorder, an inflammatory disease, an ischemic neuropathy, a diabetic neuropathy, a vascular disease, a respiratory disease, and a cartilage disease.
- amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 comprising the ASK 1 partial peptide or a salt thereof, which contains the N-terminal activation control domain of ASK 1 and does not contain the kinase domain.
- a pharmaceutical comprising an ASK 1 partial peptide or a salt thereof that does not contain
- Diseases include viral infection, endocrine disease, blood disease, organ dysplasia, transplanted organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative disease, ischemic heart disease, radiation damage, ultraviolet light damage, medium 41.
- the medicament according to claim 40 which is selected from the group consisting of a toxic disease, a nutritional disorder, an inflammatory disease, an ischemic neuropathy, a diabetic neuropathy, a vascular disease, a respiratory disease, and a cartilage disease.
- An ASK1 activation inhibitor comprising the peptide of the above-mentioned [9] or a salt thereof,
- a medicament comprising the peptide of the above-mentioned [9] or a salt thereof,
- Diseases include viral infection, endocrine disease, blood disease, organ dysplasia, transplanted organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative disease, ischemic heart disease, radiation damage, ultraviolet light damage, medium
- the present invention provides the medicament of the above-mentioned [63], which is selected from the group consisting of toxic diseases, nutritional disorders, inflammatory diseases, ischemic neuropathies, diabetic neuropathies, vascular diseases, respiratory diseases and cartilage diseases.
- FIG. 1 shows the alignment of the human (top row) and mouse (bottom row) ABP 1 amino acid sequences.
- the portion surrounded by a square indicates the partial amino acid sequence used as an antigen for producing an antibody.
- the boxes show the partial sequences used as antigenic peptides for the production of the two antibodies of the present invention (the eight antibodies! ⁇ ⁇ 1 antibody).
- FIG. 2A shows the tissue distribution of ABP 1 mRNA expression in mouse tissues.
- FIG. 2B shows the daily change of ABP1 mRNA expression in mouse fetal tissues.
- FIG. 3 shows the amount of plasmid-dependent increase in detection intensity when the ABP1 protein is overexpressed by transfecting HEK293 cells with a plasmid expressing untagged ABP1.
- Cells were collected 24 hours after gene transfer, each cell extract was divided into two, SDS-PAGE was performed, and Imno plot (IB) was performed using the ELA antibody and the LVR antibody.
- ABPl_pcDNA3 (-) indicates non-transformed HEK293 cells, and the gradient indicates that the amount of plasmid increases from left to right.
- FIGS. 4A-B show the results of a binding test between ABP1 and ASK1.
- Figure 4A Transfection of F1ag-ABP1 and Myc-ASK1 plasmids into HEK293 cells, harvested cells 24 hours later, immunoprecipitated with anti-F1ag antibody, Blot analysis was performed.
- Fig. 4B Transfection of F1ag-ASK1 and CFP-ABP1 plasmids into HEK293 cells, harvested cells 24 hours later, immunoprecipitation with anti-Flag antibody, and immunoblot analysis with anti-GFP antibody was.
- CFP is a mutant of GFP and can be recognized by an anti-GFP antibody.
- FIG. 4A the right two lanes with B, H 2 0 2 treatment (0. 5 mM, 1 hour) was also performed.
- the lower part shows a part of each cell lysate before immunoprecipitation, which was separately electrophoresed.
- IP stands for immunoprecipitation
- IB stands for immunoblot.
- FIG. 5A shows a schematic diagram of the ASK1-deficient mutant.
- An HA tag was added to the N-terminal of each plasmid containing the ASK1-deficient mutant.
- Amino acid numbers 678-936 (shaded area) indicate a kinase domain.
- FIG. 5B shows the results of a binding test between ABP1 and an ASK1-deficient mutant. The lower panel shows a portion of each cell lysate before immunoprecipitation, which was separately electrophoresed. IP represents immunoprecipitation, and IB represents immunoblot.
- FIGS. 6A-B show cell death induction by ABP1 in HeLa cells.
- Figure 6A Fluorescence microscope image (upper) and differential interference image (lower) of the same field of view 36 hours after transfection of CFP or CFP-ABP1 plasmid into HeLa cells.
- Figure 6B Percentage of cell death (%) 36 hours after gene transfer.
- FIG. 7A-B show ABP1-induced cell death in PAE-ABP1 cells.
- Figure 7 A PAE-ABP1 cells in the presence (+) and absence (-) of tetracycline (Tet) Shows a phase-contrast microscope image 36 hours after culturing.
- FIG. 7B Percentage of cell death (%) after culture in the presence of tetracycline (O h) and at 24 and 48 hours after tetracycline removal.
- FIG. 8 shows ABP1-induced cell death (DNA fragmentation) in PAE-ABP1 cells.
- DNA fragmentation DNA fragmentation
- FIGS. 9A-B show the caspase dependence of cell death by ABP1.
- Figure 9A PAE-ABP1 cells were cultured without tetracycline and caspase-3 activity was measured after 0, 12, 24, and 36 hours.
- FIGS. 10A-B show cell death induction by ABP1-deficient mutants.
- FIG. 10A shows a schematic diagram of an ABP1-deficient mutant. A CFP evening was added to the N-terminal side in the same manner as in the wild type.
- Figure 10B The percentage (%) of cell death 36 hours after gene transfer in cells into which ABP1-deficient mutants have been introduced.
- Figures 11A-B show the activation of 31 ⁇ 1, JNK :, p38 according to 88-1.
- Fig. 11 A ⁇ _ ⁇ cells were cultured after removing tetracycline, and endogenous JNK and ⁇ 38 were activated by immunoblot analysis using each anti-phosphorylation protein antibody.
- Fig. 11B PAE-ABP1 cells were cultured without tetracycline, and the activation of endogenous ASK1 was examined by immunoblot analysis using an anti-phosphorylation protein antibody. IB stands for immunoblot.
- ABSP 1 of the present invention is a protein containing an amino acid sequence identical or substantially identical to the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4.
- the ABP 1 of the present invention can be prepared from warm-blooded animals (eg, humans, mice, rats, guinea pigs, hamsters, puppies, higgs, goats, bushes, puppies, puppies, birds, cats, dogs, sal, chimpanzees).
- warm-blooded animals eg, humans, mice, rats, guinea pigs, hamsters, puppies, higgs, goats, bushes, puppies, puppies, birds, cats, dogs, sal, chimpanzees.
- Cells e.g., hepatocytes, spleen cells, nerve cells, glial cells, splenic iS cells, bone marrow cells, mesangial cells, Langerhans cells, epidermal cells, epithelial cells, goblet cells, endothelial cells, smooth muscle cells, Fibroblasts, fiber cells, muscle cells, fat cells, immune cells (e.g., macrophages, T cells, B cells, natural killer cells, mast cells, neutrophils, basophils, eosinophils, monocytes) , Megakaryocytes, synovial cells, chondrocytes, bone cells, osteoblasts, osteoclasts, breast cells, hepatocytes or stromal cells, or precursors of these cells, stem cells or cancer cells) Or any tissue where these cells are present, such as the brain, parts of the brain (eg, olfactory bulb, amygdala, basal sphere, hippocampus, thalamus, hypo
- substantially identical amino acid sequence to the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, about 70% or more, preferably about 70% or more to the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 Amino acid sequences having a homology of 80% or more, more preferably about 90% or more, particularly preferably about 95% or more, and most preferably about 98% or more.
- proteins containing an amino acid sequence substantially identical to the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 include, for example, the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 described above. Containing substantially the same amino acid sequence as that of SEQ ID NO: 2 or SEQ ID NO: 4; Proteins having activity are preferred.
- substantially the same activity examples include, for example, ASK1 or a group of kinases located downstream thereof (eg, MKK4 / 7, MKK3 / 6, JNK, p38, etc.), the activity promoting activity, and the induction of cell apoptosis. Activity and the like. Substantially the same means that the properties are qualitatively (eg, physiologically or pharmacologically) the same. Therefore, it is preferable that the activities such as promotion of ASK1 cascade activation are the same, but the quantitative factors such as the degree of these activities and the molecular weight of the protein may be different (for example, About 0.1 to 100 times, preferably about 0.1 to 10 times, and more preferably 0.5 to 2 times).
- the ASK 1 cascade activation promoting activity can be measured by a known method, for example, using a labeled phosphate group donor to ASK 1 or a kinase group located downstream thereof (eg, MKK4 / 7, MKK3 / 6, JM, p38, etc.), and apoptosis-inducing activity should be measured by measuring cell death induction rate, morphological observation of cells, detection of DNA fragmentation, etc. Can be.
- a labeled phosphate group donor to ASK 1 or a kinase group located downstream thereof eg, MKK4 / 7, MKK3 / 6, JM, p38, etc.
- apoptosis-inducing activity should be measured by measuring cell death induction rate, morphological observation of cells, detection of DNA fragmentation, etc. Can be.
- ABP 1 of the present invention includes, for example, 1) 1 or 2 or more (preferably about 1 to 30, preferably 1 to 1) in the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4; An amino acid sequence in which about 0, more preferably a number (1 to 5) amino acids have been deleted; (2) one or two or more amino acids (preferably An amino acid sequence having about 30 amino acids, preferably about 1 to 10 amino acids, and more preferably a number of (1 to 5) amino acids; 3 1 or 2 amino acid sequences shown in SEQ ID NO: 2 or SEQ ID NO: 4 An amino acid sequence into which at least (preferably about 1 to 30, preferably about 1 to 10, and more preferably about 1 to 5) amino acids have been inserted, SEQ ID NO: 2 or SEQ ID NO: 4 1 or 2 or more of the amino acid sequences shown in More preferably, an amino acid sequence in which about 1 to 30 amino acids, preferably about 1 to 10 amino acids, and more preferably a number (1 to 5) amino acids are substituted with another amino acid, or
- ABP 1 of the present invention is preferably human ABP 1 (ABP 1) having the amino acid sequence shown in SEQ ID NO: 2 or mouse ABP 1 (mABP 1) having the amino acid sequence shown in SEQ ID NO: 4, Or its homologue in other warm-blooded animals (eg, rats, guinea pigs, hamsters, egrets, higgins, goats, bush, eel, poma, birds, cats, dogs, monkeys, chimpanzees, etc.).
- ABSP 1 human ABP 1
- mABP 1 mouse ABP 1
- hABPl is a protein consisting of 127 amino acids that is encoded by a known gene (GenBank accession number: AF116272) derived from a human T cell designated as PGR1, but its function has not been reported so far.
- Mouse ABP1 is a 125 amino acid protein that is encoded in mouse brain-derived cDNA (GenBank accession number: AB041651) named clone MNCb-1039, but also reports on its function. There is no.
- ABP 1 of the present invention including a protein containing the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, has a C-terminal lipoxyl group (one CO ⁇ H), carboxylate (one CO O one) , Amide (—CONH 2 ) or ester (_CO ⁇ R).
- R in the ester e.g., methyl, Echiru, n- propyl, isopropyl, alkyl groups such as n- butyl; for example, cyclopentyl, C 3 _ 8 cycloalkyl group such as cyclohexyl; e.g., phenyl, Facial C 6 _ 12 ⁇ Li Ichiru groups, such as single naphth le; for example, benzyl, phenylene root ⁇ preparative 2 alkyl group such as phenethyl; «- 7 _ such as ⁇ - Nafuchiru ( ⁇ _ 2 alkyl group such as naphthylmethyl 14 aralkyl group; pivaloxyloxymethyl group and the like are used.
- ABP 1 has a carbonyl group (or carboxylate) other than at the C-terminus
- ABP 1 in which the carboxyl group is amidated or esterified is also included in ABP 1 of the present invention.
- the ester for example, the above-mentioned C-terminal ester and the like are used.
- amino acid residues e.g., Mechionin residues
- N-terminal Amino groups protecting group eg, C WINCH such formyl group, such as Asechiru group ( ⁇ _ 6 Al force Noiru 6- Glycyl group)
- N-terminal glutamine residue formed by cleavage in vivo, pyroglutamine oxidation
- Substituent on the side chain of amino acid in the molecule eg, one SH, amino group, imidazo Ichiru group, I command Ichiru group, Guanijino group, etc.
- a suitable protecting group e.g., formyl group, such as C E one 6 Ashiru groups such as 6 Arukanoiru groups such Asechi Le group
- complex proteins such as so-called glycoproteins to which sugar chains are bound.
- the present invention provides a peptide having the partial amino acid sequence of ABP1 described above, and having substantially the same activity as ABP1.
- substantially the same activity has the same meaning as described above.
- the “substantially the same activity” can be measured in the same manner as described above.
- the partial peptide is hereinafter referred to as “activating peptide of the present invention”.
- the activating peptide of the present invention is not particularly limited as long as it has the above properties.
- the activating peptide of the present invention has about 60 amino acids or more, preferably about 60 to about 1 Peptides having the same or substantially the same amino acid sequence as the partial amino acid sequence consisting of 100 amino acids, more preferably about 60 to about 80 amino acids, and the like.
- the partial amino acid sequence may be an N-terminal sequence of ABP1, a C-terminal sequence, or an internal sequence. Alternatively, a combination of these partial sequences may be used.
- the activating peptide of the present invention comprises at least about 60 amino acids on the N-terminal side of the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, more preferably about 60 to about 100 amino acids, particularly It preferably contains a partial amino acid sequence of about 60 to about 80 amino acids.
- the activating peptide of the present invention may show higher activity (eg, ASK1 cascade activation promoting activity, apoptosis inducing activity, etc.) than the full-length protein in some cases.
- partial peptides of ABP 1 include those that can function as (antagonistic) inhibitors of ABP 1 or the “activating peptide of the present invention”. Examples of such partial peptides include those which have binding activity to ASK1 but cannot activate the kinase. In the present specification, the partial peptide is hereinafter referred to as “the inhibitory peptide of the present invention”.
- the inhibitory peptide of the present invention contains the same or substantially the same amino acid sequence as a part of the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, and does not activate or inhibit ASK1. It is a peptide that can be activated.
- the inhibitory peptide include those containing a partial amino acid sequence consisting of about 35 amino acids or less in the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4, and preferably containing a partial amino acid sequence on the N-terminal side.
- the partial peptide of ABP1 of the present invention (including both the activating peptide of the present invention and the inhibitory peptide of the present invention; hereinafter may be simply abbreviated as “partial peptide of the present invention”) is a C-terminal. May be any of a carbonyl group (—COOH), a carboxylate (one COO—), an amide (—CONH 2 ), or an ester (—COOR).
- R in the ester include the same as described above for ABP1.
- these peptides have a carboxyl group (or carboxylate) other than the C-terminus, those in which the carbonyl group is amidated or esterified are also included in the partial peptide of the present invention.
- the ester in this case, for example, the above-mentioned C-terminal ester or the like is used.
- the partial peptides of the present invention include those in which the amino group of the N-terminal methionine residue is protected with a protecting group, and those in which the N-terminal side is cleaved in vivo to form G 1 Also includes those in which n is pyroglutamine-oxidized, those in which the substituent on the side chain of the amino acid in the molecule is protected by an appropriate protecting group, and those which are combined with a sugar chain such as a so-called glycopeptide. It is.
- Examples of the salt of ABP1 or a partial peptide thereof of the present invention include physiologically acceptable salts with an acid or a base, and a physiologically acceptable acid addition salt is particularly preferable.
- Such salts include, for example, inorganic acids (eg, hydrochloric acid, phosphoric acid, bromide water) Salts with organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzene) And sulfonic acid).
- inorganic acids eg, hydrochloric acid, phosphoric acid, bromide water
- Salts with organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid
- ABP1 or a salt thereof of the present invention can be prepared from a warm-blooded animal cell or tissue by the above-mentioned method for purifying a protein known per se. Specifically, the tissue or cells of a warm-blooded animal are homogenized, and the soluble fraction is separated and purified by chromatography such as reverse phase chromatography, ion exchange chromatography, affinity chromatography, etc. ABP1 or a salt thereof can be produced.
- ABP1 of the present invention or a partial peptide thereof or a salt thereof can also be produced according to a known peptide synthesis method.
- the peptide synthesis method may be, for example, either a solid phase synthesis method or a liquid phase synthesis method.
- the target protein can be produced by condensing a partial peptide or amino acid capable of constituting ABP 1 with the remaining portion and, when the product has a protecting group, removing the protecting group.
- condensation and the elimination of the protecting group are performed according to a method known per se, for example, the methods described in the following 1 to 5.
- the ABPs 1 thus obtained can be isolated and purified by a known purification method.
- purification methods include, for example, solvent extraction, distillation, and column chromatography. Chromatography, liquid chromatography, recrystallization, and combinations thereof.
- the free form can be converted into an appropriate salt by a known method or a method analogous thereto, and conversely, the protein (peptide) is converted into a salt.
- the salt can be converted to a free form or another salt by a known method or a method analogous thereto.
- commercially available resins for protein synthesis can be used for the synthesis of the ABP1s of the present invention.
- Such resins include chloromethyl resin, hydroxymethyl resin, benzhydrylamine resin, aminomethyl resin, 4-benzyloxybenzyl alcohol resin, 4-methylbenzhydrylamine resin, PAM resin, 41 Hydroxymethyl methylphenylacetamidomethyl resin, polyacrylamide resin, 4- (2,4, dimethoxyphenylhydroxymethyl) phenoxy resin, 4- (24'dimethoxyphenyl Fmocaminoethyl) phenoxy resin, etc.
- a resin an amino acid having an amino group and a side chain functional group appropriately protected can be converted into a target protein or peptide (hereinafter sometimes collectively referred to as “protein etc.”) by itself.
- the resin is condensed on the resin according to various known condensation methods. At the end of the reaction, proteins and the like are cut out from the resin, and at the same time, various protecting groups are removed. Further, an intramolecular disulfide bond formation reaction is carried out in a highly diluted solution to obtain a target protein or the like or an amide thereof.
- the protected amino acids described above various activating reagents that can be used for protein synthesis can be used, and carbodiimides are particularly preferable.
- carbopimides DCC, N, N, diisopropyl carbopimide, N-ethyl-N,-(3-dimethylaminoprolyl) carbopimide, and the like are used.
- the protected amino acids may be added directly to the resin along with the racemization inhibitor additives (eg, H tBt, HOOBt), or symmetric anhydrides or HOBt esters or HOOBt esters. Can be added to the resin after the protected amino acid is activated in advance.
