WO2002086131A1 - Unite de regulation de l'expression genetique et utilisation de cette unite - Google Patents
Unite de regulation de l'expression genetique et utilisation de cette unite Download PDFInfo
- Publication number
- WO2002086131A1 WO2002086131A1 PCT/JP2002/003537 JP0203537W WO02086131A1 WO 2002086131 A1 WO2002086131 A1 WO 2002086131A1 JP 0203537 W JP0203537 W JP 0203537W WO 02086131 A1 WO02086131 A1 WO 02086131A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- dna
- gene
- expression
- specific
- Prior art date
Links
- 230000014509 gene expression Effects 0.000 title claims abstract description 184
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 288
- 102000004169 proteins and genes Human genes 0.000 claims abstract description 142
- 210000004027 cell Anatomy 0.000 claims abstract description 94
- 210000002569 neuron Anatomy 0.000 claims abstract description 89
- 230000005540 biological transmission Effects 0.000 claims abstract description 73
- 230000000694 effects Effects 0.000 claims abstract description 31
- 230000006870 function Effects 0.000 claims description 93
- 238000000034 method Methods 0.000 claims description 87
- 230000001537 neural effect Effects 0.000 claims description 61
- 230000001276 controlling effect Effects 0.000 claims description 58
- 101710138657 Neurotoxin Proteins 0.000 claims description 56
- 238000004458 analytical method Methods 0.000 claims description 44
- 241001465754 Metazoa Species 0.000 claims description 33
- 235000013601 eggs Nutrition 0.000 claims description 26
- 230000001105 regulatory effect Effects 0.000 claims description 22
- 239000002581 neurotoxin Substances 0.000 claims description 20
- 231100000618 neurotoxin Toxicity 0.000 claims description 20
- 239000000126 substance Substances 0.000 claims description 20
- 230000008859 change Effects 0.000 claims description 18
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 claims description 18
- 239000002858 neurotransmitter agent Substances 0.000 claims description 16
- 230000003957 neurotransmitter release Effects 0.000 claims description 15
- 210000001671 embryonic stem cell Anatomy 0.000 claims description 13
- 210000001178 neural stem cell Anatomy 0.000 claims description 11
- 230000003213 activating effect Effects 0.000 claims description 10
- 230000008827 biological function Effects 0.000 claims description 8
- 241000283984 Rodentia Species 0.000 claims description 5
- 210000003061 neural cell Anatomy 0.000 claims description 3
- 210000004102 animal cell Anatomy 0.000 claims description 2
- 230000004720 fertilization Effects 0.000 claims 1
- 210000004556 brain Anatomy 0.000 abstract description 14
- 230000030833 cell death Effects 0.000 abstract description 4
- 230000002427 irreversible effect Effects 0.000 abstract description 4
- 210000005036 nerve Anatomy 0.000 abstract description 2
- 108020004414 DNA Proteins 0.000 description 195
- 239000012634 fragment Substances 0.000 description 82
- 239000013598 vector Substances 0.000 description 78
- 241000699670 Mus sp. Species 0.000 description 65
- 241000699666 Mus <mouse, genus> Species 0.000 description 63
- 229960003692 gamma aminobutyric acid Drugs 0.000 description 53
- BTCSSZJGUNDROE-UHFFFAOYSA-N gamma-aminobutyric acid Chemical compound NCCCC(O)=O BTCSSZJGUNDROE-UHFFFAOYSA-N 0.000 description 53
- 238000011830 transgenic mouse model Methods 0.000 description 50
- 108091008146 restriction endonucleases Proteins 0.000 description 39
- SGKRLCUYIXIAHR-AKNGSSGZSA-N (4s,4ar,5s,5ar,6r,12ar)-4-(dimethylamino)-1,5,10,11,12a-pentahydroxy-6-methyl-3,12-dioxo-4a,5,5a,6-tetrahydro-4h-tetracene-2-carboxamide Chemical compound C1=CC=C2[C@H](C)[C@@H]([C@H](O)[C@@H]3[C@](C(O)=C(C(N)=O)C(=O)[C@H]3N(C)C)(O)C3=O)C3=C(O)C2=C1O SGKRLCUYIXIAHR-AKNGSSGZSA-N 0.000 description 38
- 229960003722 doxycycline Drugs 0.000 description 38
- 241000699660 Mus musculus Species 0.000 description 35
- 239000004098 Tetracycline Substances 0.000 description 35
- 229960002180 tetracycline Drugs 0.000 description 35
- 229930101283 tetracycline Natural products 0.000 description 35
- 235000019364 tetracycline Nutrition 0.000 description 35
- 150000003522 tetracyclines Chemical class 0.000 description 35
- 239000000523 sample Substances 0.000 description 32
- 239000000243 solution Substances 0.000 description 27
- 238000009396 hybridization Methods 0.000 description 25
- 108020004684 Internal Ribosome Entry Sites Proteins 0.000 description 22
- 238000012360 testing method Methods 0.000 description 20
- 210000001638 cerebellum Anatomy 0.000 description 19
- 230000002441 reversible effect Effects 0.000 description 19
- 210000005013 brain tissue Anatomy 0.000 description 18
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 17
- 108020004999 messenger RNA Proteins 0.000 description 17
- 108700012359 toxins Proteins 0.000 description 17
- 238000011144 upstream manufacturing Methods 0.000 description 17
- 238000006243 chemical reaction Methods 0.000 description 16
- 239000002953 phosphate buffered saline Substances 0.000 description 15
- 239000012528 membrane Substances 0.000 description 14
- 239000007983 Tris buffer Substances 0.000 description 13
- 210000000782 cerebellar granule cell Anatomy 0.000 description 13
- 239000000758 substrate Substances 0.000 description 13
- 238000013519 translation Methods 0.000 description 13
- 230000014621 translational initiation Effects 0.000 description 13
- 108700019146 Transgenes Proteins 0.000 description 12
- 108020004707 nucleic acids Proteins 0.000 description 12
- 102000039446 nucleic acids Human genes 0.000 description 12
- 150000007523 nucleic acids Chemical class 0.000 description 12
- 239000013612 plasmid Substances 0.000 description 12
- 210000001519 tissue Anatomy 0.000 description 12
- 238000012546 transfer Methods 0.000 description 12
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 11
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 11
- 239000013604 expression vector Substances 0.000 description 11
- 210000004565 granule cell Anatomy 0.000 description 11
- 239000002773 nucleotide Substances 0.000 description 11
- 125000003729 nucleotide group Chemical group 0.000 description 11
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 11
- 238000002105 Southern blotting Methods 0.000 description 10
- 238000010276 construction Methods 0.000 description 10
- 210000004748 cultured cell Anatomy 0.000 description 10
- 238000001514 detection method Methods 0.000 description 10
- 230000006698 induction Effects 0.000 description 10
- 210000000449 purkinje cell Anatomy 0.000 description 10
- 238000000636 Northern blotting Methods 0.000 description 9
- 108091005804 Peptidases Proteins 0.000 description 9
- 108010076504 Protein Sorting Signals Proteins 0.000 description 9
- 108010090618 Syntaxin 1 Proteins 0.000 description 9
- 150000001413 amino acids Chemical class 0.000 description 9
- 238000007901 in situ hybridization Methods 0.000 description 9
- 238000002360 preparation method Methods 0.000 description 9
- 239000003053 toxin Substances 0.000 description 9
- 231100000765 toxin Toxicity 0.000 description 9
- 238000001262 western blot Methods 0.000 description 9
- 239000004365 Protease Substances 0.000 description 8
- 102100037486 Reverse transcriptase/ribonuclease H Human genes 0.000 description 8
- 239000000872 buffer Substances 0.000 description 8
- 239000002299 complementary DNA Substances 0.000 description 8
- 238000010586 diagram Methods 0.000 description 8
- 239000002052 molecular layer Substances 0.000 description 8
- 235000019419 proteases Nutrition 0.000 description 8
- 241000283707 Capra Species 0.000 description 7
- 108091034117 Oligonucleotide Proteins 0.000 description 7
- 206010043376 Tetanus Diseases 0.000 description 7
- 238000001962 electrophoresis Methods 0.000 description 7
- 102000034356 gene-regulatory proteins Human genes 0.000 description 7
- 108091006104 gene-regulatory proteins Proteins 0.000 description 7
- 210000000653 nervous system Anatomy 0.000 description 7
- 230000001575 pathological effect Effects 0.000 description 7
- 238000013518 transcription Methods 0.000 description 7
- 230000035897 transcription Effects 0.000 description 7
- 230000009261 transgenic effect Effects 0.000 description 7
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 6
- 102000002812 Heat-Shock Proteins Human genes 0.000 description 6
- 108010004889 Heat-Shock Proteins Proteins 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- PXIPVTKHYLBLMZ-UHFFFAOYSA-N Sodium azide Chemical compound [Na+].[N-]=[N+]=[N-] PXIPVTKHYLBLMZ-UHFFFAOYSA-N 0.000 description 6
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 6
- 210000003050 axon Anatomy 0.000 description 6
- 239000003153 chemical reaction reagent Substances 0.000 description 6
- 238000003776 cleavage reaction Methods 0.000 description 6
- 239000003814 drug Substances 0.000 description 6
- 239000000835 fiber Substances 0.000 description 6
- 239000000499 gel Substances 0.000 description 6
- 239000003550 marker Substances 0.000 description 6
- 239000000203 mixture Substances 0.000 description 6
- 238000003757 reverse transcription PCR Methods 0.000 description 6
- 239000011780 sodium chloride Substances 0.000 description 6
- SHIBSTMRCDJXLN-UHFFFAOYSA-N Digoxigenin Natural products C1CC(C2C(C3(C)CCC(O)CC3CC2)CC2O)(O)C2(C)C1C1=CC(=O)OC1 SHIBSTMRCDJXLN-UHFFFAOYSA-N 0.000 description 5
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 5
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 5
- 108091027981 Response element Proteins 0.000 description 5
- 101150026889 VAMP2 gene Proteins 0.000 description 5
- 241000607479 Yersinia pestis Species 0.000 description 5
- 230000003542 behavioural effect Effects 0.000 description 5
- 238000009395 breeding Methods 0.000 description 5
- 230000001488 breeding effect Effects 0.000 description 5
- 230000030570 cellular localization Effects 0.000 description 5
- 230000001419 dependent effect Effects 0.000 description 5
- UQLDLKMNUJERMK-UHFFFAOYSA-L di(octadecanoyloxy)lead Chemical compound [Pb+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O UQLDLKMNUJERMK-UHFFFAOYSA-L 0.000 description 5
- QONQRTHLHBTMGP-UHFFFAOYSA-N digitoxigenin Natural products CC12CCC(C3(CCC(O)CC3CC3)C)C3C11OC1CC2C1=CC(=O)OC1 QONQRTHLHBTMGP-UHFFFAOYSA-N 0.000 description 5
- SHIBSTMRCDJXLN-KCZCNTNESA-N digoxigenin Chemical compound C1([C@@H]2[C@@]3([C@@](CC2)(O)[C@H]2[C@@H]([C@@]4(C)CC[C@H](O)C[C@H]4CC2)C[C@H]3O)C)=CC(=O)OC1 SHIBSTMRCDJXLN-KCZCNTNESA-N 0.000 description 5
- 229940079593 drug Drugs 0.000 description 5
- 230000008030 elimination Effects 0.000 description 5
- 238000003379 elimination reaction Methods 0.000 description 5
- 238000005516 engineering process Methods 0.000 description 5
- 239000011521 glass Substances 0.000 description 5
- 239000007924 injection Substances 0.000 description 5
- 238000002347 injection Methods 0.000 description 5
- 230000033001 locomotion Effects 0.000 description 5
- 239000003531 protein hydrolysate Substances 0.000 description 5
- 230000007017 scission Effects 0.000 description 5
- 230000035939 shock Effects 0.000 description 5
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- 101150039504 6 gene Proteins 0.000 description 4
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 4
- 108020004705 Codon Proteins 0.000 description 4
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 4
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 4
- 102000013265 Syntaxin 1 Human genes 0.000 description 4
- 108010055044 Tetanus Toxin Proteins 0.000 description 4
- 210000001084 basket cell Anatomy 0.000 description 4
- 230000033228 biological regulation Effects 0.000 description 4
- 230000002490 cerebral effect Effects 0.000 description 4
- 238000010367 cloning Methods 0.000 description 4
- 239000003085 diluting agent Substances 0.000 description 4
- 238000012869 ethanol precipitation Methods 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 108020001507 fusion proteins Proteins 0.000 description 4