- Solvents used to activate protected amino acids and condense with resins are used in protein condensation reactions. It can be appropriately selected from solvents known to be usable.
- acid amides such as N, N-dimethylformamide, N, N-dimethylacetamide, N-methylpyrrolidone, halogenated hydrocarbons such as methylene chloride, chloroform, and trifluoroethanol.
- Alcohols, sulfoxides such as dimethyl sulfoxide, amines such as pyridine, ethers such as dioxane and tetrahydrofuran, nitriles such as acetonitrile and propionitrile, esters such as methyl acetate and ethyl acetate, or esters thereof.
- the reaction temperature is appropriately selected from the range known to be usable for the protein bond formation reaction, and is usually selected from the range of about 120 to 5 as appropriate.
- the activated amino acid derivative is usually used in a 1.5 to 4-fold excess.
- the protection of the functional group which should not be involved in the reaction of the raw materials, the protecting group, the elimination of the protective group, the activation of the functional group involved in the reaction, and the like can be appropriately selected from known groups or known means.
- Examples of the protecting group for the amino group of the starting material include Z, Boc, Yuichi Sharipentyloxycarbonyl, Isoporiloxycarbonyl, 4-methoxybenzyloxycarbonyl, C11Z, Br-Z, a Damantyloxycarbonyl, trifluoroacetyl, phthaloyl, formyl, 2-ditrophenylsulfenyl, diphenylphosphinothioyl, Fmoc and the like are used.
- the carboxyl group may be, for example, an alkyl esterified (eg, methyl, ethyl, propyl, butyl, tert-butyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 2-adamantyl, etc.) Or cyclic alkyl esterification), aralkyl esterification (eg, benzyl ester, 412-nitrobenzyl ester, 4-methoxybenzyl ester, 4-cyclobenzyl ester, benzhydryl esterification), phenacyl ester , Ben It can be protected by ziroxycarbonyl hydrazide, tertiary butoxycarbonyl hydrazide, trityl hydrazide and the like.
- an alkyl esterified eg, methyl, ethyl, propyl, butyl, tert-butyl, cyclopent
- the hydroxyl group of serine can be protected, for example, by esterification or etherification.
- a group suitable for this esterification for example, a lower alkanol group such as an acetyl group, an aroyl group such as a benzoyl group, a group derived from carbonic acid such as a benzyloxycarbonyl group, an ethoxycarponyl group, and the like are used.
- Examples of a group suitable for etherification include a benzyl group, a tetrahydropyranyl group, and a t-butyl group.
- the protecting group of the phenolic hydroxyl group of tyrosine for example, B z 1, C 1 2 - Bz l, 2- nitrobenzyl, B r- Z, such as data one tert-butyl is used.
- the protecting group for imidazole of histidine for example, Tos, 4-methoxy-2,3,6-trimethylbenzenesulfonyl, DNP, benzyloxymethyl, Bum, Boc, Trt, Fmoc and the like are used.
- the acid treatment for example, anisol, phenol, thioanisole, methacrylol, paracresol, dimethylsulfide, 1,4-butanedithiol, 1,2-1 It is effective to add a thione scavenger such as ethanedithiol, etc.
- a thione scavenger such as ethanedithiol, etc.
- the imidazole protecting group of histidine and The 2,4-dinitrophenyl group used in the reaction is removed by thiophenol treatment, and the formyl group used as an indole protecting group for tributofan is an acid in the presence of the above-mentioned 1,2-ethanedithiol and 1,4-butanedithiol.
- the activated carbonyl group of the raw material include, for example, the corresponding acid anhydride, azide, and active ester [alcohol (eg, pentachlorophenol, 2,4,5-trichlorophenol, 2,4 Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphenolimide, and esters with H ⁇ Bt).
- alcohol eg, pentachlorophenol, 2,4,5-trichlorophenol, 2,4 Dinitrophenol, cyanomethyl alcohol, paranitrophenol, HONB, N-hydroxysuccinimide, N-hydroxyphenolimide, and esters with H ⁇ Bt.
- the activated amino group of the raw material for example, a corresponding phosphoric amide is used.
- an amide form of a protein or the like for example, first, after amidating and protecting the ⁇ -hydroxyl group of the amino acid at the carboxy terminal, a peptide (protein) chain is added to the amino group side with a desired chain length. After that, a protein or the like from which only the protecting group of the amino group at the ⁇ -terminal of the peptide chain was removed and a protein or the like from which only the protecting group of the carboxyl group at the C-terminus was removed were prepared.
- the protein and the like are condensed in the above-mentioned mixed solvent. Details of the condensation reaction are the same as described above.
- the crude protein or the like is purified by various known purification means, and the main fraction is freeze-dried to obtain an amide such as a desired protein.
- an ester such as a protein
- the desired protein or the like can be obtained in the same manner as an amide such as a protein.
- An ester form can be obtained.
- the partial peptide of the present invention or a salt thereof can also be produced by cleaving the peptide 1 or a salt thereof with an appropriate peptidase.
- ABPs 1 of the present invention can be obtained by culturing a transformant containing DNA encoding ⁇ 1 or a partial peptide thereof, and isolating and purifying ⁇ ⁇ 1 from the resulting culture. It can also be manufactured.
- Examples of the DNA encoding ABP 1 or a partial peptide thereof of the present invention include genomic DNA, genomic DNA library, human or other warm-blooded animals (for example, human, mouse, rat, guinea pig, hamster, rabbit, sheep) , Goat, bush, Poma, bird, cat, dog, monkey, chimpanzee, etc., all cells (eg, spleen cells, nerve cells, glial cells, england
- RNA or mRNA fraction prepared from the cells and tissues described above can be directly amplified by reverse transcriptase polymerase chain reaction (hereinafter abbreviated as "RT-PCR"). .
- RT-PCR reverse transcriptase polymerase chain reaction
- Examples of the DNA encoding ABP 1 of the present invention include a DNA containing the nucleotide sequence shown in SEQ ID NO: 1 or SEQ ID NO: 3, or a DNA containing the nucleotide sequence shown in SEQ ID NO: 1 or SEQ ID NO: 3 and a high string An activity that is substantially the same as a protein that contains a nucleotide sequence that hybridizes under gentle conditions and that contains the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4 (eg, ASK1 cascade activation promoting activity) , Apoptosis-inducing activity, etc.).
- the DNA eg, SEQ ID NO: 1 or SEQ ID Has a homology of about 50% or more, preferably about 60% or more, more preferably about 70% or more, particularly preferably about 80% or more, and most preferably about 90% or more with the nucleotide sequence shown in No. 3
- a DNA containing a base sequence or the like is used.
- Hybridization can be performed by a method known per se or a method analogous thereto, for example, a method described in Molecular 'Cloning (Molecular Cloning) 2nd edition (J. Sambrook et al., Cold Spring Harbor Lab. Press, 1989). And so on. When a commercially available library is used, hybridization can be performed according to the method described in the attached instruction manual. Hybridization can be performed preferably under high stringent conditions. '
- High stringency conditions refer to, for example, conditions in which the sodium concentration is about 19 to 40 mM, preferably about 19 to 20 mM, and the temperature is about 50 to 70 ° C, preferably about 60 to 65 ° C. In particular, it is preferable that the sodium concentration is about 19 mM and the temperature is about 65.
- the DNA encoding ABP1 of the present invention is preferably a hABP1 DNA containing the nucleotide sequence shown in SEQ ID NO: 1 or a mABP1 DNA containing the nucleotide sequence shown in SEQ ID NO: 3, or other warm blood. And homologs thereof in animals (eg, rats, guinea pigs, hamsters, porcupines, sheep, goats, pigs, porcupines, pomas, birds, cats, dogs, monkeys, chimpanzees, etc.).
- animals eg, rats, guinea pigs, hamsters, porcupines, sheep, goats, pigs, porcupines, pomas, birds, cats, dogs, monkeys, chimpanzees, etc.
- the DNA encoding the partial peptide of the present invention includes a base sequence encoding the same or substantially the same amino acid sequence as a part of the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO: 4. If it is, any thing may be used. Further, any of genomic DNA, genomic DNA library, cDNA derived from the above-described cells and tissues, cDNA library derived from the above-mentioned cells and tissues, and synthetic DNA may be used.
- the vector used for the library may be any of bacteriophage, plasmid, cosmid, phagemid and the like. Alternatively, the mRNA can be directly amplified by RT-PCR using an mRNA fraction prepared from the cells and tissues described above. Specifically, as the DNA encoding the partial peptide of the present invention, for example,
- DNA encoding a peptide having an activity of inhibiting a protein containing an amino acid sequence encoded by the DNA is used. .
- Examples of the DNA that can hybridize with the nucleotide sequence shown in SEQ ID NO: 1 or SEQ ID NO: 3 under high stringent conditions include, for example, about 60% or more, and preferably about 70% or more with the corresponding portion in the nucleotide sequence. More preferably, a polynucleotide containing a nucleotide sequence having a homology of about 80% or more, most preferably about 90% or more is used.
- the DNA encoding ABP1 of the present invention or a partial peptide thereof may be amplified by PCR using a synthetic DNA primer having a part of the nucleotide sequence encoding the protein or peptide, or may be appropriately expressed.
- the DNA incorporated in the vector can be cloned by hybridization with a DNA fragment encoding a part or the entire region of the protein of the present invention or with a labeled synthetic DNA. it can. Hybridization can be performed, for example, according to the method described in Molecular Cloning, 2nd edition (described above). When a commercially available library is used, hybridization can be carried out according to the method described in the instruction manual attached to the library.
- the DNA base sequence can be determined using known kits, such as Mutan TM -super Express Km (Takara Shuzo Co., Ltd.), Mutan TM -K (Takara Shuzo Co., Ltd.), and the like.
- the conversion can be carried out according to a method known per se, such as the duplex method or the Kunkel method, or a method analogous thereto.
- the cloned DNA can be used as it is depending on the purpose, or after digestion with a restriction enzyme, if desired, or after adding a linker.
- the DNA may have ATG as a translation initiation codon at its 5 'end and TAA, TGA or TAG as a translation termination codon at its 3' end. These translation start codon and translation stop codon can be added using an appropriate synthetic DNA adapter.
- the DNA expression vector encoding ABP1 or a partial peptide thereof of the present invention can be obtained, for example, by cutting out a DNA fragment of interest from DNA encoding ABP1,
- It can be produced by ligating a DNA fragment downstream of a promoter in an appropriate expression vector.
- Escherichia coli-derived plasmids eg, pBR322, pBR
- plasmid derived from Bacillus subtilis eg, pUBll0, TP5, pC194
- plasmid derived from yeast eg, pSH19, pSHl
- Bacteriophages such as ⁇ phage
- Animal viruses such as retrovirus, vaccinia virus, and baculovirus
- ZCMV ZCMV, pRcZRSV, pcDNAIZNeo, and the like are used.
- Any promoter may be used as long as it is an appropriate promoter corresponding to the host used for gene expression.
- SR promoter when the host is an animal cell, SR promoter, SV40 promoter, LTR promoter, CMV (cytomegalovirus) promoter, H
- An SV-TK promoter or the like is used. Among them, CMV promoter, S
- R promoter is preferred.
- the host is a bacterium belonging to the genus Escherichia;
- re cA promoter Isseki one, AP L promoter one, lpp promoter, T7 flop Romo - evening - and the like are preferable. If the host is Bacillus, SP01 promoter, SP02 promoter First, pen P promoter is preferred.
- PHO5 promoter When the host is yeast, PHO5 promoter, PGK promoter, GAP promoter, ADH promoter, etc. are preferred.
- a polyhedrin promoter When the host is an insect cell, a polyhedrin promoter, a P10 promoter and the like are preferred.
- the expression vector may contain, in addition to the above, an enhancer, a splicing signal, a polyA addition signal, a selection marker, and an SV40 replication origin (hereinafter sometimes abbreviated as SV40 ori), if desired.
- an enhancer e.g., a splicing signal
- a polyA addition signal e.g., a polyA addition signal
- a selection marker e.g., SV40 replication origin
- SV40 ori SV40 replication origin
- the selection marker include a dihydrofolate reductase (hereinafter sometimes abbreviated as dh fr) gene [methotrexate (MTX) resistance] and an ampicillin resistance gene (hereinafter abbreviated as Amp 1).
- Amp ampicillin resistance gene
- Ne of, G418 resistance neomycin resistance gene
- a signal sequence suitable for the host may be added to the N-terminal side of the protein of the present invention. If the host is a bacterium belonging to the genus Escherichia, a PhoA signal sequence, an immediate A signal sequence, etc .; if the host is a bacillus, an a-amylase signal sequence, a subtilisin signal sequence, etc .; If the host is yeast, MF signal sequence, SUC2 signal sequence, etc .; if the host is an animal cell, insulin 'signal sequence, human interferon. Signal sequence, antibody molecule, signal sequence, etc. are used, respectively. .
- the transformant containing "DNA encoding ABP 1 of the present invention or a partial peptide thereof" obtained as described above is transformed into a host with an expression vector containing the DNA according to a known method. It can be manufactured by doing.
- the expression vectors include those described above.
- Escherichia bacteria for example, Escherichia bacteria, Bacillus bacteria, yeast, insect cells, insects, animal cells, and the like are used.
- Bacillus spp. include, for example, Bacillus subtilis MI 114 (Gene, 24, 255 (1983)), 207-21 (Journal of Biochemistry) 95, 87 (1984)].
- yeast examples include, for example, Saccharomyces cerevisiae AH22, AH22R-, NA87-11A, DKD-5D, 20B-12, Schizosaccharomyces pombe NC YC 1913, NC YC2036 And Pichia pastoris K M71.
- Insect cells include, for example, when the virus is Ac NPV, a cell line derived from the larvae of night moth (Spodoptera frugiperda cell; Sf cell), MG1 cells derived from the midgut of Trichoplusia ni, and eggs derived from eggs of Trichoplusia ni High Five TM cells, cells derived from Mamestra brassicae, cells derived from Esiigmena acrea and the like are used.
- Sf cells include Sf9 cells (ATCC CRL1711) and Sf21 cells (Vaughn, JL et al., In Vivo, 13, 213-217, (1977)) and the like. Is used.
- insects for example, silkworm larvae are used [Maeda et al., Nature, 315, 592 (1985)].
- animal cells include monkey cells C ⁇ S_7, Vero, Chinese hamster cells CHO (hereinafter abbreviated as CHO cells), and dh fr gene-deficient chicks—zhamster cells CHO (hereinafter CH ⁇ (dh fr—) Cells), mouse L cells, mouse AtT-20, mouse myeloid cells, rat GH3, and human FL cells.
- Transformation can be performed according to a known method depending on the type of host. Proc. Natl. Acad. Sci. USA, Proc. Natl. Acad. Sci. USA, Vol. 69, Proc. Transformation can be carried out according to the method described in 21 10 (1972) or Gene, 17, 107 (1982).
- Bacillus spp. Can be transformed, for example, according to the method described in Molecular & General Genetics, 168, 111 (1979) and the like.
- Yeasts are described, for example, in Methods in Enzyniology, Vol. 194, 182—187 (1991), Processings of the National Academy of Sciences, Ob. Transformation can be performed according to the method described in U.S.A. (Proc. Natl. Acad. Sci. USA), 75, 1929 (1978).
- Insect cells and insects can be transformed according to the method described in, for example, Bio / Technology, 6, 47-55 (1988).
- Animal cells can be transformed, for example, according to the method described in Cell Engineering Separate Volume 8, New Cell Engineering Experiment Protocol. 263-267 (1995) (published by Shujunsha), Virology, 52, 456 (1973). can do.
- the transformant can be cultured according to a known method depending on the type of the host.
- a liquid medium is preferably used as the culture medium.
- the medium preferably contains a carbon source, a nitrogen source, inorganic substances, etc. necessary for the growth of the transformant.
- a carbon source for example, glucose, dextrin, soluble starch, sucrose, etc .
- a nitrogen source for example, ammonium salts, nitrates, corn steep, liquor, peptone, casein, meat extract, soybean meal, Inorganic or organic substances such as potato extract; inorganic substances include, for example, calcium chloride, sodium dihydrogen phosphate, and magnesium chloride.
- the medium may be supplemented with yeast extract, vitamins, growth promoting factors and the like.
- the H of the medium is preferably about 5-8.
- an M9 medium containing glucose and casamino acid (Miller, Journal of Experimenin) Molecular Genetics, 431-433, Cold Spring Harbor Laboratory, New York 1972).
- an agent such as, for example, 3; 3-indolylacrylic acid may be added to the medium to make the mouth motor work efficiently.
- Culture of a transformant whose host is a bacterium belonging to the genus Escherichia is usually about 15 to 43, and is performed for about 3 to 24 hours. If necessary, ventilation or stirring may be performed.
- Burkholder's minimal medium As a medium for culturing a transformant in which the host is yeast, for example, Burkholder's minimal medium [Bostian, KL et al., Processings of the National Academy of Sciences] Natl. Acad. Sci. USA, 77, 4505 (1980)] and 0.
- the pH of the medium is preferably about 5-8. Culture is usually about 2
- the culture medium for culturing insect cells or insect transformants is, for example, Grace's Insect Medium (Grace, TCC, Nature-(Nature), 195,788 (1962)). For example, those to which additives such as% P serum are appropriately added are used.
- the pH of the medium is preferably between about 6.2 and 6.4. Culture is usually about 27, and is performed for about 3 to 5 days. Ventilation and stirring may be performed as necessary.
- Examples of a medium for culturing a transformant in which the host is an animal cell include a MEM medium containing about 5 to 20% fetal bovine serum [Science, 122, 501 (1952)], DMEM Medium [Virology, 8, 396 (1959)], RPMI 1640 medium [Journal of the American Medical 'Associe (The Journal of the American Medical Association) Volume 199 , 519 (1967)], and 199 medium (Proceeding of the Society for the Biological Medicine, 73, 1 (1950)). Used.
- the pH of the medium is preferably about 6-8.
- the cultivation is usually performed at about 30 ° C to 40 ° C for about 15 to 60 hours. Ventilation and stirring may be performed as necessary.
- ABP1 of the present invention or a partial peptide thereof or a salt thereof can be produced inside or outside the cells of the transformant.
- the ABPs of the present invention can be separated and purified from a culture obtained by culturing the transformant according to a method known per se.
- the cells or cells collected from the culture by a known method are suspended in an appropriate buffer, and then subjected to ultrasound, lysozyme and Z or freeze-thawing. After the cells or cells are destroyed by, for example, a method of obtaining a crude extract of a soluble protein by centrifugation or filtration is used as appropriate.
- the buffer may contain a protein denaturant such as urea or guanidine hydrochloride, or a surfactant such as Triton X-100 TM .
- the isolation and purification of the ABPs 1 of the present invention contained in the soluble fraction thus obtained can be carried out according to a method known per se.
- Such methods include methods using solubility such as salting out and solvent precipitation; mainly using differences in molecular weight such as dialysis, ultrafiltration, gel filtration, and SDS-polyacrylamide gel electrophoresis.
- a method utilizing a difference in charge such as ion exchange chromatography; a method utilizing a specific affinity such as affinity chromatography; a method utilizing a difference in hydrophobicity such as reverse phase high performance liquid chromatography.
- the free form can be converted to a salt by a method known per se or a method analogous thereto, and when the ABP 1 is obtained as a salt, The salt can be converted into a free form or another salt by a method known per se or a method analogous thereto.
- ABP1s produced by the transformant can be arbitrarily modified or the polypeptide can be partially removed before or after purification by the action of an appropriate protein modifying enzyme.
- the protein-modifying enzyme include trypsin, chymotrypsin, arginyl endopeptidase, protein kinase, glycosidase and the like.
- the presence of the ABPs 1 of the present invention thus obtained can be confirmed by, for example, enzymatic immunoassay western blotting using a specific antibody.
- the ABPs 1 of the present invention are characterized in that the RNA corresponding to the DNA encoding the above-mentioned ABP 1 or a partial peptide thereof is a type II, and that a heron reticulocyte lysate, a wheat germ lysate, and an Escherichia coli lysate are used.
- DNA encoding ABP1 or a partial peptide thereof can also be synthesized as type III using a cell-free transcription Z translation system containing RNA polymerase.
- an antibody against ABP 1 or a salt thereof of the present invention may be a polyclonal antibody or a monoclonal antibody as long as it can recognize ABP 1 or a salt thereof. Any of null antibodies may be used.
- ABP 1 or its An antibody against a salt can be produced by using the ABP 1s of the present invention as an antigen according to a method for producing an antibody or antiserum known per se.
- the ABP1s of the present invention used as an antigen any of the above-mentioned ABP1s or partial peptides thereof or salts thereof may be used.
- a monoclonal antibody or a polyclonal antibody against ABP1 or a salt thereof can be produced, for example, as follows.
- the ABP 1s of the present invention are administered to a mammal at a site where antibody production is possible upon administration, together with itself or a carrier or diluent.
- Complete Freund's adjuvant or incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration. Administration is usually performed once every 2 to 6 weeks, for a total of about 2 to 10 times.
- the mammal used includes, for example, monkeys, egrets, dogs, guinea pigs, mice, rats, sheep, and goats, and mice and rats are preferably used.
- an individual with an antibody titer is selected from a mammal immunized with an antigen, such as a mouse, and the spleen or lymph node is collected 2 to 5 days after the final immunization, and the antibody-producing cells contained therein are collected.
- an antigen such as a mouse
- myeloma cells of the same or different species a monoclonal antibody-producing hybridoma can be prepared.
- the measurement of the antibody titer in the antiserum can be performed, for example, by reacting the labeled protein described below with the antiserum, and then measuring the activity of the labeling agent bound to the antibody.
- the fusion operation can be performed according to a known method, for example, the method of Koehler and Milstein [Nature, 256, 495 (1975)].
- the fusion promoter include polyethylene glycol (PEG) and Sendai virus, and PEG is preferably used.
- myeloma cells include mammalian myeloma cells such as NS-1, P3U1, SP 2/0, and AP-1, and P3U1 is preferably used.
- the preferred ratio between the number of antibody-producing cells (spleen cells) and the number of myeloma cells used is 1: 1 to 20: about 1, PEG (preferably PEG 1000 to PEG 6000) is added at a concentration of about 10 to 80%, and 20 to 40 ° C, preferably 30 to 37 ° C for 1 to 10 minutes By performing the incorporation, cell fusion can be carried out efficiently.
- Monoclonal antibody-producing hybridomas are prepared, for example, by adding a hybridoma culture supernatant to a solid phase (eg, microplate) on which an antigen is directly or adsorbed together with a carrier, and then labeling with an immunoglobulin labeled with a radioactive substance or an enzyme.
- a solid phase eg, microplate
- Antibodies (anti-mouse immunoglobulin antibodies are used if the cells used for cell fusion are mice) or protein A, and the monoclonal antibody bound to the solid phase is detected; anti-immunoglobulin antibody or protein Screening is performed by adding the hybridoma culture supernatant to the solid phase to which A is adsorbed, adding a protein labeled with a radioactive substance or an enzyme, and detecting the monoclonal antibody bound to the solid phase. be able to.
- the selection of the monoclonal antibody can be performed according to a method known per se or a method analogous thereto.
- the selection of monoclonal antibodies can usually be performed in a medium for animal cells supplemented with HAT (hypoxanthine, aminopterin, thymidine).
- HAT hyperxanthine, aminopterin, thymidine
- any medium can be used as long as hybridomas can grow. Examples of such a medium include RPMI 1640 medium containing 1 to 20%, preferably 10 to 20% fetal bovine serum, and GIT medium containing 1 to 10% fetal bovine serum (Wako Pure Chemical Industries, Ltd.
- a serum-free medium for hybridoma culture (SFM-101, Nissui Pharmaceutical Co., Ltd.) can be used.
- the cultivation temperature is usually 20 to 40 ° (preferably about 37 ° C.)
- the cultivation time is generally 5 days to 3 weeks, preferably 1 to 2 weeks.
- the antibody titer of the culture supernatant of the eight hybridomas can be measured in the same manner as the measurement of the antibody titer in the antiserum described above.
- the monoclonal antibody thus obtained can be obtained by a method known per se, for example, an immunoglobulin separation and purification method [eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis, ion exchanger, (Eg, DEAE) adsorption / desorption method, ultracentrifugation method, gel filtration And a specific purification method in which only the antibody is collected using an antigen-binding solid phase or an active adsorbent such as protein A or protein G, and the bond is dissociated to obtain the antibody.
- an immunoglobulin separation and purification method eg, salting out method, alcohol precipitation method, isoelectric point precipitation method, electrophoresis, ion exchanger, (Eg, DEAE) adsorption / desorption method, ultracentrifugation method, gel filtration
- a specific purification method in which only the antibody is collected using an antigen-binding solid phase or an active adsorbent such as protein
- a polyclonal antibody against ABP1 or a salt thereof can be produced according to a method known per se.
- a immunizing antigen (ABPs 1) itself or a complex thereof with a carrier-protein is prepared, and a mammal is immunized in the same manner as in the above-described method for producing a monoclonal antibody. It can be produced by collecting the contents and separating and purifying the antibody.
- other than mammals, chickens and the like can be used.
- the type of carrier protein and the mixing ratio between carrier and hapten are determined by the antibody against hapten immunized by cross-linking with carrier. Any material may be cross-linked at any ratio as long as it can be efficiently used.For example, serum albumin, thyroglobulin, hemocyanin, etc., in a weight ratio of about 0.1 to 2 per hapten per hapten. A method of force-pulling at a rate of 0, preferably about 1 to 5 is used.
- various condensing agents for example, an active ester reagent containing a glutaldehyde aldehyde, a maleimide active ester, a thiol group or a dithiopyridyl group are used.
- the condensation product is administered to a mammal at a site capable of producing an antibody itself or together with a carrier and a diluent.
- Complete Freund's adjuvant / incomplete Freund's adjuvant may be administered in order to enhance the antibody-producing ability upon administration.
- the administration is usually made once every about 2 to 6 weeks, for a total of about 3 to 10 times.
- the polyclonal antibody can be collected from blood, ascites, etc., preferably from blood, of the mammal immunized by the above method.
- polyclonal antibody titer in the antiserum can be measured in the same manner as the measurement of the antibody titer in the antiserum described above. Separation and purification of polyclonal antibodies can be performed according to the same immunoglobulin separation and purification method as for monoclonal antibodies described above. You.
- a partial peptide of ABP1 When a partial peptide of ABP1 is used as an antigen, its position on ABP1 is not particularly limited, and examples include an oligopeptide having a partial amino acid sequence in a region well conserved among various warm-blooded animals. Specifically, a peptide having the amino acid sequence shown in SEQ ID NO: 5 or SEQ ID NO: 6 which is completely conserved between human and mouse is preferably exemplified.
- antisense polynucleotide of the present invention examples include: It has a nucleotide sequence completely complementary to or substantially complementary to the nucleotide sequence encoding ABP1, and has an effect of inhibiting the translation of the protein from RNA encoding ABP1. What is necessary is just to have.
- nucleotide sequence a nucleotide sequence encoding ABP1 or a partial peptide thereof and a nucleotide sequence capable of hybridizing under physiological conditions of a cell expressing the protein, more specifically About 70% or more, preferably about 80% or more, more preferably about 90% or more, most preferably about 90% or more with respect to the overlapping portion with the complementary strand of the nucleotide sequence encoding ABP1 or a partial peptide thereof.
- a nucleotide sequence having about 95% or more homology is exemplified.
- the antisense polynucleotide of the present invention can be designed and synthesized based on cloned or determined nucleotide sequence information of the polynucleotide of the present invention. Such a polynucleotide can inhibit the replication or expression of the gene encoding ABP1. That is, the antisense polynucleotide of the present invention can hybridize with RNA transcribed from the gene encoding ABP1, and inhibits mRNA synthesis (processing) or function (translation into protein). Can be.
- the length of the target region of the antisense polynucleotide of the present invention is not particularly limited as long as the translation of ABP1 is inhibited by the hybridization of the antisense polynucleotide, and the ABP1 encodes ABP1.
- the entire or partial sequence of the RNA may be short, about 15 bases long, long And the entire sequence of mRNA or early transcript. Considering the ease of synthesis and the problem of antigenicity, oligonucleotides consisting of about 15 to about 30 bases are preferred, but not limited thereto.
- the 5 'end hairpin loop of the gene encoding ABP 1, the 5' end 6—base pair 'repeat, the 5' end untranslated region, the polypeptide translation start codon, the protein coding region, the ORF translation start Codons, 3 'untranslated regions, 3' palindrome regions, and 3 'end hairpin loops can be selected as target regions, but any region within the gene can be selected as a target. For example, it is also preferable to use the intron portion of the gene as a target region.
- the antisense polynucleotide of the present invention not only inhibits translation into protein by hybridizing with mRNA or early transcript encoding ABP1, but also encodes ABP1, a double-stranded DNA. It may bind to a gene to form a triplex, thereby inhibiting RNA transcription.
- Antisense polynucleotides may be deoxyliponucleotides containing 2-dexoxy-D-report, liponucleotides containing D-report, N-glycosides of purine or pyrimidine bases, or other types of nucleosides.
- polymers with non-nucleotide backbones eg, commercially available protein nucleic acids or nucleic acid polymers specific to synthetic sequences
- polymers containing special bonds can be found in DNA or RNA
- Pairing of bases as found contains nucleotides with a configuration that allows base attachment
- They can be double-stranded DNA, single-stranded DNA, double-stranded RNA, single-stranded RNA, and also DNA: RNA hybrids, and can be unmodified polynucleotides (or unmodified oligonucleotides), or even known.
- intramolecular nucleotides Modified, eg, having uncharged bonds (eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.), charged or sulfur-containing bonds (eg, phosphorothioate, Such as proteins (nucleases, nucleases, etc.) Nucleic acid inhibitors, toxins, antibodies, signal peptides, poly-L-lysine, etc.) and sugars (for example, monosaccharides), etc.
- uncharged bonds eg, methylphosphonate, phosphotriester, phosphoramidate, olebamate, etc.
- sulfur-containing bonds eg, phosphorothioate
- proteins nucleases, nucleases, etc.
- sugars for example, monosaccharides
- nucleic acid may include not only those containing purine and pyrimidine bases but also those containing other modified heterocyclic bases. Such modifications can include methylated purines and pyrimidines, acylated purines and pyrimidines, or other heterocycles.
- Modified nucleotides and modified nucleotides may also be modified at the sugar moiety, e.g., where one or more hydroxyl groups have been replaced by halogens or aliphatic groups, or by functional groups such as ethers, amines, etc. It may be converted.
- Antisense polynucleotides are RNA, DNA, or modified nucleic acids (RNA, DNA).
- modified nucleic acid include, but are not limited to, sulfur derivatives of nucleic acids, thiophosphoate derivatives, and polynucleoside amides, which are resistant to degradation of oligonucleoside amides.
- the antisense polynucleotide of the present invention can be preferably designed according to the following policy.
- Antisense polynucleotides may contain altered or modified sugars, bases, or linkages, and are provided in specialized forms such as ribosomes and microspheres. Can be given, applied by gene therapy, or given in an added form. Such additional forms include polycations, such as polylysine, which act to neutralize the charge on the phosphate backbone, and lipids, which enhance the interaction with cell membranes or increase the uptake of nucleic acids ( For example, hydrophobic substances such as phospholipid and cholesterol) can be mentioned. Preferred lipids for addition include cholesterol and its derivatives (eg, cholesteryl chromate formate, cholic acid, etc.).
- Such a substance can be attached to the 3 'end or 5' end of a nucleic acid, and can be attached via a base, sugar, or intramolecular nucleoside bond.
- Other groups include cap groups specifically arranged at the 3 'end or the 5' end of nucleic acids for preventing degradation by nucleases such as exonuclease and RNase. Examples of such a capping group include, but are not limited to, hydroxyl-protecting groups known in the art, such as glycols such as polyethylene glycol and tetraethylene glycol.
- a liposome capable of specifically cleaving mRNA or gene early transcript encoding ABP 1 within the coding region is also included in the antisense polynucleotide of the present invention. May be included.
- the term "lipozyme” refers to an RNA having an enzymatic activity for cleaving a nucleic acid, but recently, it has been revealed that oligo DNA having the nucleotide sequence of the enzyme active site also has a nucleic acid cleaving activity. However, in the present specification, the term is used as a concept including DNA as long as it has a sequence-specific nucleic acid cleavage activity.
- the most versatile liposomes include self-splicing RNAs found in infectious RNAs such as viroid and willoid, and hammerhead and hairpin types are known.
- the hammerhead type exerts enzymatic activity at about 40 bases, and the bases at both ends (approximately 10 bases in total) adjacent to the hammerhead structure take the desired cleavage site of mRNA.
- By using a complementary sequence it is possible to specifically cleave only the target mRNA.
- This type of lipozyme has the additional advantage of not attacking genomic DNA, as it uses only RNA as substrate You.
- the lipozyme is used in the form of an expression vector containing the DNA encoding the liposome, a hybrid in which the tRNA-modified sequence is further ligated to promote the transfer of the transcript to the cytoplasm It can also be a lipozyme [Nucleic Acids Res., 29 (13): 2780-2788 (2001)].
- Double-stranded oligo RNA complementary to the mRNA encoding ABP 1 or a partial sequence in the coding region of the gene early transcript (including the intron part in the case of the early transcript) is also used. It can be included in the antisense polynucleotide of the present invention.
- RNAi RNA interference
- the antisense oligonucleotide and lipozyme of the present invention determine the target region of mRNA or early transcript based on the cDNA or genomic DNA sequence information encoding the protein of the present invention, and a commercially available DNAZRNA automatic synthesizer (Applied Biosystems, Beckman, etc.) to synthesize a sequence complementary thereto.
- a DNAZRNA automatic synthesizer Applied Biosystems, Beckman, etc.
- the sense strand and the antisense strand were synthesized using a DNA / RNA automatic synthesizer, respectively, and denatured in an appropriate annealing buffer, for example, at about 90 to about 95 ° C for about 1 minute. Thereafter, it can be prepared by annealing at about 30 to about 70 ° C for about 1 to about 8 hours.
- a longer double-stranded polynucleotide can be prepared by synthesizing complementary oligonucleotide chains so as to alternately overlap each other, annealing them, and ligating them with a ligase.
- ABP 1 or the activation peptide of the present invention binds and activates ASK 1 Therefore, induction of apoptosis via the ASK1 kinase cascade can be promoted. Therefore, (1) ABP1 or the activation peptide of the present invention, (2) a polynucleotide encoding ABP1 or the activation peptide of the present invention, (3) a polynucleotide containing a base sequence encoding ABP1 or a part thereof, (4) the present invention
- the antibody, the antisense polynucleotide of the present invention, and the inhibitory peptide of the present invention have the following uses.
- ABP1 has a function of promoting Apotosis induction through the ASK1 kinase cascade by binding and activating ASK1. Therefore, ABP1 or the activating peptide of the present invention or a salt thereof is added to the cells, or a polynucleotide encoding ABP1 or the activating peptide of the present invention is introduced into the cells to express them, and intracellular By increasing the amount of ABP1, the activation of ASK1 in the cells can be promoted, or apoptosis can be induced in the cells. For example, it can be used as a reagent for studying apoptosis.
- ABP 1 or the activating peptide of the present invention or a salt thereof is used as the above-mentioned ASK 1 activation promoter or apoptosis inducer
- water or an appropriate buffer eg, phosphate buffer, PBS, Tris-HCl buffer
- PBS phosphate buffer
- Tris-HCl buffer Tris-HCl buffer
- the polynucleotide when ABP1 or a polynucleotide encoding the activating peptide of the present invention is used as the above-mentioned ASK1 activation promoter or apoptosis inducer, the polynucleotide may be used alone or in a retrovirus vector or adenovirus vector. After insertion into an appropriate vector such as an adenovirus associated virus vector, the cells can be introduced into cells using the above-mentioned transformation methods (eg, ribosome method, electoporation method, etc.).
- transformation methods eg, ribosome method, electoporation method, etc.
- ABP 1 since ABP 1 has a function of activating ASK 1 to induce apoptosis in cells, ABP 1 or a nucleic acid encoding it in vivo (eg, Gene, mRNA, etc.) is abnormal or defective, or its expression level is abnormally decreased, and the induction of apoptosis in cells by some other factor.
- various diseases such as cancer, autoimmune diseases, viral infections, endocrine diseases, blood diseases, and organ hyperplasia occur.