- 102000037865 fusion proteins Human genes 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- QWTDNUCVQCZILF-UHFFFAOYSA-N isopentane Chemical compound CCC(C)C QWTDNUCVQCZILF-UHFFFAOYSA-N 0.000 description 4
- 239000002609 medium Substances 0.000 description 4
- 238000000520 microinjection Methods 0.000 description 4
- 230000008685 targeting Effects 0.000 description 4
- 229940118376 tetanus toxin Drugs 0.000 description 4
- 238000005406 washing Methods 0.000 description 4
- WFDIJRYMOXRFFG-UHFFFAOYSA-N Acetic anhydride Chemical compound CC(=O)OC(C)=O WFDIJRYMOXRFFG-UHFFFAOYSA-N 0.000 description 3
- 108010085238 Actins Proteins 0.000 description 3
- 229920000936 Agarose Polymers 0.000 description 3
- 241000894006 Bacteria Species 0.000 description 3
- 108030001720 Bontoxilysin Proteins 0.000 description 3
- 241000193449 Clostridium tetani Species 0.000 description 3
- 108020003215 DNA Probes Proteins 0.000 description 3
- 239000003298 DNA probe Substances 0.000 description 3
- 101710088194 Dehydrogenase Proteins 0.000 description 3
- 241000588724 Escherichia coli Species 0.000 description 3
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 3
- 239000004471 Glycine Substances 0.000 description 3
- 108091028043 Nucleic acid sequence Proteins 0.000 description 3
- 108020004518 RNA Probes Proteins 0.000 description 3
- 239000003391 RNA probe Substances 0.000 description 3
- 108091081024 Start codon Proteins 0.000 description 3
- 108091023040 Transcription factor Proteins 0.000 description 3
- 102000040945 Transcription factor Human genes 0.000 description 3
- 102000003786 Vesicle-associated membrane protein 2 Human genes 0.000 description 3
- 108090000169 Vesicle-associated membrane protein 2 Proteins 0.000 description 3
- 230000009471 action Effects 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 230000000692 anti-sense effect Effects 0.000 description 3
- 230000000903 blocking effect Effects 0.000 description 3
- 210000004899 c-terminal region Anatomy 0.000 description 3
- 210000005056 cell body Anatomy 0.000 description 3
- 238000005119 centrifugation Methods 0.000 description 3
- 238000005520 cutting process Methods 0.000 description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 3
- 238000012137 double-staining Methods 0.000 description 3
- 235000020188 drinking water Nutrition 0.000 description 3
- 239000003651 drinking water Substances 0.000 description 3
- 210000002216 heart Anatomy 0.000 description 3
- 229910001385 heavy metal Inorganic materials 0.000 description 3
- 230000006801 homologous recombination Effects 0.000 description 3
- 238000002744 homologous recombination Methods 0.000 description 3
- 230000001771 impaired effect Effects 0.000 description 3
- 210000003734 kidney Anatomy 0.000 description 3
- 238000002372 labelling Methods 0.000 description 3
- 150000002632 lipids Chemical class 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 210000003101 oviduct Anatomy 0.000 description 3
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N phenol group Chemical group C1(=CC=CC=C1)O ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 3
- 230000001766 physiological effect Effects 0.000 description 3
- 238000000746 purification Methods 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 238000010825 rotarod performance test Methods 0.000 description 3
- 238000012216 screening Methods 0.000 description 3
- 210000002966 serum Anatomy 0.000 description 3
- 235000020183 skimmed milk Nutrition 0.000 description 3
- 210000004500 stellate cell Anatomy 0.000 description 3
- 235000000346 sugar Nutrition 0.000 description 3
- 230000002103 transcriptional effect Effects 0.000 description 3
- 230000000007 visual effect Effects 0.000 description 3
- CFBILACNYSPRPM-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;2-[[1,3-dihydroxy-2-(hydroxymethyl)propan-2-yl]amino]acetic acid Chemical compound OCC(N)(CO)CO.OCC(CO)(CO)NCC(O)=O CFBILACNYSPRPM-UHFFFAOYSA-N 0.000 description 2
- 102000007469 Actins Human genes 0.000 description 2
- 239000012099 Alexa Fluor family Substances 0.000 description 2
- 108020005544 Antisense RNA Proteins 0.000 description 2
- 206010003591 Ataxia Diseases 0.000 description 2
- 101100328189 Bacillus anthracis clpP2 gene Proteins 0.000 description 2
- 238000011740 C57BL/6 mouse Methods 0.000 description 2
- VTYYLEPIZMXCLO-UHFFFAOYSA-L Calcium carbonate Chemical compound [Ca+2].[O-]C([O-])=O VTYYLEPIZMXCLO-UHFFFAOYSA-L 0.000 description 2
- 206010010947 Coordination abnormal Diseases 0.000 description 2
- 108010051219 Cre recombinase Proteins 0.000 description 2
- 238000012270 DNA recombination Methods 0.000 description 2
- 241000255581 Drosophila <fruit fly, genus> Species 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical class CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 2
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 2
- MHAJPDPJQMAIIY-UHFFFAOYSA-N Hydrogen peroxide Chemical compound OO MHAJPDPJQMAIIY-UHFFFAOYSA-N 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 101001041236 Mus musculus Ornithine decarboxylase Proteins 0.000 description 2
- 102000004390 N-Ethylmaleimide-Sensitive Proteins Human genes 0.000 description 2
- 108010081735 N-Ethylmaleimide-Sensitive Proteins Proteins 0.000 description 2
- SEQKRHFRPICQDD-UHFFFAOYSA-N N-tris(hydroxymethyl)methylglycine Chemical compound OCC(CO)(CO)[NH2+]CC([O-])=O SEQKRHFRPICQDD-UHFFFAOYSA-N 0.000 description 2
- 102100023206 Neuromodulin Human genes 0.000 description 2
- 101710144282 Neuromodulin Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 238000009004 PCR Kit Methods 0.000 description 2
- 229910019142 PO4 Inorganic materials 0.000 description 2
- 239000002033 PVDF binder Substances 0.000 description 2
- 229930040373 Paraformaldehyde Natural products 0.000 description 2
- 208000018737 Parkinson disease Diseases 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 102000000583 SNARE Proteins Human genes 0.000 description 2
- 108010041948 SNARE Proteins Proteins 0.000 description 2
- 229930006000 Sucrose Natural products 0.000 description 2
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 2
- 239000007997 Tricine buffer Substances 0.000 description 2
- 235000010724 Wisteria floribunda Nutrition 0.000 description 2
- 238000002835 absorbance Methods 0.000 description 2
- 230000006978 adaptation Effects 0.000 description 2
- 239000011543 agarose gel Substances 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 239000000074 antisense oligonucleotide Substances 0.000 description 2
- 238000012230 antisense oligonucleotides Methods 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 229940053031 botulinum toxin Drugs 0.000 description 2
- 235000011089 carbon dioxide Nutrition 0.000 description 2
- 230000001364 causal effect Effects 0.000 description 2
- 239000003184 complementary RNA Substances 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 230000000593 degrading effect Effects 0.000 description 2
- 238000012217 deletion Methods 0.000 description 2
- 230000037430 deletion Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- AFABGHUZZDYHJO-UHFFFAOYSA-N dimethyl butane Natural products CCCC(C)C AFABGHUZZDYHJO-UHFFFAOYSA-N 0.000 description 2
- 201000010099 disease Diseases 0.000 description 2
- VYFYYTLLBUKUHU-UHFFFAOYSA-N dopamine Chemical compound NCCC1=CC=C(O)C(O)=C1 VYFYYTLLBUKUHU-UHFFFAOYSA-N 0.000 description 2
- 238000001035 drying Methods 0.000 description 2
- 210000002257 embryonic structure Anatomy 0.000 description 2
- 238000010195 expression analysis Methods 0.000 description 2
- 239000000834 fixative Substances 0.000 description 2
- -1 for example Substances 0.000 description 2
- 238000010230 functional analysis Methods 0.000 description 2
- 239000005090 green fluorescent protein Substances 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 229940088597 hormone Drugs 0.000 description 2
- 238000003364 immunohistochemistry Methods 0.000 description 2
- 208000016290 incoordination Diseases 0.000 description 2
- 230000010189 intracellular transport Effects 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 230000004807 localization Effects 0.000 description 2
- 238000004519 manufacturing process Methods 0.000 description 2
- 230000007246 mechanism Effects 0.000 description 2
- 239000003068 molecular probe Substances 0.000 description 2
- 230000000877 morphologic effect Effects 0.000 description 2
- 229940126619 mouse monoclonal antibody Drugs 0.000 description 2
- 210000003205 muscle Anatomy 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 230000003955 neuronal function Effects 0.000 description 2
- 229920002866 paraformaldehyde Polymers 0.000 description 2
- 239000010452 phosphate Substances 0.000 description 2
- 239000004033 plastic Substances 0.000 description 2
- 229920003023 plastic Polymers 0.000 description 2
- 229920002401 polyacrylamide Polymers 0.000 description 2
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 239000012723 sample buffer Substances 0.000 description 2
- 238000000926 separation method Methods 0.000 description 2
- 210000000813 small intestine Anatomy 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 241000894007 species Species 0.000 description 2
- ATHGHQPFGPMSJY-UHFFFAOYSA-N spermidine Chemical compound NCCCCNCCCN ATHGHQPFGPMSJY-UHFFFAOYSA-N 0.000 description 2
- 239000005720 sucrose Substances 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 230000014616 translation Effects 0.000 description 2
- 239000001226 triphosphate Substances 0.000 description 2
- 235000011178 triphosphate Nutrition 0.000 description 2
- UNXRWKVEANCORM-UHFFFAOYSA-N triphosphoric acid Chemical compound OP(O)(=O)OP(O)(=O)OP(O)(O)=O UNXRWKVEANCORM-UHFFFAOYSA-N 0.000 description 2
- GPRLSGONYQIRFK-MNYXATJNSA-N triton Chemical compound [3H+] GPRLSGONYQIRFK-MNYXATJNSA-N 0.000 description 2
- 239000011534 wash buffer Substances 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- 210000004885 white matter Anatomy 0.000 description 2
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 2
- DNXIKVLOVZVMQF-UHFFFAOYSA-N (3beta,16beta,17alpha,18beta,20alpha)-17-hydroxy-11-methoxy-18-[(3,4,5-trimethoxybenzoyl)oxy]-yohimban-16-carboxylic acid, methyl ester Natural products C1C2CN3CCC(C4=CC=C(OC)C=C4N4)=C4C3CC2C(C(=O)OC)C(O)C1OC(=O)C1=CC(OC)=C(OC)C(OC)=C1 DNXIKVLOVZVMQF-UHFFFAOYSA-N 0.000 description 1
- PLRACCBDVIHHLZ-UHFFFAOYSA-N 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine Chemical compound C1N(C)CCC(C=2C=CC=CC=2)=C1 PLRACCBDVIHHLZ-UHFFFAOYSA-N 0.000 description 1
- VZSRBBMJRBPUNF-UHFFFAOYSA-N 2-(2,3-dihydro-1H-inden-2-ylamino)-N-[3-oxo-3-(2,4,6,7-tetrahydrotriazolo[4,5-c]pyridin-5-yl)propyl]pyrimidine-5-carboxamide Chemical compound C1C(CC2=CC=CC=C12)NC1=NC=C(C=N1)C(=O)NCCC(N1CC2=C(CC1)NN=N2)=O VZSRBBMJRBPUNF-UHFFFAOYSA-N 0.000 description 1
- AXAVXPMQTGXXJZ-UHFFFAOYSA-N 2-aminoacetic acid;2-amino-2-(hydroxymethyl)propane-1,3-diol Chemical compound NCC(O)=O.OCC(N)(CO)CO AXAVXPMQTGXXJZ-UHFFFAOYSA-N 0.000 description 1
- FTAHXMZRJCZXDL-UHFFFAOYSA-N 3-piperideine Chemical compound C1CC=CCN1 FTAHXMZRJCZXDL-UHFFFAOYSA-N 0.000 description 1
- DGZSVBBLLGZHSF-UHFFFAOYSA-N 4,4-diethylpiperidine Chemical compound CCC1(CC)CCNCC1 DGZSVBBLLGZHSF-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 101150010303 AMP2 gene Proteins 0.000 description 1
- 208000024827 Alzheimer disease Diseases 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 102100026189 Beta-galactosidase Human genes 0.000 description 1
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 1
- 102000016838 Calbindin 1 Human genes 0.000 description 1
- 108010028310 Calbindin 1 Proteins 0.000 description 1
- 241000282836 Camelus dromedarius Species 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- 241000282552 Chlorocebus aethiops Species 0.000 description 1
- 241000193403 Clostridium Species 0.000 description 1