- ABP 1 or the activating peptide of the present invention or a salt thereof may be used.
- ABP 1 or the activating peptide of the present invention or a salt thereof may be used.
- administering to the patient to supplement the amount of ABP 1, or b)
- administering ABP 1 or the DNA encoding the activating peptide of the present invention to the patient and expressing them in target cells.
- Apoptosis via ASK1 cascade can be induced in abnormal or unnecessary cells.
- ABP1 or the activation peptide of the present invention or a salt thereof or b) a polynucleotide encoding ABP1 or the activation peptide of the present invention to induce apoptosis.
- Effective diseases such as cancer (eg, leukemia, esophageal cancer, stomach cancer, colon cancer, rectal cancer, lung cancer, liver cancer, kidney cancer, breast cancer, uterine cancer, ovarian cancer, prostate cancer, melanoma, myeloma, osteosarcoma) , Brain tumors), autoimmune diseases (eg, systemic lupus erythematosus, scleroderma, rheumatoid arthritis, siedalen syndrome group, multiple sclerosis, insulin-dependent diabetes mellitus, psoriasis, ulcerative colitis, idiopathic small blood disease Platelet purpura, Crohn's disease, glomerulonephritis, etc., viral infections (eg, hemorrhagic fever, T-cell leukemia, cystic sarcoma, infectious mononucleosis, lymphoma, nasopharyngeal cancer, cervical cancer, Skin cancer, hepatitis, liver cancer, etc
- the polynucleotide encoding ABP 1 or the activating peptide of the present invention when used as the above-mentioned prophylactic / therapeutic agent, the polynucleotide may be used alone or in the form of a retrovirus vector, an adenovirus vector, an adenovirus association. After introduction into a suitable vector such as a Ted virus vector, it can be formulated according to conventional means. The polynucleotide can be administered as it is or together with an auxiliary for promoting uptake by a catheter such as a gene gun or a hydrogel catheter.
- a catheter such as a gene gun or a hydrogel catheter.
- a) ABP 1 or the activating peptide of the present invention or a salt thereof, or b) a polynucleotide encoding ABP 1 or the activating peptide of the present invention may be a sugar-coated tablet or capsule, if necessary. It can be used orally as an elixir, microcapsule or the like, or parenterally in the form of an injection such as a sterile solution with water or another pharmaceutically acceptable liquid, or a suspension.
- ABP1 or the activating peptide of the present invention or a salt thereof, or b) a polynucleotide encoding ABP1 or the activating peptide of the present invention may be prepared by using a known physiologically acceptable carrier, flavoring agent, or excipient. It can be manufactured by mixing with excipients, vehicles, preservatives, stabilizers, binders and the like in the unit dosage form generally accepted for the practice of formulations. The amount of the active ingredient in these preparations is such that an appropriate dose in the specified range can be obtained.
- Additives that can be incorporated into tablets, capsules, etc. include, for example, binders such as gelatin, corn starch, tragacanth, gum arabic, excipients such as crystalline cellulose, corn starch, gelatin, alginic acid, etc. Swelling agents such as magnesium stearate, sweeteners such as sucrose, lactose or saccharine, and flavoring agents such as peppermint, cocoa oil or cherry are used.
- the unit dosage form is a capsule, the above type of material can further contain a liquid carrier such as an oil or fat.
- Sterile compositions for injection are formulated according to standard pharmaceutical practice, such as dissolving or suspending the active substance in vehicles such as water for injection, and naturally occurring vegetable oils such as sesame oil and coconut oil.
- vehicles such as water for injection, and naturally occurring vegetable oils such as sesame oil and coconut oil.
- aqueous solution for injection for example, physiological saline, isotonic solution containing glucose and other adjuvants (eg, D-sorbitol, D-mannitol, sodium chloride, etc.) and the like are used.
- alcohols eg, ethanol
- polyalcohols eg, propylene glycol, polyethylene daricol
- non-ionic surfactants eg, Polysorbate 80 TM , HCO-50
- oily liquid for example, sesame oil, soybean oil and the like are used, and may be used in combination with solubilizers such as benzyl benzoate and benzyl alcohol.
- prophylactic / therapeutic agents examples include, for example, buffers (eg, phosphate buffer, sodium acetate buffer), soothing agents (eg, benzalkonium chloride, procaine hydrochloride, etc.), stabilizers (eg, human serum It may be combined with preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants, etc.
- buffers eg, phosphate buffer, sodium acetate buffer
- soothing agents eg, benzalkonium chloride, procaine hydrochloride, etc.
- stabilizers eg, human serum It may be combined with preservatives (eg, benzyl alcohol, phenol, etc.), antioxidants, etc.
- preservatives eg, benzyl alcohol, phenol, etc.
- the preparations obtained in this way are safe and low toxic, they can be used, for example, in humans and other warm-blooded animals (eg, rats, mice, hamsters, egrets, sheep, sheep, goats, pigs, pigs, dogs, cats). , Dogs, monkeys, chimpanzees, birds, etc.). '
- the dosage of ABP 1 or the activating peptide of the present invention varies depending on the administration subject, target organ, symptom, administration method, and the like.
- oral administration for example, a cancer patient (as 60 kg) is generally used.
- the amount is about 0.1 to 100 mg, preferably about 1.0 to 50 mg, more preferably about 1.0 to 2 Omg per day.
- the single dose varies depending on the subject, target organ, symptoms, administration method, etc., but, for example, in the case of injection, it is usually used, for example, in cancer patients (60 kg). It is convenient to administer about 0.01 to 30 mg / day, preferably about 0.1 to 20 mg / day, more preferably about 0.1 to about 0 / L / day. In the case of other animals, the dose can be administered in terms of 60 kg.
- Administration of a polynucleotide encoding ABP 1 or an activated peptide of the invention varies depending on the administration target, target organ, symptom, administration method, etc., but in the case of oral administration, for example, in a cancer patient (as 60 kg), about 0.1 mg to 100 mg per day, Preferably it is about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
- the single dose varies depending on the administration target, target organ, symptoms, administration method, etc., for example, usually in the form of injection, for example, cancer patients (60 kg) It is convenient to administer about 0.01 to 30 mg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 10 mg per day. In the case of other animals, a dose converted per 6 O kg can be administered.
- a polynucleotide containing the nucleotide sequence encoding ABP 1 or a part thereof (hereinafter, referred to as “sense polynucleotide of the present invention”) and the antisense polynucleotide of the present invention can be used as a probe to produce human or other DNA or mRNA encoding ABP 1 in warm-blooded animals (e.g., rat, mouse, eight-muster, rabbit, sheep, sheep, goat, pig, pig, pig, cat, dog, monkey, chimpanzee, bird, etc.) Since abnormalities (gene abnormalities) can be detected, for example, genetic diagnostic agents such as damage or mutation of the DNA, abnormal or reduced expression of mRNA splicing, or amplification of the DNA or excessive expression of mRNA.
- genetic diagnostic agents such as damage or mutation of the DNA, abnormal or reduced expression of mRNA splicing, or amplification of the DNA or excessive expression of mRNA.
- the polynucleotide containing a part of the nucleotide sequence encoding ABP1 is not particularly limited as long as it has a necessary length (for example, about 15 bases or more) as a probe, and encodes a partial peptide of ABP1. (It is in frame).
- the above-described genetic diagnosis using the sense or antisense polynucleotide of the present invention includes, for example, known Northern hybridization, quantitative RT-PCR, PCR-SSCP method, allele-specific PCR, PCR- It can be performed by the SSOP method, DGGE method, RNase protection method, PCR-RFLP method, and the like.
- RNA fraction extracted from the cells of the test warm-blooded animal Hybridization ⁇ Quantitative RT-PCR shows that ABP1 expression is reduced or that PCR-SSCP method is performed on RNA or genomic DNA fractions.
- a disease associated with apoptosis or suppression of inflammatory site force-in production such as cancer (eg, leukemia, esophageal cancer, stomach cancer, colorectal cancer, rectal cancer, lung cancer, liver cancer, kidney cancer, breast cancer) ) Insulin-dependent diabetes mellitus, psoriasis, ulcerative colitis, idiopathic thrombocytopenic purpura, Crohn's disease, glomerulonephritis, etc., viral infections (eg, hemorrhagic fever, T-cell leukemia, lipopositive) Tumors, infectious mononucleosis, lymphoma, nasopharyngeal cancer, cervical cancer, skin cancer, hepatitis, liver
- a disease associated with enhanced apoptosis or inflammation such as a viral infection (eg, AIDS, influenza, unknown fever) Etc.), endocrine disorders (eg, hormone deficiency, site force in deficiency, etc.), blood disorders (eg, cytopenia, renal anemia, etc.), dysplasia (eg, thyroid atrophy, cleft palate, etc.), transplant organ rejection , Graft-versus-host disease, immunodeficiency, neurodegenerative disease (eg, polyglutamine disease, Alfheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, prion disease, cerebellar degeneration, etc.), ischemic heart disease (Eg, angina pectoris, myocardial infarction, etc.), radiation damage, ultraviolet damage (eg, sunburn, etc.), toxic diseases (eg, renal tubular cell damage by heavy metals,
- a viral infection eg, AIDS, influenza, unknown fever
- endocrine disorders e
- the antibody of the present invention can specifically recognize ABP1, it can be used for detecting ABP1 in a test solution.
- the two kinds of antibodies recognize different portions of ABP1. For example, if one antibody recognizes the N-terminus of ABP 1 (eg, a portion having the amino acid sequence shown in SEQ ID NO: 5), the other antibody will recognize the C-terminus of ABP 1 (eg, And a portion having the amino acid sequence shown in SEQ ID NO: 6).
- ABP 1 can be quantified using a monoclonal antibody against ABP 1, and can be detected by tissue staining or the like.
- the antibody molecule itself may be used, or the F (ab ') 2 , Fab', or Fab fraction of the antibody molecule may be used.
- the method for quantifying ABP 1 or a salt thereof using the antibody of the present invention is not particularly limited, and may be an antibody, antigen or antibody-antigen complex corresponding to the amount of antigen (eg, ABP 1) in a test solution.
- Any measurement method may be used as long as the amount of the body is detected by chemical or physical means, and the amount is calculated from a standard curve prepared using a standard solution containing a known amount of antigen.
- nephrometry, a competitive method, an immunometric method, and a sandwich method are preferably used, but in terms of sensitivity and specificity, it is particularly preferable to use a sandwich method described later.
- a labeling agent used in a measurement method using a labeling substance for example, a radioisotope, an enzyme, a fluorescent substance, a luminescent substance and the like are used. Radioisotopes, if example embodiment, [1 2 5 I], [1 3 1 I], [3 H], and [1 4 C] used.
- the enzyme a stable enzyme having a large specific activity is preferable.
- the fluorescent substance for example, fluorescamine, fluorescein isothiosinate and the like are used.
- the luminescent substance for example, luminol, luminol derivative, reluciferin, lucigenin and the like are used.
- a biotin- (strept) avidin system can be used for binding the antibody or antigen to the labeling agent.
- the carrier include insoluble polysaccharides such as agarose, dextran, and cellulose; synthetic resins such as polystyrene, polyacrylamide, and silicon; and glass.
- a test solution is reacted with the insolubilized antibody of the present invention (primary reaction), and further reacted with another labeled antibody of the present invention (secondary reaction), followed by labeling on the insolubilized carrier.
- primary reaction the insolubilized antibody of the present invention
- secondary reaction another labeled antibody of the present invention
- the primary reaction and the secondary reaction may be performed in reverse order, may be performed simultaneously, or may be performed with a time delay.
- the labeling agent and the method of insolubilization can be in accordance with those described above.
- the antibody used for the immobilized antibody or the labeled antibody does not necessarily need to be one kind, and two or more kinds of antibodies are used for the purpose of improving measurement sensitivity and the like. A mixture may be used.
- an antibody having a different ABP1 binding site is preferably used.
- the antibody used in the secondary reaction recognizes the C-terminal of ABP 1
- the antibody used in the primary reaction is preferably the C-terminal or less.
- an antibody that recognizes the N-terminal portion is used.
- the antibody of the present invention can also be used in a measurement system other than the sandwich method, for example, a competition method, an immunometric method, or a nephrometry.
- the competition method after the antigen in the test solution and the labeled antigen are allowed to react competitively with the antibody, the unreacted labeled antigen (F) is separated from the labeled antigen (B) bound to the antibody. (BZF separation), the labeling amount of either B or F is measured, and the amount of antigen in the test solution is quantified.
- This reaction method uses a soluble antibody as an antibody, a liquid phase method in which BZF separation is performed using polyethylene glycol or a secondary antibody against the above-mentioned antibody (primary antibody), and an immobilized antibody as the primary antibody Alternatively, a solid-phase method using a soluble primary antibody and a solid-phased antibody as the secondary antibody is used.
- the antigen in the test solution and the immobilized antigen are subjected to a competitive reaction with a certain amount of labeled antibody, and then the solid phase and the liquid phase are separated. After reacting the antigen with an excess amount of the labeled antibody, the immobilized antigen is added to bind the unreacted labeled antibody to the solid phase, and then the solid phase and the liquid phase are separated. Next, the amount of the label in either phase is measured to determine the amount of the antigen in the test solution.
- nephrometry the amount of insoluble sediment resulting from the antigen-antibody reaction in a gel or in a solution is measured. Even when the amount of antigen in the test solution is small and only a small amount of sediment is obtained, laser nephrometry utilizing laser scattering is preferably used.
- ABP1 or a salt thereof can be quantified with high sensitivity by using the antibody of the present invention.
- a biological sample eg, blood, plasma, urine, biopsy, etc.
- concentration of ABP 1 or a salt thereof in the test sample is determined using the antibody of the present invention.
- apoptosis suppression-related diseases such as cancer (eg, leukemia, esophageal cancer, gastric cancer, ⁇ : bowel cancer, rectal cancer, lung cancer, liver cancer, Kidney cancer, breast cancer, uterine cancer, ovarian cancer, prostate cancer, melanoma, myeloma, osteosarcoma, brain tumor, etc., autoimmune diseases (eg, systemic lupus erythematosus, scleroderma, rheumatoid arthritis, siddaren syndrome, multiple) Sclerosis, insulin-dependent diabetes mellitus, psoriasis, ulcerative colitis, idiopathic thrombocytopenic purpura, Crohn's disease, glomerulonephritis, etc., viral infections (eg, hemorrhagic fever, T-cell leukemia, lipopositive sarcoma, transmission) Gender Disease, lymphoma
- cancer eg, leukemia, esoph
- diseases associated with enhanced apoptosis or inflammation such as viral infections (eg, AIDS, influenza, unknown fever, etc.), endocrine diseases (eg, Hormone deficiency, site force in deficiency, etc.), blood disorders Disease (eg, cytopenia, renal anemia, etc.), organ dysplasia (eg, thyroid atrophy, cleft palate, etc.), transplanted organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative disease (eg, polyglutamine disease) , Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, prion disease, cerebellar degeneration, etc.), ischemic heart disease (eg, angina, myocardial infarction, etc.), radiation damage, ultraviolet damage (eg, Toxic diseases (eg, renal tubular cell injury due to heavy metals, hepatocellular injury due to alcohol, etc.), nutritional disorders
- viral infections eg, AIDS, influenza, unknown fever, etc
- the antibody of the present invention inhibits the binding of ABP 1 to ASK 1 by specifically binding to ABP 1, and inactivates ASK1 activation and apoptosis / inflammatory cytokine production induction promoting action. (Ie, neutralize).
- the antisense polynucleotide of the present invention inhibits the expression of ABP1 and decreases the production of the protein, resulting in ASK1 activation by ABP1 and apoptotic Z inflammatory cytokine production. The induction promoting action can be reduced.
- the antibody of the present invention to cells to inactivate ABP 1, or introducing the antisense polynucleotide of the present invention into cells to reduce the amount of ABP 1 in the cells, It can inhibit ASK1 activation and suppress apoptosis and inflammatory cytokine production of the cells, and can be used, for example, as a reagent for studying apoptosis and inflammatory response.
- water or an appropriate buffer eg, phosphate buffer, PBS, Tris-HCl buffer, etc.
- an appropriate buffer eg, phosphate buffer, PBS, Tris-HCl buffer, etc.
- PBS phosphate buffer
- Tris-HCl buffer Tris-HCl buffer
- the antisense polynucleotide of the present invention is the ASK1 activation inhibitor described above.
- the polynucleotide is used alone or after being inserted into an appropriate vector such as a tedvirus vector, and then subjected to the above-mentioned transformation methods (eg, ribosome method, electro-volatilization method, etc.). It can be introduced into cells.
- ABP 1 has a function of activating ASK 1 to induce apoptosis in cells or inducing inflammation. Therefore, ABP 1 or a nucleic acid encoding it in vivo (eg, a gene, mRNA, etc.) ) Is abnormal (expression of a highly active mutant), or its expression level is abnormally increased, and abnormal induction of cell apoptosis or inflammatory cytokine production is caused by some other factor.
- a) the antibody of the present invention or b) a disease effective in preventing apoptosis or inflammation from preventing or treating apoptosis or inflammation for example, a disease caused by overexpression of ABP1, etc.
- viral infections eg, AIDS, influenza, unknown fever, etc.
- endocrine diseases eg, hormone deficiency, Blood illness (eg, cytopenia, renal anemia, etc.), dysplasia (eg, thyroid atrophy, cleft palate, etc.), transplanted organ rejection, graft-versus-host disease, immunodeficiency, God Degenerative diseases (eg, polyglutamine disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, prion disease, cerebellar degeneration), ischemic heart disease (eg, angina pectoris, myocardial infarction, etc.), Radiation damage, UV damage (eg, sunburn, etc.) Toxic disease (eg, renal tubular cell injury
- the antibody of the present invention When used as the above-mentioned prophylactic / therapeutic agent, it can be formulated in the same manner as the above-mentioned drug containing ABP1 or the activated peptide of the present invention or a salt thereof.
- the antisense polynucleotide of the present invention When used as the above-mentioned prophylactic / therapeutic agent, it may be formulated in the same manner as the above-mentioned drug containing ABP1 or a polynucleotide encoding the activating peptide of the present invention. Can be.
- the preparations obtained in this way are safe and low toxic, such as, for example, humans and other warm-blooded animals (eg, rats, mice, hamsters, egrets, sheep, sheep, goats, bushes, dogs, dogs, It can be administered to cats, dogs, monkeys, chimpanzees, and birds.
- warm-blooded animals eg, rats, mice, hamsters, egrets, sheep, sheep, goats, bushes, dogs, dogs, dogs, It can be administered to cats, dogs, monkeys, chimpanzees, and birds.
- the dose of the antibody of the present invention varies depending on the administration subject, target organ, symptom, administration method, and the like. It is about 0.1 mg to 100 mg per day, preferably about 1.0 to 50 mg, more preferably about 1.0 to 20 mg.
- the single dose varies depending on the administration target, target organ, symptoms, administration method, etc.
- it is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 Omg per day.
- the dose of the antisense polynucleotide of the present invention varies depending on the administration subject, target organ, disease state, administration method, and the like.
- parenteral administration the single dose varies depending on the administration target, target organ, disease state, administration method, etc.
- an injection it is usually used, for example, for patients with polyglutamine disease (6O). It is convenient to administer about 0.01 to 3 Omg per day, preferably about 0.1 to 2 Omg per day, more preferably about 0.1 to 1 Omg per day. In the case of other animals, the dose can be administered in terms of 6 O kg.
- 6O polyglutamine disease
- the inhibitory peptide of the present invention can function as an (antagonist) inhibitor of ABP1, that is, it can bind to ASK1, but does not activate or inactivate it.
- ABP1 an apoptosis Z inflammatory cytokine production inhibitor
- prophylactic / therapeutic agent for a disease associated with enhanced apoptosis or inflammation.
- the inhibitory peptide of the present invention When the inhibitory peptide of the present invention is used as an inhibitor of ASK1 activation and as an inhibitor of apoptosis or inflammatory cytokine production, promotion of ASK1 activation containing the above-mentioned ABP1 or the activated peptide of the present invention or a salt thereof ⁇ Reagents can be prepared in the same manner as apoptosis inducers.
- the inhibitory peptide of the present invention is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated in the same manner as the above-mentioned drug containing ABP1, the activated peptide of the present invention or a salt thereof.
- the preparations obtained in this way are safe and have low toxicity, for example, in humans and other warm-blooded animals (for example, rats, mice, hamsters, egrets, higgies, goats, bushes, dogs, dogs). , Cats, dogs, monkeys, chimpanzees, birds, etc.).
- warm-blooded animals for example, rats, mice, hamsters, egrets, higgies, goats, bushes, dogs, dogs).
- the dosage of the inhibitory peptide of the present invention depends on the administration target, target organ, symptoms, administration method, etc.
- Oral administration generally involves, for example, about 0.1 mg to 100 mg per day, preferably about 1.0 to 50 mg, and more in patients with polyglutamine disease (as 60 kg). Preferably it is about 1.0-20 mg.
- the single dose varies depending on the subject of administration, target organ, symptoms, administration method, etc.
- it is usually used, for example, in patients with polyglutamine disease (60 kg ), It is convenient to administer about 0.01 to 30 mg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 10 mg per day.
- the dose can be administered in terms of 60 kg.
- ASK1 partial peptide having no phosphorylation activity or salt thereof is an N-terminal activation control domain.
- a partial peptide of ASK1 having the activation control domain but lacking the kinase domain responsible for activating MAPKK located downstream of the cascade can function as a (antagonistic) inhibitor of ASK1. That is, since the peptide can bind to ABP1 but cannot be activated thereby, it can competitively inhibit the binding between ASK1 and ABP1 that are endogenous to cells and the activation of ASK1 thereby. Therefore, the partial peptide can be used as an ABP1 inhibitor, an apoptotic or inflammatory site force-in production inhibitor, or a prophylactic / therapeutic agent for a disease associated with enhanced apoptosis or inflammation.
- kinase domain of ASK1 for example, in the case of human ASK1, the amino acid sequence represented by amino acid numbers 678 to 936 in the amino acid sequence represented by SEQ ID NO: 1 in the sequence listing of Patent Document 1 (Japanese Patent Laid-Open No. 10-93) Is mentioned.
- a partial peptide of ASK1 having an activation control domain on the N-terminal side and lacking a kinase domain includes: Amino acid sequence identical or substantially identical to all or a part of the amino acid sequence represented by amino acid numbers 1 to 677 in the amino acid sequence of human ASK 1 (hereinafter, referred to collectively as “ASK1-N sequence”) And a peptide having the ability to bind to ABP1.
- substantially identical amino acid sequence Is preferably about 70% or more, preferably about 80% or more, more preferably about 90% or more, particularly preferably about 70% or more of all or a part of the amino acid sequence represented by amino acid numbers 1 to 677 in the amino acid sequence of the human ASK1. Is an amino acid sequence having a homology of about 95% or more, most preferably about 98% or more.
- the ASK1 partial peptide of the present invention includes, for example, 1) 1 or 2 or more in the ASK1-N sequence (preferably about 1 to 30, preferably about 1 to 10, more preferably Amino acid sequence in which the amino acid sequence has been deleted, (2) 1 or more (preferably about 1 to 30, preferably about 1 to 10, more preferably 1 to 10) in the ASKl_N sequence.
- the ASK1 partial peptide of the present invention can be prepared by the same method as the above ABP1 partial peptide.
- ASK1 activation containing the above-mentioned ABP1 or the activated peptide of the present invention or a salt thereof is used.
- Acceleration ⁇ Reagents can be prepared in the same manner as the apoptosis-inducing agent.
- the ASK1 partial peptide of the present invention is used as the above-mentioned prophylactic / therapeutic agent, it can be formulated in the same manner as the above-mentioned drug containing ABP1 or the activated peptide of the present invention or a salt thereof.
- the preparations obtained in this way are safe and low toxic, such as, for example, humans and other warm-blooded animals (eg, rats, mice, hamsters, egrets, sheep, sheep, goats, bushes, dogs, dogs, Cats, dogs, monkeys, chimpanzees, birds, etc.).
- the dose of the ASK1 partial peptide of the present invention varies depending on the administration subject, target organ, symptoms, administration method, and the like.
- oral administration in general, for example, in a polydaltamin disease patient (60 kg), About 0.1 mg to 100 mg per day, preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg.
- the single dose varies depending on the subject of administration, target organ, symptoms, administration method, etc.
- it is usually used, for example, in patients with polydaltamin disease (6 O kg ), It is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 10 mg per day.
- the equivalent dose per 60 kg can be administered.
- the present invention also provides a method of screening for an ASK1 activation-regulating substance by using ABP1 or the activating peptide of the present invention or a salt thereof, or a cell that produces it.
- the screening method includes (A) a method using the binding property of ABP1 and ASK1, (B) a method using the activation of ASK1 as an index, and (C) a method using the expression level of ABP1 as an index. It is roughly divided.
- a partial peptide thereof that retains binding to ASK1 or ABP1 that is, a partial peptide containing at least the N-terminal activation control domain
- a salt thereof or a salt thereof is produced.
- the cells used are further used.
- ABP1 can bind to and activate ASK1, PINDIN using ABP1 or the activation peptide of the present invention and ASK1 or its partial peptide containing the N-terminal activation control domain is used. Screening of compounds having the same action as ABP1 or the activating peptide of the present invention by constructing a Guassy system, screening for compounds that inhibit the action of ABP1 or the activating peptide of the present invention It can be performed. That is, the present invention provides a method for screening an ASK1 activation regulator using ABP1 or the activation peptide of the present invention and ASK1 or a partial peptide thereof containing an N-terminal activation control domain. More specifically, the present invention provides
- ASK1 or N-terminal activity ABP1 or an ASK1 or an N-terminal activation control domain thereof containing an N-terminal activation control domain, when a partial peptide containing the activation control domain is brought into contact with the ABP1 or the activated peptide of the present invention and a test substance.
- a method for screening for an ASK 1 activation-regulating substance comprising comparing the amount of binding between a partial peptide and ABP 1 or the activated peptide of the present invention,
- ABP1 or the activation peptide of the present invention is brought into contact with a cell producing ASK1 or a partial peptide thereof containing an N-terminal activation control domain; and (2) ASK1 or N Contains ASK 1 or N-terminal activation control domain when ABP 1 or the activation peptide of the present invention and a test substance are brought into contact with cells that produce the partial peptide containing the terminal activation control domain
- a method for screening for an ASK1 activation-regulating substance comprising comparing the amount of binding between the partial peptide and ABP1 or the activation peptide of the present invention, and
- ASK1 or its partial peptide containing the N-terminal activation control domain and ABP1 or the activation peptide of the present invention are produced, and when both bind, the transcription of the reporter gene is activated It is intended to provide a method for screening for an ASK1 activation modulator, which comprises comparing the expression level of the reporter gene in a cell in the presence and absence of a test substance.
- ASK1 used in the above-mentioned screening method (a) or (b) is isolated from human or other warm-blooded animal cells using a method known per se (for example, the same method as described above for ABP1). It can be purified.
- the partial peptide of ASK 1 containing the N-terminal activation control domain is not particularly limited as long as it has the amino acid sequence of the N-terminal activation control domain described above, and may be a part of the kinase domain or the C-terminal amino acid sequence. May be included.
- the partial peptide can be obtained by digesting ASK1 with an appropriate protease.
- ASK1 and its partial peptide containing the N-terminal activation control domain are genes known per se. After cloning the DNA encoding it according to engineering techniques,
- ABP1 and the activating peptide of the present invention (hereinafter sometimes simply referred to as ABP1) can be prepared according to the above-mentioned method.
- the cell that produces ASK1 is not particularly limited as long as it is a human or other warm-blooded animal cell that expresses ASK1, and examples include HeLa cells and HEK293 cells.
- Cells that produce ASK1 or its partial peptide containing the N-terminal activation control domain include the transformant prepared by the above-described genetic method. Is exemplified.
- Test substances include, for example, proteins, peptides, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, etc., and these substances are novel Or a known one.
- the binding amount is measured by, for example, Western blot analysis using a labeled anti-ABP1 antibody and anti-ASK1 antibody, labeling either ABP1 or ASK1 (for example, [ 3 H], [ 125 I], 14 C], leaving in be carried out by such [35 S] binding, such as in) to Atsusi Ya Gerushi Futoatsusi or surface plasmon resonance, (SPR).
- ABP1 or ASK1 for example, [ 3 H], [ 125 I], 14 C
- SPR surface plasmon resonance
- a compound that binds to ASK1 and inhibits the binding between ABP1 and ASK1 can be selected as an ASK1 activation modulator.
- the reaction between ABP1 and ASK1 can usually be performed at about 37 ° C for about several hours.
- ASK1 standard by suspending ASK1 or cells producing it in a buffer suitable for screening.
- the buffer may be any buffer that does not inhibit the binding between ABP1 and ASK1, such as a phosphate buffer having a pH of about 4 to 10 (preferably, a pH of about 6 to 8) or a tris-hydrochloride buffer.
- CHAPS, T Surfactants such as we en-80 TM (Kao-Atlas), digitonin, and dexcholate can also be added to the buffer.
- protease inhibitor such as PMSF, leptin, bacitracin, aprotinin, E-64 (manufactured by Protein Research Laboratories), or papstatin is added to reduce the degradation of ABP 1 and ASK 1 by proteases. You can also.
- the ABP1 and ASK1 cells are left immobilized in the incubator, that is, while the cells are grown, or using cells fixed with daltaraldehyde / paraformaldehyde. Can be combined.
- the buffer a medium / Hank's solution or the like is used.
- a fixed amount for example, about 1000 cpm to l OOOOOO c pm for 20000 Ci / mmo 1
- labeled ABP 1 for example, [ was added ABP 1) labeled with 125 1
- NBS non-specific binding
- the force (B.-NSB) is calculated by subtracting the non-specific binding amount (NSB) from the count (B 0 ) when there is no antagonistic substance, and setting it to 100%.
- a test substance having a specific binding amount (B-NSB) of, for example, 50% or less of the count (B-NSB) can be selected as an ASK1 activation modulator.
- the screening kit of the present invention contains ABP1, preferably further contains ASK1 or a cell that produces it.
- screening kit of the present invention examples include, but are not limited to, the following.
- HeLa cells or HEK293 cells were subcultured on a 12-well plate at 5 ⁇ 10 5 holes, and cultured for 2 days in 37, 5% CO 2 , 95% air.
- ABP 1 was dissolved in PBS containing 0.1% ⁇ serum albumin (manufactured by Sigma) to a concentration of 0.1 mM and stored at 1-20.
- test substance solution M After adding ⁇ 10_ 1G 5 1 a test substance solution M, 5 nM of labeled and AB P 1 5 l was added and reacted at room temperature for 1 hour. To determine the nonspecific binding A supplementary ABP 1 to 5 1 of 10- 4 M in place of the test substance.
- Cells used in the above screening method (c) include: (1) DNA binding of ASK1 or its partial peptide containing the N-terminal activation control domain to a transcription factor (eg, GAL4, VP16, etc.) DNA encoding a fusion protein with either the domain or the transcriptional activation domain, (2) DNA encoding a fusion protein between the ABPI or the activation peptide of the present invention and the other domain of the transcription factor, And (3) a cell containing a repo-one-gene under the control of a promoter activated by the transcription factor, preferably a yeast cell, a mammalian cell, and the like.
- a transcription factor eg, GAL4, VP16, etc.
- repo overnight gene examples include luciferase gene, iQ-galactosidase gene, alkaline phosphatase gene, peroxidase gene, chloramphenico-l-acetyltransferase gene, green fluorescent protein (GFP) gene and the like.
- luciferase gene examples include luciferase gene, iQ-galactosidase gene, alkaline phosphatase gene, peroxidase gene, chloramphenico-l-acetyltransferase gene, green fluorescent protein (GFP) gene and the like.
- GFP green fluorescent protein
- the above cells are cultured for several hours to about one day under culture conditions suitable for the host cell to be used, and then a cell extract or supernatant is obtained. Perform detection.
- a compound that inhibits reporter gene expression can be selected as an ASK1 activation modulator.
- the effect of the test substance on the interaction between ABP1 and ASK1 can be determined by measuring the activation of ASK1 to determine whether the substance inhibits or promotes ASK1 activation. Can be evaluated. That is, the present invention provides activation of the protein or the peptide in a cell producing ASK1 or an N-terminal activation control domain and a partial peptide thereof containing a kinase domain, by (1) ABP1 or A method for screening for an ASK1 activation modulator by measuring and comparing in the presence of the activating peptide of the present invention and (2) in the presence of ABP1 or the activating peptide of the present invention and a test substance I will provide a.
- ASK1 or N-terminal activation control domain examples include cells that endogenously produce ASK1 (eg, HeLa cells, HEK293 cells, etc.), Animal cells into which DNA encoding ASK1 or its N-terminal side activation control domain and its partial peptide containing a kinase domain have been introduced.
- ASK1 producing cells include cells that endogenously produce ASK1 (eg, HeLa cells, HEK293 cells, etc.), Animal cells into which DNA encoding ASK1 or its N-terminal side activation control domain and its partial peptide containing a kinase domain have been introduced.
- Partial peptides of ASK 1 containing an N-terminal activation control domain and a kinase domain include amino acid numbers 1 to 936 in the amino acid sequence shown in SEQ ID NO: 1 in the sequence listing of Patent Document 1 (JP-A-10-93). Contains the same or substantially the same amino acid sequence as all or part of the amino acid sequence shown (hereinafter, referred to as "ASK1-NK sequence"), and is activated by binding to ABP1 Peptides that can be used.
- substantially identical amino acid sequence refers to about 70% or more, preferably about 80% or more, more preferably about 90% or more, particularly preferably about 95% or more, Preferably, an amino acid sequence having about 98% or more homology is exemplified.
- the partial peptides include, for example, 1) 1 or 2 or more in the ASK1-NK sequence (preferably about 1 to 30, preferably about 1 to 10, more preferably number (1 to 5) Amino acid sequence in which the amino acid sequence is deleted (2) 1 or more (preferably about 1 to 30, preferably about 1 to 10, more preferably 1 to 5) in the ASK1-NK sequence 3) ASK1— 1 or 2 or more amino acid sequences (preferably about 1 to 30, preferably about 1 to 10, more preferably number (1 to 5)) 1 1 or 2 or more in the ASK1-NK sequence (preferably about 1 to 30, preferably about 1 to 10, more preferably number (1 to 5) Amino acid sequence in which one amino acid has been replaced with another amino acid, or Mutant peptides containing the combined amino acid sequence are also included.
- ABP1 or the activating peptide of the present invention may be added to cells from the outside, or may be produced by ASK1-producing cells themselves. In the latter case, ABP 1 may be produced endogenously,
- the transformant may be a genetically engineered transformant using the ASK1-producing cell as a host according to the method for expressing ABP1 described above.
- Test substances include, for example, proteins, peptides, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, etc., and these substances are novel Or a known one.
- ASK 1 activation can be an autophosphorylation of ASK 1, a kinase located downstream of the ASK1 cascade (eg, MKK4 / 7, MK3 / 6, JN, p38, etc.) or a substrate for ASK1 It can be evaluated by measuring the phosphorylation of other proteins or synthetic peptides, the rate of inducing cell death, and the like.
- a kinase located downstream of the ASK1 cascade eg, MKK4 / 7, MK3 / 6, JN, p38, etc.
- a substrate for ASK1 It can be evaluated by measuring the phosphorylation of other proteins or synthetic peptides, the rate of inducing cell death, and the like.
- ASK1-producing cells are cultured on a multiwell plate or the like. Before performing the screening, replace the medium with a fresh medium or an appropriate buffer that is not toxic to the cells in advance, and add the test substance (or ABP 1 if the cells do not produce ABP 1) etc. for a certain period of time. After incubation, cells are extracted or the supernatant is collected, and the products and phenomena generated are quantified according to each method.
- Phosphorylation of ASK1 or other proteins or peptides can be detected by Western blot analysis using antibodies specific to phosphorylated proteins (peptides), or by incorporation of [ 32 P] -labeled ATP into substrate proteins (peptides). It can be performed by a method such as detection by electrophoresis and autoradiography.
- a cell death assay can be performed using a system that can confirm the expression of ABP 1 (for example, a cell into which DNA encoding the fusion protein of ABP 1 and a fluorescent protein has been introduced, or an ABP 1 expression vector that further contains a selectable marker gene).
- ABP 1 for example, a cell into which DNA encoding the fusion protein of ABP 1 and a fluorescent protein has been introduced, or an ABP 1 expression vector that further contains a selectable marker gene.