- 241000193155 Clostridium botulinum Species 0.000 description 1
- 108091026890 Coding region Proteins 0.000 description 1
- 208000019736 Cranial nerve disease Diseases 0.000 description 1
- 238000000018 DNA microarray Methods 0.000 description 1
- 238000001712 DNA sequencing Methods 0.000 description 1
- 241000252212 Danio rerio Species 0.000 description 1
- 102000016911 Deoxyribonucleases Human genes 0.000 description 1
- 108010053770 Deoxyribonucleases Proteins 0.000 description 1
- 208000020401 Depressive disease Diseases 0.000 description 1
- SNRUBQQJIBEYMU-UHFFFAOYSA-N Dodecane Natural products CCCCCCCCCCCC SNRUBQQJIBEYMU-UHFFFAOYSA-N 0.000 description 1
- 101150039808 Egfr gene Proteins 0.000 description 1
- 241000710188 Encephalomyocarditis virus Species 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- HVLSXIKZNLPZJJ-TXZCQADKSA-N HA peptide Chemical compound C([C@@H](C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](C(C)C)C(=O)N1[C@@H](CCC1)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](C)C(O)=O)NC(=O)[C@H]1N(CCC1)C(=O)[C@@H](N)CC=1C=CC(O)=CC=1)C1=CC=C(O)C=C1 HVLSXIKZNLPZJJ-TXZCQADKSA-N 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 102000010789 Interleukin-2 Receptors Human genes 0.000 description 1
- 108060001084 Luciferase Proteins 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- 239000007993 MOPS buffer Substances 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 239000012901 Milli-Q water Substances 0.000 description 1
- 101001135571 Mus musculus Tyrosine-protein phosphatase non-receptor type 2 Proteins 0.000 description 1
- 241000244206 Nematoda Species 0.000 description 1
- 208000012902 Nervous system disease Diseases 0.000 description 1
- 108091092724 Noncoding DNA Proteins 0.000 description 1
- 206010029719 Nonspecific reaction Diseases 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 240000007817 Olea europaea Species 0.000 description 1
- 240000007594 Oryza sativa Species 0.000 description 1
- 235000007164 Oryza sativa Nutrition 0.000 description 1
- 102000001675 Parvalbumin Human genes 0.000 description 1
- 108060005874 Parvalbumin Proteins 0.000 description 1
- 102000035195 Peptidases Human genes 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 229940124158 Protease/peptidase inhibitor Drugs 0.000 description 1
- 102000015799 Qa-SNARE Proteins Human genes 0.000 description 1
- 108010010469 Qa-SNARE Proteins Proteins 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- LCQMZZCPPSWADO-UHFFFAOYSA-N Reserpilin Natural products COC(=O)C1COCC2CN3CCc4c([nH]c5cc(OC)c(OC)cc45)C3CC12 LCQMZZCPPSWADO-UHFFFAOYSA-N 0.000 description 1
- QEVHRUUCFGRFIF-SFWBKIHZSA-N Reserpine Natural products O=C(OC)[C@@H]1[C@H](OC)[C@H](OC(=O)c2cc(OC)c(OC)c(OC)c2)C[C@H]2[C@@H]1C[C@H]1N(C2)CCc2c3c([nH]c12)cc(OC)cc3 QEVHRUUCFGRFIF-SFWBKIHZSA-N 0.000 description 1
- 229920005654 Sephadex Polymers 0.000 description 1
- 239000012507 Sephadex™ Substances 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 description 1
- UZMAPBJVXOGOFT-UHFFFAOYSA-N Syringetin Natural products COC1=C(O)C(OC)=CC(C2=C(C(=O)C3=C(O)C=C(O)C=C3O2)O)=C1 UZMAPBJVXOGOFT-UHFFFAOYSA-N 0.000 description 1
- 101710137500 T7 RNA polymerase Proteins 0.000 description 1
- 108020005038 Terminator Codon Proteins 0.000 description 1
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Triethanolamine Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 1
- 108091023045 Untranslated Region Proteins 0.000 description 1
- SXEHKFHPFVVDIR-UHFFFAOYSA-N [4-(4-hydrazinylphenyl)phenyl]hydrazine Chemical compound C1=CC(NN)=CC=C1C1=CC=C(NN)C=C1 SXEHKFHPFVVDIR-UHFFFAOYSA-N 0.000 description 1
- YVNQAIFQFWTPLQ-UHFFFAOYSA-O [4-[[4-(4-ethoxyanilino)phenyl]-[4-[ethyl-[(3-sulfophenyl)methyl]amino]-2-methylphenyl]methylidene]-3-methylcyclohexa-2,5-dien-1-ylidene]-ethyl-[(3-sulfophenyl)methyl]azanium Chemical compound C1=CC(OCC)=CC=C1NC1=CC=C(C(=C2C(=CC(C=C2)=[N+](CC)CC=2C=C(C=CC=2)S(O)(=O)=O)C)C=2C(=CC(=CC=2)N(CC)CC=2C=C(C=CC=2)S(O)(=O)=O)C)C=C1 YVNQAIFQFWTPLQ-UHFFFAOYSA-O 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 230000001800 adrenalinergic effect Effects 0.000 description 1
- 108010045649 agarase Proteins 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 238000013528 artificial neural network Methods 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 230000037429 base substitution Effects 0.000 description 1
- BLFLLBZGZJTVJG-UHFFFAOYSA-N benzocaine Chemical compound CCOC(=O)C1=CC=C(N)C=C1 BLFLLBZGZJTVJG-UHFFFAOYSA-N 0.000 description 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- 230000008033 biological extinction Effects 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 230000003925 brain function Effects 0.000 description 1
- UDSAIICHUKSCKT-UHFFFAOYSA-N bromophenol blue Chemical compound C1=C(Br)C(O)=C(Br)C=C1C1(C=2C=C(Br)C(O)=C(Br)C=2)C2=CC=CC=C2S(=O)(=O)O1 UDSAIICHUKSCKT-UHFFFAOYSA-N 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 229910002092 carbon dioxide Inorganic materials 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000003915 cell function Effects 0.000 description 1
- 230000006727 cell loss Effects 0.000 description 1
- 210000000170 cell membrane Anatomy 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 210000003591 cerebellar nuclei Anatomy 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 235000013330 chicken meat Nutrition 0.000 description 1
- XFIOKOXROGCUQX-UHFFFAOYSA-N chloroform;guanidine;phenol Chemical compound NC(N)=N.ClC(Cl)Cl.OC1=CC=CC=C1 XFIOKOXROGCUQX-UHFFFAOYSA-N 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 230000002759 chromosomal effect Effects 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 101150034144 ci gene Proteins 0.000 description 1
- 230000009194 climbing Effects 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000000576 coating method Methods 0.000 description 1
- 230000003750 conditioning effect Effects 0.000 description 1
- 238000002425 crystallisation Methods 0.000 description 1
- 230000008025 crystallization Effects 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 230000003247 decreasing effect Effects 0.000 description 1
- 239000007857 degradation product Substances 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000018044 dehydration Effects 0.000 description 1
- 238000006297 dehydration reaction Methods 0.000 description 1
- 210000001787 dendrite Anatomy 0.000 description 1
- 230000003001 depressive effect Effects 0.000 description 1
- 229960000633 dextran sulfate Drugs 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- KCFYHBSOLOXZIF-UHFFFAOYSA-N dihydrochrysin Natural products COC1=C(O)C(OC)=CC(C2OC3=CC(O)=CC(O)=C3C(=O)C2)=C1 KCFYHBSOLOXZIF-UHFFFAOYSA-N 0.000 description 1
- 238000007865 diluting Methods 0.000 description 1
- 238000010790 dilution Methods 0.000 description 1
- 239000012895 dilution Substances 0.000 description 1
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- 230000006806 disease prevention Effects 0.000 description 1
- UKWLRLAKGMZXJC-QIECWBMSSA-L disodium;[4-chloro-3-[(3r,5s)-1-chloro-3'-methoxyspiro[adamantane-4,4'-dioxetane]-3'-yl]phenyl] phosphate Chemical compound [Na+].[Na+].O1OC2([C@@H]3CC4C[C@H]2CC(Cl)(C4)C3)C1(OC)C1=CC(OP([O-])([O-])=O)=CC=C1Cl UKWLRLAKGMZXJC-QIECWBMSSA-L 0.000 description 1
- 208000035475 disorder Diseases 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 229960003638 dopamine Drugs 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 108700021358 erbB-1 Genes Proteins 0.000 description 1
- DNJIEGIFACGWOD-UHFFFAOYSA-N ethyl mercaptane Natural products CCS DNJIEGIFACGWOD-UHFFFAOYSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000003203 everyday effect Effects 0.000 description 1
- 230000001747 exhibiting effect Effects 0.000 description 1
- 239000004744 fabric Substances 0.000 description 1
- 239000012894 fetal calf serum Substances 0.000 description 1
- 238000009408 flooring Methods 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 230000008014 freezing Effects 0.000 description 1
- 238000007710 freezing Methods 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 238000012239 gene modification Methods 0.000 description 1
- 238000012637 gene transfection Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 230000005017 genetic modification Effects 0.000 description 1
- 235000013617 genetically modified food Nutrition 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 230000003284 homeostatic effect Effects 0.000 description 1
- 238000000265 homogenisation Methods 0.000 description 1
- 238000011532 immunohistochemical staining Methods 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- JEIPFZHSYJVQDO-UHFFFAOYSA-N iron(III) oxide Inorganic materials O=[Fe]O[Fe]=O JEIPFZHSYJVQDO-UHFFFAOYSA-N 0.000 description 1
- 238000011813 knockout mouse model Methods 0.000 description 1
- 210000005240 left ventricle Anatomy 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 230000001665 lethal effect Effects 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000003137 locomotive effect Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 238000000504 luminescence detection Methods 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 239000012139 lysis buffer Substances 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 230000004879 molecular function Effects 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 239000003176 neuroleptic agent Substances 0.000 description 1
- 230000000701 neuroleptic effect Effects 0.000 description 1
- 230000007604 neuronal communication Effects 0.000 description 1
- 210000004940 nucleus Anatomy 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 229920006284 nylon film Polymers 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000032696 parturition Effects 0.000 description 1
- 238000010827 pathological analysis Methods 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 235000021401 pellet diet Nutrition 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 230000010412 perfusion Effects 0.000 description 1
- 230000002572 peristaltic effect Effects 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000008363 phosphate buffer Substances 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000003518 presynaptic effect Effects 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 235000019833 protease Nutrition 0.000 description 1
- 229940076376 protein agonist Drugs 0.000 description 1
- 229940076372 protein antagonist Drugs 0.000 description 1
- 238000002731 protein assay Methods 0.000 description 1
- 230000007026 protein scission Effects 0.000 description 1
- 230000012743 protein tagging Effects 0.000 description 1
- 208000020016 psychiatric disease Diseases 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- BJOIZNZVOZKDIG-MDEJGZGSSA-N reserpine Chemical compound O([C@H]1[C@@H]([C@H]([C@H]2C[C@@H]3C4=C([C]5C=CC(OC)=CC5=N4)CCN3C[C@H]2C1)C(=O)OC)OC)C(=O)C1=CC(OC)=C(OC)C(OC)=C1 BJOIZNZVOZKDIG-MDEJGZGSSA-N 0.000 description 1
- 229960003147 reserpine Drugs 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 239000003161 ribonuclease inhibitor Substances 0.000 description 1
- 210000003705 ribosome Anatomy 0.000 description 1
- 235000009566 rice Nutrition 0.000 description 1
- MDMGHDFNKNZPAU-UHFFFAOYSA-N roserpine Natural products C1C2CN3CCC(C4=CC=C(OC)C=C4N4)=C4C3CC2C(OC(C)=O)C(OC)C1OC(=O)C1=CC(OC)=C(OC)C(OC)=C1 MDMGHDFNKNZPAU-UHFFFAOYSA-N 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 201000000980 schizophrenia Diseases 0.000 description 1
- 230000035945 sensitivity Effects 0.000 description 1
- 239000012679 serum free medium Substances 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 230000003584 silencer Effects 0.000 description 1
- 238000001179 sorption measurement Methods 0.000 description 1
- 230000021595 spermatogenesis Effects 0.000 description 1