- the percentage of dead cells in all ABP1-expressing cells using exfoliation from the plate and morphological observation (eg, membrane blebbing, fragmentation, etc.) as indicators. be able to.
- the substance when ASK1 is activated when a test substance is added to an ASK1 producing cell, the substance can be selected as an ASK1 activation promoting substance.
- the substance when ASK 1 was inactivated when the test substance was added, In this case, the substance can be selected as an ASK1 activation inhibitor.
- the present invention relates to, for example, ABP contained in (i) a) blood of a non-human mammal, b) a specific organ, c) a tissue or cell isolated from an organ, or (ii) a transformant.
- ABP1 mRNA amount or protein amount is specifically performed as follows.
- non-human mammals eg, mice, rats, rabbits, sheep, pigs, horses, cats, dogs, monkeys, etc.
- drugs eg, TNF-a, After applying IL-1, Fas, anticancer drugs, etc., or physicochemical stress (eg, UV, reactive oxygen, ischemia, etc.), and after a certain period of time, blood or specific organs (eg, brain) , Liver, kidney, etc.) or tissues or cells isolated from organs.
- ABP1 mRNA contained in the obtained cells can be quantified by, for example, extracting mRNA from cells or the like by a usual method and using a method such as RT-PCR, or It can also be quantified by Northern blot analysis known per se.
- the amount of ABP1 protein can be quantified by extracting the protein from cells or the like by an ordinary method and performing, for example, Western blot analysis using a labeled anti-ABP1 antibody.
- a transformant expressing ABP 1 or the activating peptide of the present invention is prepared according to the above-described method, and ABP 1 or the activating peptide of the present invention or the mRNA thereof contained in the transformant is prepared. It can be quantified and analyzed in the same way.
- test substance is administered, and after a lapse of a certain time after the administration (30 minutes to 3 days, preferably 1 hour to 2 days, more preferably 1 hour to 24 hours), the ABP 1 contained in the cells It can be performed by quantifying and analyzing the amount of mRNA or the amount of protein.
- test substance When the transformant is cultured according to a conventional method, the test substance is mixed into the medium, and after culturing for a certain period of time (from 1 day to 7 days, preferably from 1 day to 3 days, more preferably after 2 days) 3 days later), by quantifying and analyzing the mRNA amount or protein amount of ABP1 or the activating peptide of the present invention contained in the transformant.
- Test substances include peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, and the like. These substances may be novel substances or known substances.
- a substance that increases the expression level of ABP1 can be selected as an ASK1 activation promoting substance, and a substance that decreases the expression level of ABP1 can be selected as an ASK1 activation inhibitor.
- ASK1 activation promoting substance (hereinafter, used synonymously in the present specification))
- ASK1 activation promoter When the ASK1 activation promoter is used as an apoptosis-inducing agent, dissolve it in water or an appropriate buffer (eg, phosphate buffer, PBS, Tris-HCl buffer, etc.) to an appropriate concentration. Can be prepared. If necessary, commonly used preservatives, stabilizers, reducing agents, tonicity agents and the like may be added. As described above, since the ASK-1 activation promoting substance has the function of inducing apoptosis in cells, patients who cannot expect removal of unnecessary cells or abnormal cells by apoptosis due to a decrease in ABP1 or other factors are considered. ASK 1 activation enhancer is administered to the patient to activate ASK 1 to induce apoptosis via ASK 1 cascade in abnormal or unnecessary cells in the patient's body. can do.
- an appropriate buffer eg, phosphate buffer, PBS, Tris-HCl buffer, etc.
- inducing apoptosis of ASK1 activation promoting substance is a prophylactic and therapeutically effective disease, such as cancer (eg, leukemia, esophageal cancer, stomach cancer, colorectal cancer, rectal cancer, lung cancer, liver cancer, kidney) Cancer, breast cancer, uterine cancer, ovarian cancer, prostate cancer, melanoma, bone myeloma, osteosarcoma, brain tumor, etc., autoimmune disease (eg, systemic lupus erythematosus, scleroderma, rheumatoid arthritis, Shederdalen syndrome, multiple occurrence Sclerosis, insulin-dependent diabetes mellitus, psoriasis, ulcerative colitis, idiopathic thrombocytopenic purpura, Crohn's disease, glomerulonephritis, etc., viral infections (eg, hemorrhagic fever, T-cell leukemia, lipopositive sarcoma, Infectious mono
- the ASK1 activation inhibitor When used as the above-mentioned prophylactic / therapeutic agent, it can be formulated in the same manner as the above-mentioned drug containing ABP1 or the activating peptide of the present invention or a salt thereof.
- the preparations obtained in this way are safe and low toxic, they can be used, for example, in humans and other warm-blooded animals (eg, rats, mice, hamsters, egrets, sheep, sheep, goats, pigs, pigs, dogs, cats). , Dogs, monkeys, chimpanzees, birds, etc.).
- warm-blooded animals eg, rats, mice, hamsters, egrets, sheep, sheep, goats, pigs, pigs, dogs, cats.
- the dose of the ASK-1 activation promoting substance varies depending on the administration target, target organ, symptoms, administration method, and the like. Is about 0.1 mg to 100 mg per day, preferably about 1.0 mg 5050 mg, more preferably about 1.0-20 mg. In the case of parenteral administration, the single dose varies depending on the administration target, target organ, symptoms, administration method, etc. It is convenient to administer about 0.01 to 3 Omg per day, preferably about 0.1 to 20 mg, more preferably about 0.1 to 1 Omg per day. For other animals, the dose can be administered in terms of 60 kg.
- ASK1 activation inhibitor (hereinafter synonymous in the present specification) Can be used to inhibit signal transduction by the ASK1 cascade to suppress cell apoptosis and inflammatory cytokine production. Therefore, by adding an ASK 1 activation inhibitor to cells, apoptosis / inflammatory site force-in production of the cells can be suppressed.
- reagents for research on apoptosis, inflammatory reaction, etc. Can be used as
- an ASK1 activation inhibitor When used as an inhibitor of apoptosis or inflammatory cytokine production, make it to an appropriate concentration in water or an appropriate buffer (eg, phosphate buffer, PBS, Tris-HCl buffer). It can be prepared by dissolving in water. If necessary, commonly used preservatives, stabilizers, reducing agents, tonicity agents and the like may be added.
- an appropriate buffer eg, phosphate buffer, PBS, Tris-HCl buffer.
- ASK1 activation inhibitors have the function of suppressing cell apoptosis and the production of inflammatory cytokines, so that apoptosis causes loss of cells necessary for the living body due to an increase in ABP1, etc. If an ASK1 activation inhibitor is administered to the patient to inhibit the activation of ASK1 in patients with known or developing inflammatory diseases, Can suppress cis death and inflammatory response.
- Treatmentally effective diseases such as viral infections (eg, AIDS, influenza, unknown fever, etc.), endocrine diseases (Eg, hormone deficiency, cytokine deficiency, etc.), blood diseases (eg, cytopenia, renal anemia, etc.), organ dysplasia (eg, thyroid atrophy) Congenital, cleft palate, etc.), transplanted organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative disease (eg, polyglutamine disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, prion disease, cerebellar degeneration) Etc.), ischemic heart disease (eg, angina pectoris, myocardial infarction, etc.), radiation damage, UV damage (eg, sunburn, etc.) Toxic disease (eg, renal tubular cell damage by heavy metals, liver cell
- the ASK1 activation inhibitor When used as the above-mentioned prophylactic / therapeutic agent, it can be formulated in the same manner as the above-mentioned medicament containing ABP1 or the activating peptide of the present invention or a salt thereof.
- the preparations obtained in this way are safe and low toxic, such as, for example, humans and other warm-blooded animals (eg, rats, mice, hamsters, egrets, sheep, sheep, goats, bushes, dogs, dogs, Cats, dogs, monkeys, chimpanzees, birds, etc.).
- warm-blooded animals eg, rats, mice, hamsters, egrets, sheep, sheep, goats, bushes, dogs, dogs, Cats, dogs, monkeys, chimpanzees, birds, etc.
- the dose of the ASK1 activation inhibitor varies depending on the administration target, target organ, symptoms, administration method, and the like.
- oral administration in general, for example, in polyglutamine disease patients (60 kg) Is about 0.1 to 100 mg per day, preferably about 1.0 to 5 Omg, more preferably about 1.0 to 20 mg per day.
- parenteral administration the single dose varies depending on the administration target, target organ, symptoms, administration method, etc.
- it is usually used, for example, in patients with polyglutamine disease (6 O kg ), It is convenient to administer about 0.01 to 30 mg per day, preferably about 0.1 to 2 Omg, more preferably about 0.1 to 10 mg per day.
- the amount converted per 60 kg can be administered.
- the present invention relates to an exogenous DNA encoding ABP 1 (hereinafter, exogenous DN of the present invention).
- exogenous DN of the present invention A) or a mutant DNA thereof (may be abbreviated as the foreign mutant DNA of the present invention).
- Non-human mammal used in the present invention is not limited to,
- Non-human mammals having the exogenous DNA of the present invention or the mutant DNA thereof include unfertilized eggs, fertilized eggs, germ cells including spermatozoa and their progenitor cells, and the like.
- the calcium phosphate method, the electric pulse method, the ribofusion It can be produced by transferring the desired DNA by a method such as a coagulation method, a coagulation method, a microinjection method, a particle gun method, or a DEAE-dextran method.
- the exogenous DNA of the present invention intended for somatic cells, organs of living organisms, tissue cells, and the like can be transferred and used for cell culture, tissue culture, and the like.
- non-human mammals for example, porcupines, pigs, higgins, goats, magpies, dogs, cats, guinea pigs, hamsters, mice, rats and the like are used.
- a hybrid line a Be CS Fi line, a BDF line, a B6D2Fi line, a BALBZc line, an ICR line, etc.
- a rat eg, Wistar, SD, etc.
- mammal in the recombinant vector that can be expressed in mammals, human and the like can be mentioned in addition to the above-mentioned non-human mammals.
- the exogenous DNA of the present invention refers to ABP1 once isolated and extracted from a mammal, not DNA encoding ABP1 originally possessed by a non-human mammal. Refers to the DNA to be loaded.
- mutant DNA of the present invention DNA having a mutation (for example, mutation) in the base sequence of the original ABP1-encoding DNA, specifically, base addition, deletion, substitution with another base DNA that has been used is used, and abnormal DNA is also included.
- mutation for example, mutation
- the abnormal DNA means a DNA that expresses abnormal ABP1, and for example, a DNA that expresses abnormal ABP1 that suppresses the function of normal ABP1 is used.
- the exogenous DNA of the present invention may be derived from a mammal which is the same or different from the target animal.
- DNA construct eg, vector
- a DNA-transferred mammal that highly expresses DNA encoding ABP1 can be produced.
- ABP1 expression vectors include Escherichia coli-derived plasmids, Bacillus subtilis-derived plasmids, yeast-derived plasmids, bacteriophages such as ⁇ phage, retroviruses such as Morrovirus 21 leukemia virus, vaccinia virus and baculovirus, etc. Animal viruses and the like are used. Among them, a plasmid derived from Escherichia coli, a plasmid derived from Bacillus subtilis or a plasmid derived from yeast are preferably used.
- promoters that regulate DNA expression include: (1) Promoters of DNA derived from viruses (eg, Simian virus, cytomegalovirus, Moroni leukemia virus, JC virus, breast cancer virus, poliovirus, etc.) , (2) Promoters derived from various mammals (humans, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.), for example, albumin, insulin II, perolabrakin II, Eras Yuichi, erythropoietin, endothelin, muscle Creatine kinase, glial fibrillary acidic protein, dalyuthion S-transferase, platelet-derived growth factor / 3, keratin Kl, K10 and K14, collagen type I and II, Cyclic AMP-dependent protein kinase j8 I subunit, dysto-oral fin, tartrate-resistant alkaline phosphatase, atrial natriuretic factor, endot
- the vector preferably has a sequence that terminates transcription of the target mRNA in a DNA-transferring mammal (generally referred to as Yuichi Mineta-1).
- a DNA-transferring mammal generally referred to as Yuichi Mineta-1
- An animal-derived sequence of each DNA can be used, and preferably, Simian virus SV40 or the like is used.
- the splicing signal of each DNA, the enhancer region, a part of the intron of eukaryotic DNA, etc. are used 5 'upstream of the promoter region, between the promoter region and the translation region in order to further express the target foreign DNA. Alternatively, it can be linked to the 3 'downstream of the translation region depending on the purpose.
- the normal ABP1 translation region is derived from liver, kidney, thyroid cells, fibroblast DNA from humans or various mammals (eg, egrets, dogs, cats, guinea pigs, hamsters, rats, mice, etc.) and commercially available DNA. All or part of genomic DNA can be obtained from various genomic DNA libraries, or complementary DNA prepared by known methods from RNA derived from liver, kidney, thyroid cells, fibroblasts, etc. can be used as a raw material. I can do it.
- a translation region in which a normal ABP1 translation region obtained from the above-described cells or tissue is mutated by a point mutagenesis method can be prepared from the exogenous abnormal DNA.
- the translation region can be prepared as a DNA construct that can be expressed in a transgenic animal by a conventional DNA engineering technique in which it is ligated downstream of the above promoter and, if desired, upstream of the transcription termination site.
- Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal.
- the presence of the exogenous DNA of the present invention in the germinal cells of the transgenic animal after the DNA transfer indicates that all the progeny of the transgenic animal retain the exogenous DNA of the present invention in all of the germinal and somatic cells. Means to do.
- the progeny of this type of animal that has inherited the exogenous DNA of the present invention has the exogenous DNA of the present invention in all of its germinal and somatic cells.
- the non-human mammal to which the exogenous normal DNA of the present invention has been transferred is confirmed to stably maintain the exogenous DNA by mating, and can be subcultured as an animal having the DNA in a normal breeding environment. I can do it.
- Transfer of the exogenous DNA of the present invention at the fertilized egg cell stage is ensured to be present in excess in all germ cells and somatic cells of the target mammal.
- Excessive presence of the exogenous DNA of the present invention in the germinal cells of the produced animal after DNA transfer is a result of the excess.
- the progeny of such animals that have inherited the exogenous DNA of the present invention have an excess of the exogenous DNA of the present invention in all of their germinal and somatic cells.
- the normal DNA of the present invention is highly expressed, and the function of endogenous normal DNA is ultimately enhanced by promoting the function of endogenous normal DNA.
- the disease may develop and can be used as a model animal for the disease.
- using the normal DNA-transferred animal of the present invention it is possible to elucidate the pathological mechanism of ABP1 hyperfunction and diseases associated with ABP1, and to examine treatment methods for these diseases.
- diseases associated with hyperactivity of ABP1 for example, viral infection (eg, AIDS, Influenza, unknown fever, etc., endocrine disorders (eg, hormone deficiency, cytotoxic deficiency, etc.), blood disorders (eg, cytopenia, renal anemia, etc.), dysgenesis (eg, thyroid atrophy, cleft palate) Etc.), transplant organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative diseases (eg, polyglutamine disease, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, prion disease, cerebellar degeneration, etc.) , Ischemic heart disease (eg, angina pectoris, myocardial infarction, etc.), radiation damage, UV damage (eg, sunburn, etc.) Toxic disease (eg, renal tubular cell damage by heavy metals,
- non-human mammals having the exogenous abnormal DNA of the present invention should be subcultured in a normal breeding environment as an animal carrying the DNA after confirming that the exogenous DNA is stably maintained by mating. Can be done.
- the target foreign DNA can be incorporated into the above-mentioned plasmid and used as a raw material.
- a DNA construct with a promoter can be prepared by ordinary DNA engineering techniques. The transfer of the abnormal DNA of the present invention at the fertilized egg cell stage is ensured to be present in all germ cells and somatic cells of the target mammal. Germ cells from the animal after DNA transfer The presence of the abnormal DNA of the present invention means that the offspring of the produced animal have the foreign abnormal DNA of the present invention in all of its germ cells and somatic cells. By obtaining a homozygous animal having the introduced DNA on both homologous chromosomes and mating the male and female animals, it is possible to breed the cells so that all offspring have the DNA.
- the non-human mammal having the exogenous abnormal DNA of the present invention has a high level of expression of the abnormal DNA of the present invention.
- Refractory eg, cancer (eg, leukemia, esophageal cancer, stomach cancer, colorectal cancer, rectal cancer, lung cancer, liver cancer, kidney cancer, breast cancer, uterine cancer, ovarian cancer, prostate cancer, melanoma, myeloma, osteosarcoma) , Brain tumors, etc.
- autoimmune diseases eg, systemic lupus erythematosus, scleroderma, rheumatoid arthritis, Shederdalen syndrome, multiple sclerosis, insulin-dependent diabetes mellitus, psoriasis, ulcerative colitis, idiopathic
- Thrombocytopenic purpura Crohn's disease, glomerulonephritis, etc.
- viral infections eg, hemorrhagic fever, T-cell leukemia
- the animal with high expression of abnormal DNA of the present invention is capable of inhibiting abnormal ABP1 function (dominant negative action) by abnormal ABP1 in ABP1 inactive type refractory disease.
- the mammal to which the foreign abnormal DNA of the present invention has been transferred has abnormal ABP1 increasing symptoms, it can also be used in a therapeutic drug screening test for ABP1 function inactive refractory disease.
- the above-mentioned test was conducted. It is possible to provide an effective and rapid screening method for the disease preventive / therapeutic agent by using an inspection method and a quantitative method. Further, using the DNA transgenic animal of the present invention or the exogenous DNA expression vector of the present invention, it is possible to examine and develop a gene therapy method for ABP1-related diseases.
- ABP1 DNA-deficient non-human mammalian embryonic stem cells and ABP1 DNA-deficient non-human mammal used in the present invention are ABP1 DNA-deficient non-human mammalian embryonic stem cells and ABP1 DNA-deficient non-human mammal used in the present invention.
- a non-human mammalian embryonic stem cell in which the DNA encoding ABP 1 is inactivated [2] the DNA introduces a reporter gene (eg, an i3-galactosidase enzyme gene derived from Escherichia coli)
- a reporter gene eg, an i3-galactosidase enzyme gene derived from Escherichia coli
- the DNA is inactivated by introducing a reporter gene (eg, a / 3-galactosidase gene derived from Escherichia coli), and a promoter for a DNA encoding the reporter gene ABP1 is deleted.