- 229940063673 spermidine Drugs 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 210000003523 substantia nigra Anatomy 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- FRGKKTITADJNOE-UHFFFAOYSA-N sulfanyloxyethane Chemical compound CCOS FRGKKTITADJNOE-UHFFFAOYSA-N 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 210000002504 synaptic vesicle Anatomy 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 230000002194 synthesizing effect Effects 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 230000000451 tissue damage Effects 0.000 description 1
- 231100000827 tissue damage Toxicity 0.000 description 1
- 230000002588 toxic effect Effects 0.000 description 1
- 230000005758 transcription activity Effects 0.000 description 1
- 238000012250 transgenic expression Methods 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- PIEPQKCYPFFYMG-UHFFFAOYSA-N tris acetate Chemical compound CC(O)=O.OCC(N)(CO)CO PIEPQKCYPFFYMG-UHFFFAOYSA-N 0.000 description 1
- 210000000689 upper leg Anatomy 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- 238000011816 wild-type C57Bl6 mouse Methods 0.000 description 1
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/635—Externally inducible repressor mediated regulation of gene expression, e.g. tetR inducible by tetracyline
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
-
- A—HUMAN NECESSITIES
- A01—AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
- A01K—ANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
- A01K2217/00—Genetically modified animals
- A01K2217/05—Animals comprising random inserted nucleic acids (transgenic)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2830/00—Vector systems having a special element relevant for transcription
- C12N2830/001—Vector systems having a special element relevant for transcription controllable enhancer/promoter combination
- C12N2830/002—Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor
- C12N2830/003—Vector systems having a special element relevant for transcription controllable enhancer/promoter combination inducible enhancer/promoter combination, e.g. hypoxia, iron, transcription factor tet inducible
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2840/00—Vectors comprising a special translation-regulating system
- C12N2840/20—Vectors comprising a special translation-regulating system translation of more than one cistron
- C12N2840/203—Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/195—Assays involving biological materials from specific organisms or of a specific nature from bacteria
- G01N2333/33—Assays involving biological materials from specific organisms or of a specific nature from bacteria from Clostridium (G)
Definitions
- the present invention relates to a method for analyzing the function of a specific nerve cell. More specifically, a DNA having a structure capable of reversibly controlling the expression of a protein having a neuronal transmission function controlling activity in a specific nerve cell, a host into which the DNA has been introduced, and a host into which the DNA has been introduced.
- the present invention relates to a method for analyzing the function of a specific nerve cell by reversibly controlling the expression of a protein having a neuronal communication function controlling activity in the specific nerve cell.
- mice and knockout mice have been produced by remarkable progress in molecular biology in recent years, and it has become possible to artificially change the expression level of genes in individuals.
- this technique can be used to determine the phenotype of a gene when the expression of a gene increases or, conversely, when the expression of a gene is lost. It was limited to the functions of “molecules” such as what kind of work it did. In other words, there was no effective means to specifically analyze the functions of nerve cells, which are located at a higher level than molecules and are also basic units of neural networks.
- IMCT Immunonotoxin—Mediated Ceil Targeting
- Watanabe and Nakanishi who are also inventors of the present invention
- the IMCT method uses molecular biology to create transgenic mice that express a fusion protein of human IL-2 receptor and a marker in a specific region of the brain. This is a revolutionary method that allows specific cells to be destroyed and deleted at any stage of growth by administering a recognizing antibody toxin.
- the IMCT method has its limitations, and is not necessarily a sufficient method for analyzing neuronal function.
- the cerebral nervous system is an extremely plastic organ, and when a certain neuron is deleted, compensation and adaptation by other cells occur in the course of long-term observation.
- the IMCT method irreversibly causes nerve cells to be lost due to cell death. Therefore, it is possible to convert a mouse from a normal state to a defective state, but it is not possible to return the mouse from the defective state to a normal state again. Therefore, irreversible neuronal cell loss methods such as the IMCT method have a problem that they cannot accurately evaluate cell functions hidden by compensation and adaptation phenomena.
- Toxin proteins produced by tetanus and botulinum bacteria when taken up into nerve cells, release proteins (SNARE: Soluble N-ethylmaleimide-sensitive fusion protein Attachment proteins REceptor) that release neurotransmitters. Acts as a specifically degrading protease. As a result, nerve cells affected by this neurotoxin protein are unable to release neurotransmitters, preventing the transmission of information to subsequent cells without relying on cell death.
- SNARE Soluble N-ethylmaleimide-sensitive fusion protein Attachment proteins REceptor
- this toxin protein derived from tetanus and botulinum is expected to be useful as a protein having a neuronal transmission function controlling activity.
- spermatogenesis is abnormal (Eisel, U., et al., (1993) ⁇ J., 12, 3365-3372).
- the present invention provides a method for controlling the cerebral nervous system by reversibly controlling the expression of a protein having a neuronal transmission function-regulating activity, rather than deleting specific neurons by irreversible cell death.
- the task is to provide a very effective tool for analyzing networks.
- the present inventors have, GABA A a 6 pro motor (Jones that are specifically activated in certain nerve cells, A., et al, (1996 ) J. Neurochem, 67, 907 - 916;.. Bahn, Natl. Acad. Sci. USA, 94, 9417-9421), using a tetracycline-dependent transcriptional regulatory system, to produce neurotoxin genes from botulinum and tetanus bacteria.
- DNA having a structure capable of reversible regulation of expression was injected into mouse fertilized eggs by mouth, and genetically modified mice were produced.
- GA- BA A c 6 promoter specifically neurotransmitter release in cells activated has found to be controlled.
- the present invention has been accomplished based on such knowledge.
- the present invention relates to (1) reversibly controlling the expression of a protein having a nerve cell transmission function regulating activity in a specific nerve cell, comprising DNA encoding a protein having a nerve cell transmission function regulating activity.
- the host according to (5) which reversibly controls the expression of a protein having a neurotransmitter function controlling activity in a specific nerve cell, and the biological function in the nerve cell or a cell associated therewith.
- a method for analyzing the functions of biological functions and molecules characterized by analyzing physical and chemical changes of molecules.
- (9) (1) (a) activated by a specific stimulus, which is linked to a transcription control region DNA that is specifically activated in a specific nerve cell and is placed under the control of the transcription control region; And DN'A encoding a protein capable of activating a specific promoter, and (b) a promoter controlled by the protein and a neurotransmitter release controlling activity linked so as to be under the control thereof.
- a DNA encoding a protein having the same is introduced into a host; Biofunction A method for analyzing biological function and molecular function, characterized by analyzing physical and chemical changes of offspring.
- (11) (a) A transcriptional control region DNA specifically activated in a specific nerve cell and linked to be placed under the control of the transcriptional control region, activated by a specific stimulus, and DNA encoding a protein capable of activating a specific promoter, and (b) a promoter controlled by the protein and a protein having a neurotransmitter release controlling activity linked so as to be under the control of the promoter. Fertilized eggs, embryonic stem cells, and neural stem cells of non-human animals that carry the encoding DNA *
- FIG. 1 is a diagram showing a DNA encoding a tetracycline-dependent transcriptional regulator constructed in an example of the present invention.
- (1) shows the structure of the SphI / NotI9.3 Kbp fragment of the pVEC6 (ll) vector used for microinjection into mouse fertilized eggs.
- (2) shows the structure of the endogenous GABA A a6 gene.
- 1, wide frame gray both 2 is a Ipushironkaiomikuron'ita ⁇ area of GABA A alpha 6, the white width Hosowaku are IRES, wide frame diagonal line represents the rtTA gene.
- the numbers indicate the size (kbp) of the DNA in each region as a bold line indicating the lower probe setting region.
- FIG. 2 is a diagram showing a DNA encoding a neuronal transmission function control protein constructed in an example of the present invention.
- the numbers indicate the size (kbp) of the DNA in each region.
- the bold line shown as the Probe setting area at the bottom shows the area corresponding to the probe sequence used in Southern hybridization.
- BoNT There is only one BoNT. A, BoNT. B, BoNT. E, and TeNT.
- BoNT BoNT. CI is another area in other areas.
- Xbal # There is only one location for BoNT. A, BoNT. B, and BoNT. E.
- BoNT.E is another in other areas.
- 3 is a diagram showing the results of Genomusazanha Eve lida I See Chillon analysis of genetically modified mice introduced with GABA A ct 6- rtTA.
- (2) shows the results of genomic Southern hybridization analysis of genetically modified mice into which treTeNTdl has been introduced.
- the upper numbers indicate the individual numbers of the genetically modified mice as samples, and the underline added to the individual numbers indicates that the mice are transgene-positive mice.
- the size marker of DNA is shown on the left side of the figure.
- the long arrow indicates the position of the signal derived from the endogenous gene, and the short arrow indicates the position of the signal derived from the transgene. Transgene positive lines are underlined below the number.
- FIG. 4 is a diagram showing the correspondence between the structure of the neurotoxin substrate protein and the site of cleavage by the neurotoxin used in this example.
- the letters A, B, Cl, and E indicate the serotype of the neurotoxin BoNT, and the added arrows indicate the sites where each neurotoxin cleaves the substrate.
- the hatched area indicates the Four-helix bundle area (Fasshauer, D., et al., (1998) Proc. Natl. Acad. Sci. USA, 95, 15781-15786), and is blacked out.
- the region indicates a transmembrane region.
- the numbers at the bottom represent the sequence numbers of the amino acids on the N-terminal side at each boundary.
- FIG. 4 is a diagram showing the results of Western blotting analysis of the substrate protein cleavage activity of the toxin for quality.
- the molecular weight markers of the proteins are shown on the left side of the figure.
- the long arrow indicates the position of the target protein that has not been cleaved, and the short arrow indicates the position of the target protein that has been cleaved.
- Figure 6 is a graph showing the results of mRNA Northern blot analysis in GABA A a6- rtTA transgenic mice.
- FIG. 7 is a diagram showing the results of in situ hybridization analysis in GABA A a6-tTA transgenic mice.
- FIG. 8 shows the results of Western blotting for induction and elimination of neurotoxin protein expression in mice transgenic for the ABA A a6-rtTA gene and the treToxin gene.
- Figure 9 shows wild-type mice treated with doxycycline for 1 week (6 weeks old)
- Figure 9A Genotype:-/-, D0X: +
- Figure 9B Genotype: + / +, D0X:-
- Fig. 9C Genotype: + / +, D0X: +
- FIG. 2 shows the results of the anti-EGFP antibody and the secondary antibody and subsequent antibodies performed by the ABC method.
- FIG. 10 is a diagram showing the results of behavioral analysis by a rota-rod test using mice.
- Fig. 11 is a diagram showing the results of behavior analysis by a balance beam test using mice. BEST MODE FOR CARRYING OUT THE INVENTION
- a protein having an activity of controlling a neuronal transmission function and a DNA encoding the protein refers to a protein having an activity of controlling information transmission between a specific nerve cell and another cell in a living body. .
- a protein capable of directly controlling the release of a neurotransmitter hereinafter referred to as a “neurotransmitter release control protein” is preferable. Any device that can control the cell transmission function may be used.
- the neuronal transmission function control protein used in the present invention for example, a neurotransmitter release control protein, reversibly inhibits the release of a neurotransmitter when introduced into a living body or conversely when removed from the living body.
- a neurotransmitter release control protein reversibly inhibits the release of a neurotransmitter when introduced into a living body or conversely when removed from the living body.
- neurotoxin examples include tetanus toxin protease (TeNT) and botulinum toxin protease (BoNT). These may be derived from any species or type of tetanus or Clostridium botulinura, but are preferably tetanus (Clostridium tetani Harvard A-47; accession number: KZ1174, Japanese Society of Cytology, 1995, Vol. 50, No. 4, 1023) or from botulinum (BoNT. A (Binz, T., et al., (1990) J. Biol. Chem., 265, 9153-9158), ⁇ . ⁇ (Kurazono, ⁇ ., Et al., (1992) J.
- TeNT tetanus toxin protease
- BoNT botulinum toxin protease
- neuronal transmission function control proteins for example, those described in the above-mentioned documents can be used, but one or several amino acid substitutions or deletions as long as the protein has the neuronal transmission function control activity Those having, or addition or inversion can also be used as neuronal transmission function control proteins.
- a marker and a tag sequence can be added to the N-terminus or C-terminus of the protein, or can be inserted therein.
- the marker for example, GFP (Green Fluorescent Protein) ⁇ -galactosidase, luciferase and the like can be used.
- Any tag sequence may be used as long as it is commonly used as an epitope tag.
- a Flag tag, an HA tag, an HIS tag, or the like can be used.
- a sequence that affects the intracellular transport and localization of the protein, and a sequence that regulates the stability of the protein can be added to the N-terminal or C-terminal of the protein, or inserted into the protein. You can also.
- Sequences that can regulate protein stability include PEST sequences such as Mouse Orni-thine Decarboxylase (Ghoda, Shi, et al., (1989) Science, 243, -1493-1495; Li, X., et. al., (1998) J. Biol. Chem., 273, 34970-34975).
- the DNA encoding the neuronal transmission function controlling protein used in the present invention can be obtained, for example, from cells expressing the above-described neuronal transmission function controlling protein.
- the type of DNA may be any as long as it encodes a protein having a neuronal transmission function controlling activity, and may be either genomic DNA or cDNA, but is preferably cDNA.
- cDNA contains only coding region or non-coding region
- mRNA in cells expressing a neuronal transmission function controlling protein such as RT-PCR (Reverse Transcription—Polymerase Chain Reaction), PCR, or hybridization, from a cDNA library. It can be obtained by a method known per se such as the zation method.
- the DNA encoding the neuronal transmission function-controlling protein used in the present invention is not limited to the DNA obtained as described above, but also to the extent that the protein encoded by the DNA does not inhibit the neuronal transmission function-regulating activity. Transcription, translation, or intracellular localization of mRNA on the 'side or 5' side, or inside the DN.A ( Mayford, M., et al., (1996) Proc. Natl. Acad. Sci.
- a polyA signal sequence linked to the 3 ′ side of the translation region DNA can be used.
- the polyA signal sequence a sequence contained in the 3, untranslated region of the cDNA used may be used, or an exogenous sequence such as Rabbit / 3-globin ⁇ ⁇ olyA signal sequence may be used. Good.
- BoNT.A BoNT, T., et al., (1990) J. Biol. Chem., 265, 9153-9158
- BoNT.B Kurazono, H., et al., ( 1992) J. Biol. Chem., 267, 14721-1 ⁇ 4729
- BoNT. CI Hauser, D., et al., (1990) Nucleic Acids Res., 18, 4924 ⁇
- BoNT. D Boz , T et al., (1990) Nucleic Acid's Res., 18, 5566
- BoN-TE BoN-TE
- Neuronal transmission function control DNA DNA encoding the above-mentioned neuronal transmission function control protein
- neuronal transmission function control DNA has an expression capable of reversibly controlling its expression in specific nerve cells.
- the expression control unit of the neuronal transmission function control protein of the present invention hereinafter, this may be referred to as “gene expression control unit” or “expression control unit of neuronal transmission function protein”
- gene expression control unit or “expression control unit of neuronal transmission function protein”
- a gene expression control unit examples include (a) a DNA structure capable of reversibly controlling the expression of a protein having a neuronal transmission function controlling activity in a specific nerve cell; Those containing DNA encoding a protein having a neuronal transmission function controlling activity linked so as to be placed under the control of the DNA structure.
- a specific nerve cell means a cell in which a network between the cell and a cell associated with the cell changes due to the expression of a neuronal transmission function control protein in the cell.
- Specific examples include cerebellar granule cells and the like.
- a system for reverse control is a system that can control gene expression irreversibly under specific conditions and that can reversibly control gene expression specifically in specific nerve cells. If it is, any thing may be used.
- a system for reversibly controlling the expression of a gene can use the pedestrian method itself.
- a tetracycline expression control system that controls gene expression by the presence or absence of tetracycline (Gossen, M., and Bujard, H., (1992) Proc. Natl. Acad. Sci. USA., 89, 5547- 5551; U.S. Patent No.
- the system is controlled under the control of a motor that is activated only in a specific nerve cell.
- a method is used in which a system is connected or a specific condition for reversibly controlling gene expression in the system can be selectively performed only in a specific nerve cell. Whether the expression control unit of the present invention is selectively activated only in a target neuron is determined by reversibly controlling gene expression after introducing the unit into a suitable host.
- the above-described tetracycline expression control system is used. Things. Examples of specific configurations of the unit include: (a) a DNA in which a tetracytaline-responsive element and a promoter controlled by a tetracycline-responsive element are linked to the 5′-side upstream of the neuronal transfer function-control DNA of the present invention; *, And (b) DNA linked so that the DNA encoding the tetracycline expression regulator is under the control of a promoter sequence that is specifically activated in a specific nerve cell. These two DNA fragments are each constructed as independent DNA fragments, their ligations, or plasmids into which they have been inserted.
- the ligation of these DNAs is preferably as described above, but by regulating the presence or absence or the amount of tetracytaline in cells into which the expression control unit of the present invention has been introduced, a protein for controlling a neuronal transmission function can be obtained. If the expression of can be controlled reversibly, it may be moved to another site or the sequence may be changed. Further, it is preferable that at least one appropriate intron is inserted in addition to the above-described configuration.
- the above-mentioned tetracycline response element may be any DNA as long as it can bind to a tetracycline expression control factor and has a function of activating a downstream promoter by this binding. Linoperator (Gossen, M. and Bujard, H., (1992) Proc. Natl. Acad. Sci. USA, 89, 55-47-5551) and the like are used.
- any promoter may be used as long as it has no activity by itself and is controlled by a tetracycline response element.
- a CMV minimum promoter can be used.
- tetracycline or a derivative thereof Factors that induce gene expression under the control of the tetracycline response element when administered or not administered are included.
- rtTA Reverse tetracycli ne-controlled transactivator: Gossen, M., et al., (1995) Science, 268, 17-66-1769
- a certain tTA Tetracycline-controlled transactivator: Gossen, ⁇ M., and Bujard, H., (1992) Proc. Natl. Acad. Sci. USA, 89, 5547-5551).
- tetracycline expression regulatory DNA DNA encoding the above-mentioned tetracycline expression regulator (hereinafter sometimes referred to as “tetracycline expression regulatory DNA”) is ligated so as to be under the control of a promoter that is activated in a specific nerve cell.
- a promoter sequence that is activated in a specific nerve cell is linked to the 5 ′ upstream of tetracycline expression regulatory DNA, or a cell that is introduced by homologous recombination.
- DNA in which tetracycline expression regulatory DNA is located downstream of a promoter that is activated in specific endogenous neurons on the chromosome can also be used.
- RNA Ribonucleic acid
- DNA Ribonucleic acid
- GABA A a 6 pro motor
- the DNA sequence of the promoter may include a gene DNA controlled by the promoter downstream of the 3 ′ side.
- the IRES Internal
- the IRES is located between the gene and the tetracycline expression regulatory DNA of the present invention. ribosomal entry site).
- tetracytaline expression control unit of the present invention (a) a tetracytaline responsive element and a promoter sequence controlled by the tetracycline responsive element are linked to the 5 'upstream of the neuronal transfer function control DNA. And (b) transcription or translation of the DNA under specific conditions upstream of a non-specific or tetracycline expression regulatory DNA linked under the control of a promoter that is activated in specific neurons. (C) a DNA linked to a promoter that activates a specific neuron-inducing factor in a specific nerve cell. Be composed. These three DNAs are preferably constructed as independent separate DNA fragments or plasmids into which they have been inserted.However, three or two are arranged side by side to form one DNA fragment, or It can also be built as a plus.
- the arrangement of these DNAs is preferably as described above. However, by regulating the presence or absence or the amount of tetracycline in the cells into which these DNAs have been introduced, the expression of the neuronal transmission function control protein can be reversibly controlled. If so, you can move it to another location or change the arrangement. Furthermore, it is preferable that at least one intron is included in these transcription units.
- tetracycline responsive element and the promoter sequence controlled by the tetracycline responsive element those described in the above (i) can be used.
- those described in the above (i) can be used.
- the promoter that functions in specific nerve cells as the promoter that functions in all cells, for example, one actin promoter is preferably used. .
- the mechanism for inhibiting or releasing the expression of the tetracytaline expression regulator induced by this promoter under specific conditions includes, for example, Cre-lox system (S-ternberg, N., and Hamilton, D. , (1981) J. Mol. Biol., 150, 467-486; Sauer, B., and Henderson, N., (1988) Proc. Natl. Acad. Sci. USA, 85, 5166-5170; Gu. , H., et al., (1994) Science, 265, 103-106) and the FLP-FRT system. (G-olic, KG, and Lindquist, SL, (1989) Cell, 44, 499-509) can be used.
- Cre-lox system When the Cre-lox system is used, a DNA is used in which a stop codon having a ⁇ sequence linked to both ends is ligated between the non-specific promoter and the tetracycline expression control DNA. Furthermore, as a factor inducing a specific condition, Cre recombinase having an activity of deleting a region flanked by a sequence by DNA recombination is used. The DNA encoding the factor is ligated downstream of the promoter that is activated in the specific nerve cell described in (i) above.
- gene expression control units include: (a) a promoter sequence for an appropriate stress protein, a neuronal transmission function control DN-A operably linked to the sequence, (B) a factor linked to a DNA upstream of which a DNA that inhibits the transcription or translation of the gene under specific conditions is activated; It is composed of DNA that is linked to be under control.
- these two DNAs are constructed as independent and separate DNA fragments or as plasmids into which they are inserted, however, the two are arranged side by side as one DNA fragment or as a plasmid into which they are inserted. Can also be built.