- a reporter gene eg, a / 3-galactosidase gene derived from Escherichia coli
- a non-human mammalian embryonic stem cell in which the DNA encoding ABP 1 has been inactivated is an artificially mutated ABP 1 DNA possessed by the non-human mammal, which suppresses the expression ability of the DNA, Alternatively, by substantially losing the activity of ABP1 encoded by the DNA, the DNA does not substantially have the ability to express ABP1 (hereinafter sometimes referred to as the knockout DNA of the present invention).
- ES cells human mammalian embryonic stem cells
- the method of artificially mutating the DNA encoding ABP 1 can be performed, for example, by deleting part or all of the DNA sequence by genetic engineering techniques, and inserting or substituting another DNA.
- the knockout DNA of the present invention may be prepared by, for example, shifting the codon reading frame or disrupting the function of the promoter or exon by these mutations.
- non-human mammalian embryonic stem cells in which the DNA encoding ABP 1 has been inactivated include: ABP1 DNA of the target non-human mammal is isolated, and its exon portion is a drug resistance gene typified by a neomycin resistance gene or a hygromycin resistance gene, or 1acZ (] S-galactosidase gene)
- the exon function is destroyed by inserting a repo allele gene, such as cat (chloramphenicol acetyltransferase gene), or the gene is transcribed into the intron between exons.
- a DNA sequence such as a polyA addition signal
- a DNA strand having a DNA sequence constructed so as to be ruptured (hereinafter abbreviated as a targeting vector) is introduced into the chromosome of the animal by, for example, a homologous recombination method, and the obtained ES cell is subjected to AB P1 DNA DNA sequence in the vicinity region other than the ABP 1 DNA used for Southern hybridization analysis or the targeting vector and the DNA sequence on the targeting vector using the DNA sequence on or near the probe as a probe Can be obtained by analyzing the primers by a PCR method and selecting the knockout ES cells of the present invention.
- ES cells from which the DNA encoding ABP 1 is inactivated by the homologous recombination method or the like for example, those already established may be used as described above, and the known Evans and Kaufman method may be used. It may be newly established according to. For example, in the case of mouse ES cells, currently, 129 ES cells are generally used, but since the immunological background is not clear, it is an alternative pure immunological and genetic background. For example, to obtain ES cells in which C57BL / 6 BDFi mice in which the number of eggs collected by mice and C57BLZ6 has been reduced by crossing with DBAZ2 (such as those established using F of C57B LZ6 and DBA / 2) can also be used successfully.
- DBAZ2 such as those established using F of C57B LZ6 and DBA / 2
- the C57BL / 6 mouse has a background, so that ES cells obtained using this can It can be advantageously used in that the genetic background can be replaced by C57BLZ6 mice by backcrossing with C57BLZ6 mice.
- blastocysts 3.5 days after fertilization are generally used, but in addition to this, a large number of cells can be efficiently collected by collecting 8-cell embryos and culturing them up to blastocysts. Early embryos can be obtained.
- male ES cells are generally more convenient for producing a germline chimera. It is also desirable to discriminate between males and females as soon as possible in order to reduce cumbersome culture labor.
- An example of a method for determining the sex of ES cells is a method of amplifying and detecting a gene in the sex-determining region on the Y chromosome by PCR. With this method, the number of ES cells in a colony was about 1 (about 50), compared to about 10 6 cells for karyotype analysis.
- the primary selection of ES cells in the initial stage can be performed by gender discrimination, and the early selection of male cells can greatly reduce the labor required in the initial culture.
- the secondary selection can be performed, for example, by confirming the number of chromosomes by the G-banding method.
- the embryonic stem cell line obtained in this way usually has very good growth potential, but it must be carefully subcultured because it tends to lose its ability to generate individuals.
- LIF (1-1000) on a suitable feeder cell such as STO fibroblasts OU / ml
- a CO2 incubator preferably 5% CO2, 95% air or 5% oxygen, 5% CO2, 90% air
- the cells are treated with a trypsin / EDTA solution (usually 0.001-0.5% trypsin 0.1--5 mM EDTA, preferably about 0.1% trypsino, ImM EDTA) to obtain single cells.
- Such subculture is usually performed every 1 to 3 days. At this time, it is desirable to observe the cells and, if any morphologically abnormal cells are found, discard the cultured cells.
- ES cells can be transformed into various types of cells, such as parietal, visceral, and cardiac muscle, by monolayer culture up to high density or suspension culture until cell clumps are formed under appropriate conditions. It is possible to differentiate [MJ Evans and MH Kaufman, Nature, 292, 154, 1981; GR Martin Proceedings of National Academy of Sciences, USA. Proc. Natl. Acad. Sci. USA) 78, 7634, 1981; TC Doetschman et al., Journal 'Ob. Embriologi. And Xperimental' Morphology, Vol. 87, 27, 1985.
- the DNA-deficient cell of the present invention obtained by differentiating the ES cell of the present invention is useful in the cell biological examination of ABP1 in the in vivo mouth.
- the non-human mammal deficient in DNA expression of the present invention can be distinguished from a normal animal by measuring the mRNA level of the animal using a known method and indirectly comparing the expression level.
- non-human mammal the same one as described above is used.
- the non-human mammal deficient in DNA expression of the present invention may be obtained, for example, by introducing the evening getter vector prepared as described above into a mouse embryonic stem cell or a mouse egg cell, and introducing the ABP of the evening getter vector into the mouse.
- Cells in which the DNA encoding ABP 1 was knocked out were analyzed by Southern hybridization analysis using DNA sequences on or near ABP 1 DNA as a probe, or DNA sequences on a targeting vector and a targeting vector.
- chimeric embryo is injected into a non-human mammal embryo or blastocyst, and the resulting chimeric embryo is transplanted into the uterus of the pseudo-pregnant non-human mammal.
- the animals produced are chimeric animals composed of both cells with a normal ABP1 locus and cells with an artificially mutated ABP1 DNA locus.
- all the tissues are artificially mutated from a population obtained by crossing such a chimeric individual with a normal individual. It can be obtained by selecting individuals composed of cells having the added ABP1 DNA locus, for example, by judging the color. The individuals obtained in this manner are usually individuals with ABP1 heterozygous expression deficiency. Can be obtained.
- a transgenic non-human mammal having the targeting vector introduced into the chromosome can be obtained by injecting a DNA solution into the egg cell nucleus by the microinjection method. It can be obtained by selecting a gene having a mutation in the ABP1 DNA locus by homologous recombination with a gene, as compared to an non-human mammal.
- the individual in which ABP1 DNA has been knocked out can be reared in a normal breeding environment after confirming that the DNA has been knocked out in the animal obtained by mating. .
- the germline can be obtained and maintained according to a conventional method. That is, by crossing male and female animals having the inactivated DNA, the inactivated DNA is A homozygous animal having both homologous chromosomes can be obtained. The obtained homozygote animal can be efficiently obtained by rearing the mother animal in such a manner that one normal individual and a plurality of homozygotes are obtained. By mating male and female heterozygous animals, homozygous and heterozygous animals having the inactivated DNA are bred and subcultured.
- a non-human mammalian embryonic stem cell in which ABP1 DNA has been inactivated is very useful for producing a non-human mammal deficient in DNA expression of the present invention.
- non-human mammal deficient in DNA expression of the present invention lacks various biological activities that can be induced by ABP1, it can serve as a model for a disease caused by inactivation of the biological activity of ABP1. However, it is useful for investigating the causes of these diseases and examining treatment methods.
- the non-human mammal deficient in DNA expression of the present invention may be a disease caused by ABP1 DNA deficiency or damage, such as cancer (eg, leukemia, esophageal cancer, stomach cancer, colon cancer, rectum cancer, lung cancer, liver cancer, Kidney cancer, breast cancer, uterine cancer, ovarian cancer, prostate cancer, melanoma, myeloma, osteosarcoma, brain tumor, etc., autoimmune disease (eg, systemic lupus erythematosus, scleroderma, rheumatoid arthritis, siedalen syndrome, polymorphism) Sclerosis, insulin-dependent diabetes, psoriasis, ulcerative colitis, idiopathic thrombocytopenic purpura, Crohn's disease, glomerulonephritis, etc., inflammation (eg, arthritis, nephritis, etc.), viral infection (eg, hemorrhagic fever) , T-cell
- the present invention relates to a disease caused by ABP1 DNA deficiency or damage, which comprises administering a test compound to a non-human mammal deficient in expressing the DNA of the present invention and observing and measuring a change in the animal. Or a compound that has a therapeutic or preventive effect on And a method for screening for a salt.
- Examples of the non-human mammal deficient in DNA expression of the present invention used in the screening method include those described above.
- Test compounds include, for example, peptides, proteins, non-peptidic compounds, synthetic compounds, fermentation products, cell extracts, plant extracts, animal tissue extracts, and plasma.These compounds are novel compounds. Or a known compound.
- a non-human mammal deficient in expression of the DNA of the present invention is treated with a test compound, compared with a non-treated control animal, and changes in each organ, tissue, disease symptoms, etc. of the animal are used as indicators. Therapeutic and prophylactic effects of test compounds can be tested.
- test compound for example, oral administration, intravenous injection and the like are used, and it can be appropriately selected according to the symptoms of the test animal, the properties of the test compound, and the like.
- the dose of the test compound can be appropriately selected according to the administration method, the properties of the test compound, and the like.
- the symptom of the test animal is, for example, about 10% or more, preferably about 30% or more, more preferably
- the test compound can be selected as a compound having a therapeutic / preventive effect on the above-mentioned diseases.
- Compounds obtained by the screening method are compounds selected from the test compounds described above, and are used as pharmaceuticals such as safe and low toxic therapeutic and prophylactic agents for the above-mentioned diseases caused by ABP1 deficiency or damage. Can be used. Furthermore, compounds derived from the compounds obtained by the above screening can be used in the same manner.
- the compound obtained by the screening method may form a salt.
- the salt of the compound include physiologically acceptable acids (eg, inorganic acids, organic acids, etc.) and bases (eg, alkali metals, etc.). And the like, and physiologically acceptable acid addition salts are particularly preferable.
- Such salts include, for example, salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) or organic acids (eg, acetic acid, formic acid, Salts with on-acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc. are used.
- inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.
- organic acids eg, acetic acid, formic acid, Salts with on-acid, fumaric acid, maleic acid, succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.
- a drug containing the compound or a salt thereof obtained by the screening method can be produced and used in the same manner as the drug containing ABP1 or the activating peptide of the present invention.
- the present invention provides a compound which promotes or inhibits the activity of a promoter for ABP1 DNA, which comprises administering a test compound to a non-human mammal deficient in expressing DNA of the present invention, and detecting the expression of a reporter gene.
- a salt screening method Provided is a salt screening method.
- the non-human mammal deficient in expressing DNA of the present invention may be a non-human mammal deficient in expressing DNA of the present invention, wherein ABP1 DNA is introduced by introducing a repo overnight gene. Those inactivated and capable of expressing the reporter gene under the control of a promoter for ABP1 DNA are used.
- test compound examples include the same compounds as described above.
- reporter gene the same one as described above is used, and a 3-galactosidase gene (1 ac Z), a soluble alkaline phosphatase gene or a luciferase gene is suitable.
- the reporter gene is coded because the repo overnight gene is under the control of a promoter for ABP1 DNA.
- ABP 1-expressing tissue should be replaced with / BP instead of ABP 1.
- 3-galactosidase is expressed. So, for example, By staining with a reagent that serves as a substrate for jS_galactosidase, such as 5-bromo-4-chloro-1-3-indolyl_jS-galactopyranoside (X-ga1), ABP can be easily performed. It is possible to observe the expression state of one animal in vivo.
- jS_galactosidase such as 5-bromo-4-chloro-1-3-indolyl_jS-galactopyranoside (X-ga1)
- an ABP1-deficient mouse or a tissue section thereof is fixed with daltaraldehyde, washed with phosphate buffered saline (PBS), and then stained with X-ga1 at room temperature or at 37 ° C. After reacting for about 30 minutes to 1 hour in the vicinity of C, the jS-galactosidase reaction may be stopped by washing the tissue specimen with a 1 mM EDTAZPBS solution, and the coloration may be observed.
- mRNA encoding 1 ac Z may be detected according to a conventional method.
- the compound or a salt thereof obtained by using the above-mentioned screening method is a compound selected from the above-mentioned test compounds and a compound that promotes or inhibits the activity of promoter to ABP1 DNA.
- the compound obtained by the screening method may form a salt.
- the salt of the compound include physiologically acceptable acids (eg, inorganic acids) and bases (eg, organic acids). Are used, and physiologically acceptable acid addition salts are particularly preferred.
- such salts include salts with inorganic acids (eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.) and organic acids (eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid) Succinic acid, tartaric acid, citric acid, malic acid, oxalic acid, benzoic acid, methanesulfonic acid, benzenesulfonic acid, etc.).
- inorganic acids eg, hydrochloric acid, phosphoric acid, hydrobromic acid, sulfuric acid, etc.
- organic acids eg, acetic acid, formic acid, propionic acid, fumaric acid, maleic acid
- Succinic acid tartaric acid, cit
- a compound or a salt thereof that promotes promoter A activity against ABP 1 DNA can promote ABP 1 expression and promote ABP 1 function.
- prevention of a disease associated with ABP 1 dysfunction It can be used as a medicament such as a therapeutic agent.
- the compound includes, for example, cancer (eg, leukemia, esophageal cancer, stomach cancer, colon cancer, rectal cancer, lung cancer, liver cancer, kidney cancer, breast cancer, uterine cancer, ovarian cancer, prostate cancer, melanoma, Myeloma, osteosarcoma, brain tumor, etc.), autoimmune disease (eg, systemic:
- Viral infections eg, hemorrhagic fever, T-cell leukemia, Tumors, infectious mononucleosis, lymphoma, nasopharyngeal cancer, uterine face cancer, skin cancer, hepatitis, liver cancer, etc.
- endocrine diseases eg, hyperhormoneism, site hyperintensity, etc.
- blood diseases eg, Polycythemia, B-cell lymphoma, polycythemia, etc., organ hyperplasia (eg, semi-yin-yang, stagnant testicles, teratoma, nephroblastoma, polycystic kidney disease, cardiac and aortic malformations, syndactyly, etc.) It can be used as a low-toxicity and safe drug such as an agent for prevention and treatment of restenosis after angioplasty and recurrence after resection of cancer.
- a compound or a salt thereof that inhibits the activity of a promoter against ABP 1 DNA can inhibit the expression of ABP 1 and the function of ABP 1; for example, a disease associated with overexpression of ABP 1 It is useful as a drug for preventive and therapeutic agents.
- the compound may be, for example, a viral infection (eg, AIDS, influenza, unknown fever, etc.), an endocrine disease (eg, hormone deficiency, cytotoxic deficiency, etc.), a blood disease (eg, cytopenia, Renal anemia, etc.), organ dysplasia (eg, thyroid atrophy, cleft palate, etc.), transplant organ rejection, graft-versus-host disease, immunodeficiency, neurodegenerative disease (eg, polyglutamine disease, Alzheimer's disease, Parkinson's disease, muscle) Amyotrophic lateral sclerosis, prion disease, cerebellar degeneration, etc.), ischemic heart disease (eg, angina, myocardial infarction, etc.), radiation damage, UV damage (eg, sunburn, etc.) Toxic disease (eg, heavy metal) Renal tubular cell injury due to alcohol, liver cell injury due to alcohol, etc.), nutritional disorders (eg, atrophy of the thymus due to vitamin and
- a medicament containing the compound or a salt thereof obtained by the screening method can be produced and used in the same manner as the medicament containing ABP1 or the activating peptide of the present invention.
- the non-human mammal deficient in DNA expression of the present invention It is extremely useful in screening for compounds or salts thereof that promote or inhibit the activity of promoters against them, and greatly contribute to the investigation of the causes of various diseases caused by insufficient expression of ABP1 DNA or the development of therapeutic drugs. Can be.
- genes encoding various proteins are ligated downstream thereof and injected into egg cells of an animal to produce a so-called transgenic animal (gene).
- transgenic animal gene
- ABP1 has a function to specifically promote or suppress the production ability of ABP1 itself in the body. It can be used as a search system for molecular compounds.
- bases, amino acids, and the like are indicated by abbreviations based on the abbreviations by the I UPAC-I UB Commission on Biochemical Nomenclature or commonly used abbreviations in the art, and examples thereof are described below.
- the L-form is indicated unless otherwise specified.
- HONB 1-hydroxy-5-norporene-2,3-dicarpoxyimide
- FIG. 1 shows the nucleotide sequence of the amino acid coding region of human ABP1 cDNA.
- [SEQ ID NO: 3] 2 shows the nucleotide sequence of the amino acid coding region of mouse ABP1c DNA.
- the amino acid sequence of a peptide used as an antigen for producing an anti-ABP1 antibody is shown.
- ASK1-KR in which lysine (K) at the ATP-binding site (position 709) in the kinase domain of human ASK1 was replaced with arginine (R) as a bait.
- K lysine
- R arginine
- a polyclonal antibody of Egret against ABP1 was prepared. Based on the amino acid sequence information obtained from the nucleotide sequence of ABP 1 cDNA, two types of peptide antibodies with ABP 1 specific peptides (SEQ ID NO: 5 and SEQ ID NO: 6) as antigens were prepared, each of which was an ELA antibody, LVR It was named Fold. Both antibodies were purified using the affinity of the antigen peptide.
- HeLa cells and HEK293 cells high glucose Modified Eagle Medium Dulbecco containing (4. 5mg / m 1); a (DMEM SIGMA) and culture was incubated in the presence of 5% C_ ⁇ 2.
- the culture medium was supplemented with 10% fetal calf serum (FBS) and 100 units of Zml lnicillin.
- FBS fetal calf serum
- PEE bush aortic endothelial
- Each cell was treated with a lysis buffer [1 50 mM NaCl, 2 OmM Tris—HC1 (pH 7.5), 1 OmM EDTA, 1% Triton X-100, 1% dexcholate, 1.5% aprotinin, ImM PMS F]
- the lysate is centrifuged, and the supernatant is taken.
- Add SDS sample buffer [lO OmM Tris-HC1 (pH8.8), 0.01% bromophenol blue, 36% glycerol, 4% SDS].