- these DNAs are preferably as described above, but the expression of the neuronal transmission function control protein is reversible by regulating the presence or absence or the degree of appropriate stress shock in the cells into which these DNAs have been introduced. If you can control it, you may move it to another location or change the arrangement. Furthermore, these expression control units preferably contain at least one intron.
- any one can be used as long as it can be activated by applying an appropriate stress to cells into which the unit has been introduced.
- heat Shock protein promoters any one can be used as long as it can be activated by applying an appropriate stress to cells into which the unit has been introduced.
- the mechanism that inhibits or releases the expression of the neuronal transmission function control DNA induced by this promoter under specific conditions includes, for example, the Cre-lox system (Stern-berg, N., and Hamilton, D., (1981) J. Mol. Biol., 150, 467-486; Sauer, B., and Henderson, N., (1988) Proc. Natl. Acad. Sci. USA, 85, 5166-5170 Gu, ⁇ ⁇ , et al., (1994) Science, 265, 103-106) and FLP-FRT system (Golic-, KG, and Lindquist, S. Shishi, (1989) Cell, 44, 499-509) Can be used.
- Cre-lox system Stern-berg, N., and Hamilton, D., (1981) J. Mol. Biol., 150, 467-486; Sauer, B., and Henderson, N., (1988) Proc. Natl. Acad. Sci. USA, 85, 5166-5170 Gu, ⁇ ⁇ , e
- Cre-lox system When the Cre-lox system is used, a DNA in which a stop codon having both ends bound to each other between the stress protein promoter and the DNA for controlling a neuronal transmission function can be used. Further, as a factor for inducing a specific condition, Cre recombinase having an activity of deleting a region flanked by the ⁇ sequences by DNA recombination is used. The DNA encoding the factor is ligated downstream of the promoter that is activated in the specific nerve cell described in (i) above.
- a method in which a DNA having a structure in which a nerve cell transmission function control DNA is linked is introduced into a host downstream of the stress protein promoter, and stress is directly applied only to specific nerve cells (Halfon, MS, Natl. Acad. Sci. USA, 94, 6255-6260; Halloran, MC, et al., (2000) Development, 127, 1953-1960), etc. Can also be used.
- the gene expression control unit of the present invention is prepared by a method known per se, and a recombinant is obtained by introducing the same into an appropriate host. can do.
- a host into which the gene expression control unit of the present invention can be introduced any host can be used as long as the unit can be introduced and the recombinant can reversibly control the expression of a neurotransmitter release control protein. It may be something. Specifically, for example, cultured cells such as PC12 (Green, Shi, et al., (1976) Proc. Natl. Acad. Sci. USA, 73, 2 ⁇ 424-2428), nervous system cultured cells, primary cells Nervous system tissue Examples include rice culture, fertilized eggs, embryonic stem cells, neural stem cells, and organisms other than humans.
- Embryonic stem cells refer to all cells that can develop into an individual, and neural stem cells refer to all cells that can differentiate into nerve cells.
- living organisms include nematodes, Drosophila, zebrafish, birds such as chickens, and mammals such as monkeys, rats, and mice. Of these, rodents are preferred, and mice are more preferred.
- rodents are preferred, and mice are more preferred.
- As a mouse strain it is preferable to use the C57BL / 6 strain in consideration of the phenotypic analysis of the above-described expression control unit-introduced transgenic cells, but it is difficult to breed and obtain fertilized eggs, Other strains can be used, taking into account other factors such as cell lineage.
- a method for introducing the expression control unit of the neuronal transmission function controlling protein of the present invention into such a host a commonly used method known per se can be used.
- the host is a cell such as a cultured cell, a nervous system cultured cell, a primary cell, a nervous tissue slice culture, a fertilized egg, an embryonic stem cell, or a neural stem cell, specifically, for example, a microinjection method, a calcium phosphate method, A lipofection method, an electoral poration method, a virus infection method, or the like can be used.
- the gene expression control unit is composed of a plurality of DNA fragments
- the whole expression control unit can be constructed in the host body by breeding the independently introduced animals.
- a gene transfectant in each host such as a method of introducing DNA into the above-mentioned fertilized egg or embryonic stem cell and then generating the DNA, a homologous recombination method, or a transposon method.
- the method usually used for the method can be used.
- a commonly used method known per se can be used. You. Specifically, for example, a method of introducing a drug-resistant gene together with the above expression control unit, treating the host with the drug, and selecting a strain having resistance,
- a method using the expression of the marker gene described in (2) as an index is exemplified.
- it can be confirmed by Southern blotting, Northern blotting, Western blotting, or the like using DNA, RNA, or protein extract prepared from part or all of the introduced host.
- the host into which each of the thus obtained DNA fragments of the expression control unit of the present invention has been introduced may be simply referred to as a “DNA transductant”.
- a “DNA transductant” The host into which each of the thus obtained DNA fragments of the expression control unit of the present invention has been introduced.
- the method for introducing the expression control unit of the neuronal transmission function control protein of the present invention (the method for producing a genetically modified animal) will be specifically described, taking as an example the case where the host is an animal individual such as a mouse.
- animals into which the expression control unit of the neuronal transmission function controlling protein has been introduced are prepared by the above (2)
- those that do not need to be position-specifically introduced are purified after removing unnecessary regions such as the vector sequence used for cloaking.
- inject into the pronucleus of the fertilized egg of the target animal by the method described in (3) above, specifically, by microinjection.
- the injected fertilized egg is preferably transplanted into the oviduct of a pseudopregnant animal.
- the tail of a litter after weaning is cut, genomic DNA is purified, fragmented with an appropriate restriction enzyme, and then the nucleotide sequence of the introduced expression control unit of the present invention is identified.
- a method for performing Southern hybridization using a probe capable of performing the hybridization is described. It can also be confirmed by amplifying the nucleotide sequence of the introduced expression control unit of the present invention by PCR.
- mice into which each DNA fragment of the above has been introduced.
- mice in which tetracycline expression regulatory DNA has been introduced so as to be controlled by a neuron-specific promoter hereinafter sometimes referred to as “Tet mouse”
- Neuronal transfer function control Mice into which DNA encoding a protein in a state where expression of the protein can be regulated has been introduced (hereinafter sometimes referred to as “TransReg mouse”).
- a DNA having homology with the DNA sequence at the target position of the host to be introduced is added before and after the DNA fragment of the expression control unit of the present invention. It can be performed by using the targeting vector thus prepared. Specifically, homologous recombination is induced in animal embryonic stem cells using the method described in Thomas, KR, et al., (1987) Cell, 51, 503-512. The method can be performed by obtaining a chimeric individual by the method.
- the genetically modified animal obtained above can be used as F0 to obtain F1 by crossing with a wild-type animal of the same strain.
- the method described in (4) can be used to determine whether the DNA introduced into the obtained F1 is transmitted.
- breeding passages with F2 and F3 are performed, and the genomic DNA of the offspring obtained in the same manner is analyzed to confirm the presence or absence of the introduced ⁇ '.
- an animal carrying a DNA having a specific nucleotide sequence can be made into a line, it can be subcultured in a normal breeding environment.
- the gene expression control unit of the present invention is constructed in a recombinant host. can do.
- whether the introduced gene is actually transcribed in the individual and translated into a protein, and whether the transcription and translation are performed in the target region. Whether it is performed or not is determined by the usual methods such as Northern blotting using RNA extracted from each tissue, in situ hybridization, Western blotting using proteins extracted from each tissue, or immunochemistry. It can be analyzed and confirmed by the methods that have been used.
- the DNA transfectant of the present invention can be obtained by placing the transfectant under conditions suitable for the introduced gene expression control system. Can reversibly control the expression of the neuronal transmission function regulatory protein.
- the conditions suitable for the gene expression control system are, for example, when the gene expression control system used is a tetracytaline gene expression control system, the transcription activity of a tetracycline expression regulator such as doxycycline is determined. It means to regulate the administration of the substance to be regulated and the like.
- the expression of the protein can be reversibly controlled by adjusting the addition, non-addition, or addition concentration of the substance to the culture solution.
- the expression of the protein can be reversibly regulated by administering or not administering the substance to the DNA transductant.
- a method of administering the substance to an individual a method of administering the substance by mixing it with feed or water is preferable.
- Direct injection into the digestive tract, intraperitoneal injection, direct administration to the brain, etc. can also be used.
- a cell, a tissue, or a slice derived from the above-described DNA-introduced individual organism is used to control a neuronal transmission function control protein, it is preferable to control the concentration of the substance in a medium.
- the analysis of the phenotype expressed in the DNA transductant by reversible expression control of the neuronal transmission function control protein was performed by adjusting the expression level of the neuronal transmission function control protein in the transductant by the method described in (6) above. This can be done by analyzing differences in biochemical, physiological, morphological, or behavioral changes that appear in cells, tissues, living organisms, etc., before, after, and after, or from the wild type. it can.
- the phenotype to be analyzed and the method for analyzing the phenotype can be determined by appropriately combining known methods which are generally used.
- the phenotypes to be analyzed in the present invention include those that can be observed with the eyes that appear in cells and living organisms, those that require physical and chemical analysis, and those that express the above-mentioned neuronal transmission function controlling proteins. Includes all phenotypes that differ by presence, absence, or degree. Among these, phenotypes that can be observed with the eye include the shape of the transductant, which is determined by morphological analysis, and, if the host is an animal, its behavior, motor ability, learning ability, habit, and the like.
- the phenotype to be analyzed is the shape of the DNA transfectant described above
- examples include an optical microscope, an electron microscope, and visual observation.
- a maze, a rotarod, a conditioning preference test, and the like which are usually used as a means of analyzing the behavior of the animal, can be mentioned.
- observing the potential inside the cell or outer membrane, the capacitance of the membrane, etc. include commonly used electrophysiological analysis methods.
- the phenotype is a chemical change or a quantitative change of a biological function molecule such as nucleic acid, protein, lipid, sugar, etc.
- Northern blotting, RT-PCR, in situ hybridization, DNA chip, Western blotting, 2D Analysis can be performed by electrophoresis, chromatography, immunochemistry, mass spectrometry, and the like.
- the function of a specific nerve cell can be analyzed.
- a neuronal transmission function control protein By analyzing the expression of a neuronal transmission function control protein, the above-mentioned phenotype that appears when reversibly changing information transmission between other cells to which a signal is transmitted from a specific nerve cell, The causal relationship between neuronal function and phenotype can be clarified.
- biofunctional molecule means a substance having a function on a living body, such as a nucleic acid, a protein, a lipid, and a sugar.
- a method for analyzing these functions for example, the expression of a neuronal transmission function control protein is controlled in a specific neuron by the method of the present invention, and information transmission between other cells to which a signal is transmitted from the neuron is reversibly performed.
- a physical change such as a quantitative change of a biofunctional molecule, a change of a three-dimensional structure, or a change of intracellular localization in the nerve cell and other cells described above, a phosphorylation, etc.
- a method of analyzing the degree of chemical modification of the compound or whether or not a chemical change such as a change in location occurs. By performing such an analysis method, it is possible to clarify the causal relationship between the phenotype appearing in the living body and the function of the biological functional molecule.
- the DNA transfectant of the present invention can be used as various pathological model cells or pathological model animals based on the phenotype and the gene possessed.
- Specific diseases showing the pathological conditions of these models include, for example, psychiatric diseases such as schizophrenia and depression, and neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease.
- the genetically modified animal having the expression control unit of the neuronal transmission function controlling protein of the present invention can be crossed with another genetically modified animal or a host exhibiting a characteristic phenotype.
- a pathological model of a disease involving a plurality of genes can be prepared, and identified using the model. It is possible to screen a substance having a physiological activity.
- This PCR resulted in a specific amplified band of about 400 bp. This band was excised, purified, and subcloned into the pCR2.1 vector (Invitrogen). This plasmid was used as pCRExonl vector.
- the pCRExonl prepared in (11) above was cut with the restriction enzyme EcoRI (manufactured by T0Y0B0), and the DNA fragment containing the insert was purified.
- the purified DNA fragment of about 400 bp was converted into type I, and the hybridization probe ( ⁇ 3.2-dCTP (Amersham Pharmacia Biotech)) was labeled by the multiprime method using BcaBEST Labeling Kit (TAKARA). Exonl probe) was prepared.
- genomic BAC library derived from 129SVJ mouse ES cells 92160 clones contained in Mouse BAC filter Release II (Genome Systems) were screened by hybridization, and 9 Positive clones (Grid7, field2, 21-f, 308; Grid7, field3, 6-j, 333; G-rid7, field3, 16-1, 315; Grid7, field5, 1g, 329; Grid8, fieldl, 15-0, Grid9, field4, 23-f, 400; Grid9, field4, 24-b, 388; Grid9, field. 5, 9-b, 425; Grid10, fieldl, 6-h, 451) were obtained.
- the rtTA gene to which the IRES (Internal Ribosome Entry Site: GenBank Accession No .: 277 to 874 of X74312) derived from Encephalomyocarditis virus is added on the 5 'side is 5 of the ATg translation initiation sequence of the rtTA gene and the IRES sequence in the upstream region. It was obtained by repeating PCR in which partial regions were sequentially added as a fore-primer sequence. All primers used were prepared by requesting Hokkaido System Science.
- the PCR primers used were the forward primers corresponding to the IRES sequence from 277 “C” to 874 "g” of GenBank Accession No .: X74312 (MYK151a to 1511 ⁇ : SEQ ID NOs: 3 to 11, Hokkaido System Science) And rtTA A reverse primer (MYK152: SEQ ID NO: 12, produced by request of Hokkaido-System Science) for the vicinity of the Sacl region in the gene was used.
- Type ⁇ pTet-On vector (Clontech) was used for PCR using MYK151a (SEQ ID NO: 3) and MYK152, and the second PCR using MYK151b / MY-K152
- the plasmid obtained by subcloning the DNA fragment amplified in the above was purified to form type I.
- the forward primer ⁇ 1 ⁇ 51a
- a base sequence of ggC is inserted immediately below the ATg translation start sequence so as to obtain a base sequence that does not shift the reading frame of the rtTA gene (glycine is inserted as the amino acid sequence). Designed to introduce the Ncol (CCATgg) restriction enzyme site.
- ⁇ 151 ⁇ (SEQ ID NO: 11, produced by request of Hokkaido System Science) has a base sequence obtained by adding a linker sequence to the 5 'upstream of the above IRES sequence.
- a DNA fragment of about 1470 bp was amplified by PCR using MYK151i / MY-K152 as a primer. The DNA fragment was subcloned into a pCR2.1 vector (manufactured by Invitrogen) to obtain a pSEQ2i vector.
- the rtTA gene encoded in the pTet-On vector contains S A Sph-I restriction enzyme site (gCATg / C) is present (base position 1615 in the pTet-On vector). Substituting "C” for "T” in the Sphl restriction enzyme site can eliminate the Sphl restriction enzyme site while preserving the amino acid sequence to be encoded.
- MY-K149 (SEQ ID NO: 14, produced by requesting Hokkaido System Science) is a forward primer for performing this base substitution.
- MYK150 (SEQ ID NO: 15, produced by Hokkaido System Science) introduces a restriction enzyme site in the order of NotI, BglII, and EcoRI from the 5 'side to the 3' downstream region of the SV40 polyA signal sequence. This is a reverse primer for performing
- the obtained pSEQl vector was digested with Sacl and EciRI, a fragment of about 700 bp was purified, and then subcloned into the SacI / EcoRI region of a pUC18 vector (manufactured by T0Y0B0) to obtain a pVE-C1 vector.
- the obtained PSEQ2 vector was cut with Sphl and Sacl, a fragment of about 1460 bp was purified, and then subcloned into the Sphl / Sacl region of the pVECl vector to obtain a pVEC2 vector.
- the PVEC2 vector is a vector containing a sequence in which the IRES, rtTA, and SV40 late polyA signal sequences are functionally linked in order from the 5 'side in the pUC18 cloning site.
- Reverse PCR: YK158 SEQ ID NO: 19, produced by request of Hokkaido System Science
- PCR with PVEC2 as type III performed An approximately 700 bp fragment was amplified from the vicinity containing the second ATg-sequence (ATg translation initiation sequence) to the vicinity of the sequence containing the BsiWI (Spll) restriction enzyme site in the rtTA gene. This DNA fragment was designated as fragment B.
- Fragment A and Fragment B contain a sequence overlapping near the 11th ATg sequence (ATg translation initiation sequence) in the IRES region. Using the overlapping sequences, fragment A and fragment B were ligated (PCR Ligation).
- the prepared pAB vector was cut with PmaCI and BsiWI (SphI), a fragment of about 900 bp was purified, and ligated into a pVEC2 vector cut out with PmaCI / BaiWI (SphI).
- the prepared vector was used as a pVEC2 (11) vector.
- PVEC6 (11) is, GABA A a 6 expression regulatory region in order from the 5 'side during the cloning site of the P UO 18, IRES ,: rtTA, This is a vector containing a sequence in which the SV40 polyA signal sequence is functionally linked. This structure is shown in Figure 11.
- Example 2 Construction of a transgene encoding a neuronal transmission function regulatory protein
- the neurotoxin (TeNT) gene derived from tetanus was cultured from Clostridium tetani strain KZ1174 (transferred from Prof. Shinichi Nakamura, Department of Microbiology, School of Medicine, Kanazawa University) (Professor Mitsuaki Nishibuchi, Kyoto University Southeast Asian Studies Center) MYK121 (SEQ ID NO: 20, produced by Hokkaido System Science) and MYK126 (SEQ ID NO: 21, produced by Hokkaido System Science) after boiling the bacteria in the culture for 15 minutes. Prepared by request) Obtained by PCR using primers. By this PCR, a DNA fragment of about 1.4 kbp was amplified and purified, followed by subcloning into a pCR2.1 vector (manufactured by Invitrogen) to obtain a pCRTeNT vector.
- MYK121 (SEQ ID NO: 20) is a forward primer for adding a Mlu I restriction enzyme site to the region upstream of the ATg translation initiation codon of the TeNT light chain gene.
- MYK126 (SEQ ID NO: 21) is a reverse primer for adding a Not I restriction enzyme site to the 3, downstream region of the TeNT light chain gene.
- MYK121, MYK126 primer arrangement In designing the column, the registered sequence of GenBank Accession No .: # 0443 was referred to.
- MYK098 (SEQ ID NO: 22) adds an S-acll restriction enzyme site and Kozak sequence (Kozak,., (1986) Cell, 44, 283-292) to the 5 'upstream region of the ATg translation initiation codon of the EGFP gene.
- MYK099 (SEQ ID NO: 23) is used to add a Mlul restriction enzyme site to the 3 'downstream region of the AAg sequence encoding lysine 239 of the dlEGFP gene in the pdlEGF-P-N1 vector (Clontech).
- Reverse primer is used to add a Mlul restriction enzyme site to the 3 'downstream region of the AAg sequence encoding lysine 239 of the dlEGFP gene in the pdlEGF-P-N1 vector (Clontech).
- telomere sequence When PCR was performed using the pdl EGFP-N1 vector as a type I, MYK098 and MYK099 as primers, a DNA fragment of about 750 bp was amplified. This fragment was digested with SacII and Mlul, and the purified fragment of about 740 bp was ligated to the SacII / Mul restriction site of the pTRE2 vector. The prepared vector was used as pTREEGFP vector.
- YK119 (SEQ ID NO: 24, produced by request of Hokkaido System Science) is a Notl restriction enzyme in the 5 and upstream regions of the AgC sequence encoding the amino acid number 242 serine of the dlEGF. This is a forward primer for adding a site and one base of "T" (adjusting the reading frame to the protein).
- MYK101 (SEQ ID NO: 25, produced by request of Hokkaido System Science) is a reverse primer for adding an Xbal restriction enzyme site to the downstream region of the dlEGFP gene at the TAg translation termination codon.
- telomere sequence When PCR was performed using the pdlEGFP-N1 vector as a type I and MYK119 and MYK101 as primers, a DNA fragment of about 150 bp was amplified. This fragment was cut with Notl and Xbal, and the purified fragment of about 140 bp was ligated to the Notl / Xbal restriction enzyme site of pTREEGFP. The prepared vector was used as a pTREEGFPdl vector.
- PCR was carried out to add Mlul for cloning to the 5 'end and Notl at the 3' end of each BoNT gene obtained in (1-1a) above.
- Mlul restriction in 5 'upstream of ATg translation codon of each BoNT light chain gene A MYK102 (SEQ ID NO: 26) for BoNT.
- A YK105 (SEQ ID NO: 27) for BoNT.
- B MYK103 (SEQ ID NO: 28) for BoNT.
- C1 oNT For E, thigh 091 (SEQ ID NO: 29) was produced by request of Hokkaido System Science.
- a reverse primer a Notl restriction enzyme site was added to the 3 ′ downstream region of each BoNT light chain gene.
- BoNT.A MYK107 (SEQ ID NO: 30), and for BoNT.B, MYK110 (SEQ ID NO: 31), MYK108 (SEQ ID NO: 32) for BoNT. C1, and YK096 (SEQ ID NO: 33) for BoNT. E were produced by requesting Hokkaido System Science. PCR was performed using the plasmid obtained in the above (l_a) for the type I DNA and the combination of the primers for each BoNT described above.
- BoNT genes were amplified by this PCR. About 1.4 kbp- DNA fragment for BoNT. A, about 1.3 kbp DNA fragment for BoNT. B, about 1.4 kbp- DNA fragment for BoNT. C1, and about 1. A 3 kbp DNA fragment was amplified, purified, and subcloned into a pCR2.1 vector (manufactured by Invitorogen).
- the plasmid obtained by subcloning each BoNT gene obtained above was cut with Mlul and Notl, respectively, to obtain a DNA fragment of about 1.3 to 1.4 Kbp.
- the Mlul / Notl restriction enzyme site of the pTREEGFPdl vector obtained above was subjected to sparing.
- the prepared vectors were referred to as pTREBoNT. Adl, ⁇ REBoNT. Bdl, pTREBoNT. Cldl, pTREBoNT. Edl, and pTRETeNTdl vectors, respectively.
- These vectors contain a PhCMV promoter containing a tetracycline response element, a gene encoding a neurotoxin protein fused with EGFP on the N-terminal side, and a PEST sequence of Mouse Ornithine Decarboxylase on the C-terminal side.
- a vector containing a functionally arranged region This structure is shown in FIG. Note that an intron is included near the Rabbit] 3-globin polyA signal sequence.
- DNA sample preparation for injection into the pronucleus of mouse fertilized eggs The pVEC6 (11) prepared in Example 1 (3) above was cleaved with Sphl and Notl, and the cleavage reaction solution was 1% low melting point agarose (Life Technologies, Inc .: (Catalog 15517-014) /0.5 X TBE (0 The gel was electrophoresed on 045M Tris-borate, 0.001M EDTA) gel.Agarose gel around 9.3kbp was cut out, the agarose was melted with 70, and then ⁇ ⁇ with GELase (EPICENTR-E TECHNOLOGIES).
- the DNA fragment of about 9.3 kbp was purified by phenolic mouth-mouth treatment and ethanol precipitation, and the DNA after ethanol precipitation was transferred to TE (10 mM Tris (pH 8.0), IraM EDTA). After dissolving, the concentration was estimated by measuring the absorbance at 260 nm, and adjusted to lOng / ⁇ ⁇ with calcium- and magnesium-free Dulbecco's PBS (Phosphate®-uffered Saline: manufactured by Life Technologies). It was used for gene transfer into mouse fertilized eggs.