- SDS polyacrylamide gel electrophoresis SDS-PAGE
- Two fusion proteins, F1ag-ABP1 and Myc_ASK1 were co-expressed in HEK293 cells.
- the cells are lysed using a lysis buffer (described above), the lysate is centrifuged, and the supernatant is collected. After reaction with an anti-F1ag antibody (Clone M2; SIGMA), Protein A-sepharose4B (Zyied Laboratories) is added. Incubate for 30 minutes, wash three times with lysis buffer, add SDS sample buffer (described above), and confirm binding of both proteins by the same procedure as in the immunoblotting method. ( Figure 4A).
- Antibodies used were an anti-F1ag antibody (described above) and an anti-Myc antibody (Clone 9E10, Calbiochem). This binding was more enhanced H 2 0 2 treatment an activator of ASK 1.
- Antibodies used were an anti-F1ag antibody (described above) and an anti-GFP antibody (Medical & Biological Laboratories CO.). The binding was also enhanced with H 2 ⁇ 2 process. Therefore, ABP 1 and ASK1 engages binding even in mammalian cells, the binding was found to be enhanced in H 2 0 2 treatment.
- Example 5 Identification of ASK 1 binding site for ABP 1
- Binding of various ASK1 deficient mutants with HA tag shown in Fig. 5A to ABP1 was co-expressed with F1ag-ABP1, as in Example 3, and then immunoprecipitated with anti-F1ag antibody.
- Antibodies used were an anti-HA antibody (Clone 3F10, Roche Diagnostics K.K) and an anti-F1ag antibody (Clone M2, SIGMA).
- the expression vector for AS Kl used was that described in Saitoh et al. (Supra).
- ABP 1 co-precipitated with ASK 1-NT, ASK 1- ⁇ (3, but did not show co-precipitation with ASK 1-AN and ASK 1-K (FIG. 5B).
- P1 was found to bind to the N-terminal domain of ASK 1.
- Example 6 Induction of Cell Death by AB P1
- ABP 1 was considered to be a protein having cell death inducing ability.
- a cell line 0PAE-ABP1 cell capable of inducing ABP1 expression in PAE cells in a tetracycline-dependent manner was established.
- the production of PAE- ⁇ cells was performed by partially modifying the method of Takeda et al. (J. Biol. Chem., 275, 9805-9913, 2000).
- the entire length of the Myc-tagged ⁇ ⁇ 1 cDNA was subcloned into the pTet-Splice-neo vector, and this and the pTet-tTAk-hyg plasmid were simultaneously transfected into PAE cells, unlike the method of Takeda et al.
- the cells were cultured in a culture medium supplemented with 500 ng / m 1 tetracycline (Sigma), 400 units Zm 1 hygromycin B (Wako), and 240 mg / m 1 neomycin (Geneticin, Life Technologies, Inc.). Cells that survived and formed colonies were designated as PAE-ABP1 cells.
- the cells were cultured for maintenance by adding 500 ng / m 1 tetracycline, 200 units 71111 hygromycin B, and 3 Omg / m 1 neomycin to the culture solution for maintaining the PAE cells described above.
- PAE-ABP1 cells do not express ABP1 when cultured in the presence of tetracycline, but in the absence of tetracycline, expression of Myc-tagged ABP1 protein by immunoblot analysis after about 6 hours. ABP1 expression can be confirmed stably thereafter. Immunostaining with anti-Myc antibody (Clone 9E10, Calbiochem) showed stable expression of Myc—ABP1 in most cells. I did. When these cells were cultured in the absence of tetracycline, cell death was induced with the expression of ABP1 protein as in the case where ABP1 protein was transiently expressed in HeLa cells (FIG. 7A).
- the percentage of dead cells in all cells was calculated. The results are shown as the average of three visual fields) was determined. The time reached about 55% (Fig. 7B). Also in this experimental system, it was confirmed that ABP1 induced cell death.
- the DNA was purified by phenol-chloroform extraction and ethanol precipitation. After redissolving in (A), the mixture was electrophoresed on a 2% agarose gel, stained with ethidium bromide, and the migration pattern was photographed.
- the PAE-ABP1 cells formed a DNA ladder with the expression of ABP1 protein at the same time as the appearance of cell death when observed under a microscope (FIG. 8). This indicated that cell death due to ABP 1 was so-called apoptosis accompanied by DNA fragmentation. This was also confirmed by TUNEL staining of PAE-ABP1 cells.
- DNA fragmentation during apoptosis usually follows caspase activation. Therefore, caspase-3 activity was measured during induction of ABP1 protein expression using PAE-ABP1 cells.
- the measurement was performed using the synthetic fluorescent peptide DEVD-7-amino-4-trifluororaethyl coumarine (AFC) as a substrate using a CPP32 / caspase-3 fluorometric protease assay kit (MBL).
- the released AFC was measured for fluorescence intensity at an excitation wavelength of 360 nm and an emission wavelength of 530 nm using a fluorescence spectrophotometer. The degree was measured. Each sample was measured twice. The results are shown as relative values, where 1 is the value when tetracycline is not removed.
- ABP 1 does not have a functional domain of an existing apoptosis-related factor. Therefore, in order to examine the domains required for ABP 1-induced apoptosis, plasmids expressing fusion proteins of various ABP1-deficient mutants and CFP shown in Fig. 10A were constructed, and HeLa was used in the same manner as in Example 6 (1). It was expressed in cells and examined for cell death.
- ABP1 expression vectors were prepared by subcloning ABP1 cDNA or ABP1-deficient mutant cDNA A obtained by PCR into pECFP-CI (Clontech). Gene transfer of each plasmid into cells was carried out using FuGENE6 (Roche Diagnostics I. II) as described in the instruction manual.
- the ABP1 of the present invention has an action of activating the ASK1 cascade to induce apoptosis in cells or inducing the production of inflammatory site force. Therefore, the ABP1, the polynucleotide encoding the same, and the like of the present invention are useful as a prophylactic / therapeutic agent for diseases that can be expected to have a preventive / therapeutic effect by inducing apoptosis in cells.
- the ABP 1 inhibitor of the present invention for example, anti-ABP 1 antibody, ABP 1 antisense polynucleotide, etc.
- suppresses apoptosis and production of inflammatory cytokines suppresses apoptosis and production of inflammatory cytokines. It is useful as a prophylactic / therapeutic agent for promising diseases or inflammatory diseases.
- the use of ABP1 and ASK1 of the present invention provides a means for screening a novel preventive / therapeutic agent for apocysis or inflammation-related disease.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Diabetes (AREA)
- Hematology (AREA)
- Rheumatology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Gastroenterology & Hepatology (AREA)
- Zoology (AREA)
- Toxicology (AREA)
- Physical Education & Sports Medicine (AREA)
- Heart & Thoracic Surgery (AREA)
- Oncology (AREA)
- Obesity (AREA)
- Orthopedic Medicine & Surgery (AREA)
- Neurosurgery (AREA)
- Neurology (AREA)
- Biomedical Technology (AREA)
- Pain & Pain Management (AREA)
- Cardiology (AREA)
- Virology (AREA)
- Transplantation (AREA)
- Communicable Diseases (AREA)
Abstract
Description
Claims
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU2003284597A AU2003284597A1 (en) | 2002-11-22 | 2003-11-20 | Apoptosis-associated protein and use thereof |
EP03774087A EP1582586A4 (en) | 2002-11-22 | 2003-11-20 | APOPTOSIS ASSOCIATED PROTEIN AND ITS USE |
US10/535,931 US7390625B2 (en) | 2002-11-22 | 2003-11-20 | Apoptosis-associated protein and use thereof |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2002-340077 | 2002-11-22 | ||
JP2002340077 | 2002-11-22 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2004048565A1 true WO2004048565A1 (ja) | 2004-06-10 |
WO2004048565A9 WO2004048565A9 (ja) | 2005-06-16 |
Family
ID=32375809
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2003/014794 WO2004048565A1 (ja) | 2002-11-22 | 2003-11-20 | アポトーシス関連蛋白質およびその用途 |
Country Status (4)
Country | Link |
---|---|
US (1) | US7390625B2 (ja) |
EP (1) | EP1582586A4 (ja) |
AU (1) | AU2003284597A1 (ja) |
WO (1) | WO2004048565A1 (ja) |
Cited By (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012080735A1 (en) | 2010-12-16 | 2012-06-21 | Convergence Pharmaceuticals Limited | Ask1 inhibiting pyrrolopyrimidine derivatives |
WO2019012284A1 (en) | 2017-07-12 | 2019-01-17 | Galapagos Nv | PYRROLOPYRIMIDINE AND ASK1 INHIBITING PYRROLOPYRIDINE DERIVATIVES |
Families Citing this family (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2058309A4 (en) * | 2006-08-04 | 2010-12-22 | Takeda Pharmaceutical | CONDENSED HETEROCYCLIC COMPOUND |
US8892184B2 (en) | 2010-10-18 | 2014-11-18 | Siemens Medical Solutions Usa, Inc. | Systems and methods for reducing interference in a dual modality imaging system |
US20130236441A1 (en) * | 2010-11-18 | 2013-09-12 | University Of Delaware | Methods of treating and preventing thrombotic diseases using ask1 inhibitors |
US9327009B2 (en) * | 2012-03-02 | 2016-05-03 | Council Of Scientific And Industrial Research | Peptide inhibitors as novel anti-HIV therapeutics |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1997040143A1 (fr) * | 1996-04-19 | 1997-10-30 | Japanese Foundation For Cancer Research | Proteine inductrice de l'apoptose et gene codant pour elle |
WO1999057144A2 (en) * | 1998-05-05 | 1999-11-11 | Incyte Pharmaceuticals, Inc. | Human transcriptional regulator molecules |
EP1130094A2 (en) * | 1999-07-08 | 2001-09-05 | Helix Research Institute | Primers for synthesizing full length cDNA clones and their use |
WO2002018632A2 (de) * | 2000-09-01 | 2002-03-07 | Epigenomics Ag | Verfahren zur bestimmung des methylierungsgrades von bestimmten cytosinen in genomischer dna im sequenzkontext 5'-cpg-3' |
WO2002038179A1 (fr) * | 2000-11-10 | 2002-05-16 | Kissei Pharmaceutical Co., Ltd. | Preventifs ou remedes de maladies du reticulum endoblastique liees au stress |
Family Cites Families (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2003063905A1 (fr) | 2002-01-31 | 2003-08-07 | Center For Advanced Science And Technology Incubation, Ltd. | Medicaments preventifs ou remedes pour maladies immunologiques |
US7485414B2 (en) * | 2002-08-30 | 2009-02-03 | Rigel Pharmaceuticals, Inc. | Modulators of angiogenesis |
-
2003
- 2003-11-20 EP EP03774087A patent/EP1582586A4/en not_active Withdrawn
- 2003-11-20 US US10/535,931 patent/US7390625B2/en not_active Expired - Fee Related
- 2003-11-20 WO PCT/JP2003/014794 patent/WO2004048565A1/ja not_active Application Discontinuation
- 2003-11-20 AU AU2003284597A patent/AU2003284597A1/en not_active Abandoned
Patent Citations (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1997040143A1 (fr) * | 1996-04-19 | 1997-10-30 | Japanese Foundation For Cancer Research | Proteine inductrice de l'apoptose et gene codant pour elle |
JPH1093A (ja) | 1996-04-19 | 1998-01-06 | Japan Found Cancer Res | アポトーシス誘導タンパク質およびそれをコードする遺伝子 |
WO1999057144A2 (en) * | 1998-05-05 | 1999-11-11 | Incyte Pharmaceuticals, Inc. | Human transcriptional regulator molecules |
EP1130094A2 (en) * | 1999-07-08 | 2001-09-05 | Helix Research Institute | Primers for synthesizing full length cDNA clones and their use |
WO2002018632A2 (de) * | 2000-09-01 | 2002-03-07 | Epigenomics Ag | Verfahren zur bestimmung des methylierungsgrades von bestimmten cytosinen in genomischer dna im sequenzkontext 5'-cpg-3' |
WO2002038179A1 (fr) * | 2000-11-10 | 2002-05-16 | Kissei Pharmaceutical Co., Ltd. | Preventifs ou remedes de maladies du reticulum endoblastique liees au stress |
Non-Patent Citations (19)
Title |
---|
"Antisense Research and " Applications", 1993, CRC PRESS |
DATABASE GENSEQ [online] Database accession no. AK015523 * |
DATABASE GENSEQ [online] Database accession no. AK087716 * |
GYURIS ET AL., CELL, vol. 75, 1993, pages 791 - 803 |
HARUAKI YAJIMA: "Peptide Synthesis", vol. 14, HIROKAWA SHOTEN |
HARUAKI YAJIMA; SHUNPEI SAKAKIBARA; SEIKAGAKU JIKKEN, KOZA 1, TANPAKUSHITSU NO KAGAKU IV, vol. 205, 1977 |
J. KAWAKAMI ET AL., PHARM TECH JAPAN, vol. 8, 1992, pages 247 |
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495 |
LEUNG ET AL., J. BIOL. CHEM., vol. 276, no. 42, 2001, pages 39171 - 39178 |
NOBUO IZUMIYA ET AL.: "Peptide Gosei-no-Kiso to Jikken", 1975, MARUZEN CO. |
NUCLEIC ACIDS RES., vol. 29, no. 13, 2001, pages 2780 - 2788 |
PHARM TECH JAPAN, vol. 8, 1992, pages 395 |
PROC. NATL. ACAD. SCI. USA, vol. 98, no. 10, 2001, pages 5572 - 5577 |
SCHROEDER; LUEBKE: "The Peptide", 1965, ACADEMIC PRESS |
See also references of EP1582586A4 |
THE FANTOM CONSORTIUM AND THE RIKEN GENOME EXPLORATION RESEARCH GROUP PHASE I & II TEAM: "ANALYSIS OF THE MOUSE TRANSCRIPTOME BASED ON FUNCTIONAL ANNOTATION OF 60,770 FULL-LENGTH CDNAS", NATURE, vol. 420, 2002, pages 563 - 573, XP002252038 * |
THE RIKEN GENOME EXPLORATION RESEARCH GROUP PHASE II TEAM AND THE FANTOM CONSORTIUM: "FUNCTIONAL ANNOTATION OF FULL-LENGTH MOUSE CDNA COLLECTION", NATURE, vol. 409, 2001, pages 685 - 690, XP002176449 * |
TOBIUME ET AL., EMBO REP., vol. 2, 2001, pages 222 - 228 |
ZERVOS ET AL., CELL, vol. 72, 1993, pages 223 - 232 |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012080735A1 (en) | 2010-12-16 | 2012-06-21 | Convergence Pharmaceuticals Limited | Ask1 inhibiting pyrrolopyrimidine derivatives |
US9045485B2 (en) | 2010-12-16 | 2015-06-02 | Convergence Pharmaceuticals Limited | ASK 1 inhibiting pyrrolopyrimidine derivatives |
US9585886B2 (en) | 2010-12-16 | 2017-03-07 | Calchan Limited | ASK1 inhibiting pyrrolopyrimidine derivatives |
WO2019012284A1 (en) | 2017-07-12 | 2019-01-17 | Galapagos Nv | PYRROLOPYRIMIDINE AND ASK1 INHIBITING PYRROLOPYRIDINE DERIVATIVES |
US11136325B2 (en) | 2017-07-12 | 2021-10-05 | Galapagos Nv | Pyrrolopyrimidine and pyrrolopyridine derivatives |
Also Published As
Publication number | Publication date |
---|---|
EP1582586A4 (en) | 2007-03-14 |
WO2004048565A9 (ja) | 2005-06-16 |
AU2003284597A1 (en) | 2004-06-18 |
EP1582586A1 (en) | 2005-10-05 |
US7390625B2 (en) | 2008-06-24 |
US20060240023A1 (en) | 2006-10-26 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2005067971A1 (ja) | 動脈硬化の予防・治療用医薬 | |
WO2002053738A1 (fr) | Nouvelles proteines et adn correspondant | |
WO2003106683A1 (ja) | 新規スクリーニング方法 | |
WO2004048565A1 (ja) | アポトーシス関連蛋白質およびその用途 | |
WO2001000799A1 (fr) | Nouvelle proteine et son adn | |
JP4488720B2 (ja) | アポトーシス関連蛋白質およびその用途 | |
WO2003091434A1 (fr) | Nouvelle proteine, et adn de cette proteine | |
WO2004028558A1 (ja) | 神経変性疾患の予防・治療剤 | |
JP4320151B2 (ja) | 新規ポリペプチドおよびその用途 | |
JP2004101509A (ja) | 新規スクリーニング方法 | |
JP4300008B2 (ja) | 新規タンパク質およびそのdna | |
JP2004075569A (ja) | Il20受容体およびil20の新規用途 | |
JP2001299364A (ja) | 新規タンパク質およびそのdna | |
WO2004078208A1 (ja) | MIP-3α抑制薬の医薬用途および脳・神経細胞保護剤のスクリーニング方法 | |
JP2001299363A (ja) | 新規タンパク質およびそのdna | |
JP2001228146A (ja) | 疾患関連遺伝子の用途 | |
JP2004026692A (ja) | Mepeの新規用途 | |
JP2002348299A (ja) | Irap結合タンパク質 | |
WO2004081039A1 (ja) | 新規タンパク質およびそのdna | |
WO2002016428A1 (fr) | Proteine de liaison a l'irap | |
JP2000053700A (ja) | 新規タンパク質およびそのdna | |
JP2004173677A (ja) | 新規タンパク質およびそのdna | |
JPH10108683A (ja) | 新規タンパク質およびそのdna | |
JP2004261172A (ja) | 疾患関連遺伝子の用途 | |
JP2004121245A (ja) | 新規タンパク質およびそのdna |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A1 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG |
|
DFPE | Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101) | ||
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
WWE | Wipo information: entry into national phase |
Ref document number: 2003774087 Country of ref document: EP |
|
COP | Corrected version of pamphlet |
Free format text: PAGE 9/10, DRAWINGS, ADDED |
|
WWP | Wipo information: published in national office |
Ref document number: 2003774087 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2006240023 Country of ref document: US Ref document number: 10535931 Country of ref document: US |
|
NENP | Non-entry into the national phase |
Ref country code: JP |
|
WWW | Wipo information: withdrawn in national office |
Country of ref document: JP |
|
WWP | Wipo information: published in national office |
Ref document number: 10535931 Country of ref document: US |