- pTREBoNT. Adl, pTREBoNT. Bdl, pTREBoNT. Edl, and pTRETeNTdl obtained in Example 2 (2) above were cut with Xhol and Sapl, and the obtained DNA fragment of about 4 kbp was prepared in the same manner as described above. This sample was used for gene transfer into mouse fertilized eggs.
- Example 2 (2) the pTREBoNT.Cldl obtained in Example 2 (2) above was digested with Aatll and Sapl (the Xhol restriction enzyme site was present in the NT.CI gene, so the Aatll restriction enzyme site was used instead of Xhol. ) And the resulting DNA fragment of about 4 kbp was prepared in the same manner as described above. Each of these samples was used for gene transfer into mouse fertilized eggs.
- Pronuclear stage fertilized eggs obtained by crossing C57BL / 6 mice were collected by fallopian tube perfusion and used as test eggs to be used for the injection operation.
- each DNA solution adjusted to 10 ng // il was filled in a microinjection capillary and injected into the pronucleus of a pronuclear fertilized egg.
- the eggs that survived after the injection were transferred to a fresh culture solution, washed, and cultured in a carbon dioxide incubator until they became 2-cell stage embryos.
- the two-cell stage embryos were transferred into the oviduct of a foster female mouse that induced pseudopregnancy and obtained a litter.
- the obtained offspring were confirmed for sex and weaned 3 weeks after delivery.
- the resulting genetic modification Approximately 4 weeks after parturition, mice were individually identified, and the success or failure of gene transfer was analyzed by Southern hybridization.
- a 6-rtTA is introduced genetically modified Mausu tail destination end about 1cm at the 3 (2), in a lysis buffer (manufactured by KURAB0 companies), 55 ° C, 6 Incubate for ⁇ 16 hours to dissolve most of the tail. Residues were removed from the lysate by centrifugation, and genomic DNA was purified through phenol / cloth form treatment and ethanol precipitation. In order to degrade the contaminating RNA, the genomic DNA was dissolved in TE containing lOOngZml RNaseA (manufactured by Na-kalai tesque).
- the genomic DNA purified from the tail above was digested with restriction enzymes, BamHI, and Sphl, and electrophoresed with 0.9% agarose gel Z1X TAE (0.04M Tris-acetate, 0.001M EDTA). Blotting was carried out on a nylon membrane (GeneScreen Plus: NEN Lifescience) by the capillary method using 4N NaOH, 0.6M NaCl as a blocking solution.
- the blotted membrane is dried in a hybridization buffer (1 M NaCl 50 mM Tris (pH 7.5) 10% Dextran Sulfate ⁇ 200ug / ml Sonicated Salmon Sperm DNA- (STRATAGENE: Catalog # 201190-81) in 1% SDS), o to prepare a pre-hybridization die internalization in row-,, at 60 ° C; the addition of P32- dCTP in ⁇ ⁇ onl probe GABA a a 6 labeled at 60, over ⁇ A hybridization reaction was performed.
- a hybridization buffer 1 M NaCl 50 mM Tris (pH 7.5) 10% Dextran Sulfate ⁇ 200ug / ml Sonicated Salmon Sperm DNA- (STRATAGENE: Catalog # 201190-81) in 1% SDS
- mice In wild type mice, about 14kbp only the sheet Gunaru of GABA A a 6 gene endogenous (arrow in FIG 31) is detected, in addition, signaling of about 8. Lkbp (arrowheads in Fig 31) The line in which the gene is detected is the transgene-positive mouse.
- Mice The GABA A ct 6- rtTA. Was introduced could 10 lines obtained in total. These mice GABA A a 6 - - it was rtTA mice.
- Genome was prepared in the same manner as in 1), and genomic DNA prepared from the genetically modified mouse in which treBoNT.Edl was introduced was Kpnl and Xbal, and treBoNT.Adl, treBoNT.Bd-1, treBoNT.Cldl, or treTeNTdl were introduced. Genomic DNA prepared from the genetically modified mouse was cut with Kpnl and EcoRI and blotted.
- an EGFR gene DNA fragment was obtained by the following PCR.
- Forward primer: MYK189 (SEQ ID NO: 34) has a sequence in which an EcoRI restriction enzyme site and a Kozak sequence are added from the 5 'side to the 5' upstream of the ATg translation initiation sequence of the EGFP gene.
- MYK220 (SEQ ID NO: 35) an oligo DNA having a sequence in which a Hindlll restriction enzyme site was added to the 3 'downstream region of the EGFP gene was produced by requesting Hokkaido System Science.
- a DNA fragment of about 750 bp was amplified, purified, and subcloned into a pCR2.1 vector (manufactured by Invitorogen) to prepare a pCREGFP vector.
- This pCREGFP vector was cut with EcoRI and Hindlll to purify a DNA fragment of about 720 bp, and then subcloned into the EcoRI and Hindlll restriction sites of the pGEM-4Z (Promega) vector to produce pGEMEGFP.
- This pCREGFP was cleaved with EcoRI and Hindlll, and a fragment of about 720 bp was used as type II. The fragment was labeled with a P32-dCTP (Araersham Pharmacia Biotech) by the multiprime method using the BcaBEST Labeling Kit (TAKARA). Preparation of hybridization probe (EGFP probe) did.
- Example 3 Using this EGFP probe and performing hybridization in the same manner as in Example 3 (3-1) above, a representative treTeNTdl-modified mouse is shown in FIG. 32. No signal is detected in wild-type mice, but about 2.3 kbp
- Adl, pTREBoNT. Bdl, pTREBoNT. Edl, or pTRETeNTdl were introduced were referred to as treBoNT.
- the mouse into which the about 4 Kbp Aatll / Sapl fragment of pTREBoNT. Cldl was introduced was designated as treBoNT. Cldl mouse.
- a vector that constantly expresses the neurotoxin protein contained in the vector DNA encoding a protein serving as a substrate for a neurotoxin was co-expressed in cultured cells, and the resulting cleavage of the substrate protein was analyzed.
- Syntaxin 1A (288 amino acid residues, calculated molecular weight 33. lkD)
- SNAP25A Synaptosomal Associated Ro ⁇ Rotein of 25KD (206 amino acid residues, calculated molecular weight 23.3 kD)
- VAMP2 Vesicle Associated Membrane Protein (116 amino acid residues, calculated molecular weight 12.7 kD).
- SNARE soluble N-ethylmaleimide-sensitive factor attachraen * t protein receptors proteins, which are known to play an important role in the process of synaptic vesicles containing neurotransmitters fusing to the presynaptic membrane.
- BoNT. C1 requires about 1000 times more protein than BoNT.
- BoNT. E and the main toxic activity is thought to be that Syntaxin 1A is used as a substrate. .
- the pTREBoNT.Adl vector prepared in Example 2 (2) above was digested with SacII, and the resulting SacII-cut end was blunt-ended with K0D DM Polymerase (T0Y0B0). Next, the fragment was cut with Mlul, and a fragment of about 730 bp was purified. This fragment is the EGFP gene DNA. This fragment was cut with Xhol, blunt-ended in the same manner as described above, and inserted into a pCI vector (Promega) cut with Mlul.
- the plasmid into which EGFP was inserted was cut with Mlul / Xbal, and pCIBoNT.Adl Neurotoxin gene DNA fragments obtained by cutting pCIBoNT.Bdl and pCIBoNT.Edl with Mlul / Xbal were introduced.
- BoNT.Cl and TeNT were inserted into the vector twice, since there were two Xbal sites in the gene DNA.
- the pTREBoNT. Cldl and pTRETeNTdl vectors were cut with Mlul / Xbal, and fragments of about 300 bp and about 90 bp were purified, respectively. This fragment was inserted into the Mlul / Xbal restriction enzyme site of the expression vector into which EGFP was inserted. The resulting plasmid was further cut with Xbal to purify a fragment of about 1200p. This fragment was inserted into the Xbal restriction enzyme site of the expression vector into which a part of the toxin gene prepared above was inserted. In this ligation, the Xbal fragment of about 1200 bp- can be ligated in each of two possible directions. Clones in the correct direction were selected by analyzing the nucleotide sequence.
- Syntaxin 1A gene DNA was obtained from mouse brain RNA by RT-PCR. Based on the principle of the AGPC (Acid Guanidinium-Phenol-Chloroform) method from the brain tissue of C57BL6 mice (Chomczynski, P., and Sacchi, N., (1987) Anal.Biochem., 162, 162, 156-159), Total RNA was purified using RNAzol B (manufactured by TEL-TEST).
- AGPC Acid Guanidinium-Phenol-Chloroform
- CDNA was synthesized from the purified mouse brain total RNA using the Oligo dT—Adaptor Primer and AMV reverse transcriptase XL TM attached to RNA LA PCR Kit (AMV) Ver. 1.1 (TAKARA). Reverse transcription reaction).
- MYK201 as a forward primer for adding a Mlul restriction enzyme site and a Kozak sequence from the 5 'side to the 5' upstream region of the ATg translation initiation sequence of the Syntaxin 1A gene
- PCR was performed using MYK201 and MYK202 as primers, the amplified approximately 900 bp fragment was purified, subcloned into pCR2.1 vector (manufactured by Invitrogen), and did.
- pCISYN An expression vector into which Syntaxin 1A gene DNA was inserted: pCISYN was prepared.
- SNAP25A gene DNA was obtained from mouse brain RNA by RT-PCR as in (3-1) above.
- the primers for PCR were a forward primer for adding a Mlul restriction enzyme site and a Kozak sequence from the 5 'side to the 5' upstream region of the ATg translation initiation sequence of the SNAP25A gene: MYK205 (SEQ ID NO: 38) and SNAP25A MYK208 (SEQ ID NO: 39), a reverse primer for adding a Sail restriction enzyme site to the 3 'downstream region of the TAA translation termination sequence of the gene, was produced by request of Hokkaido system Science.
- the primer sequence the registered sequence of GenBank Accession No .: M22012 was referred to.
- PCR was performed using the cDNA prepared in (3-1) above as type I, MYK205 and MYK208 as primers, and the amplified fragment of about 640 bp was purified and placed in the pCR2.1 vector (Invitrogen). Subcloning was performed to obtain a pCRSNAP25A vector.
- the pCRSNAP25A vector is cleaved with Mlul / Sall, a DNA fragment of about 640 bp is purified, and ligated to the Mlul / Sall restriction enzyme site of a pCI vector (Promega), and the SNAP25A gene DNA is inserted into the expression vector.
- PCISNAP25A was prepared.
- VAMP2 gene DNA was obtained from mouse brain RNA by RT-PCR in the same manner as in (3-1) above.
- PCR primers are used to add a Mlul restriction enzyme site and a Kozak sequence from the 5 'side to the 5, upstream region of the ATg translation initiation sequence of the VAMP2 gene.
- the registered sequence of GenBank Accession No .: U60150 was referred to.
- PCR was performed using the cDNA prepared in (3-1) above as type I, MYK212 and MYK213 as primers, and the amplified fragment of about 380 bp was purified and placed in the pCR2.1 vector (Invitrogen). It was subcloned into the pCRVAMP2 vector.
- the pCRVAMP2 vector one cut with Mlul / Sall was purified a DNA fragment of about 380 bp, and Raigeshiyon the Mlul / Sall restriction sites of P CI vector (Promega Corp.), were inserted V ⁇ AMP2 gene DNA expression base Kuta one: to prepare a P CIVAMP2.
- C0S7 cells Cultured cell line C0S7 cells (ATCC: CRL1651) derived from the kidney of African green monkeys were cultured in DMEM (Dulbecco's Modified Eagle Medium) medium (Life Technologies) containing 10% fetal calf serum (Clontech). in 10% C0 2, 37 ° C the cells were cultured. SuperFect reagent (QIAGEN) was used for gene transfer into cultured cells. For gene transfer, the C0S7 cells cultured as described above were plated on a 6-well culture plate at a cell density of 5 ⁇ 10 5 cells / well, and cultured for 24 hours under the same conditions.
- DMEM Dynabecco's Modified Eagle Medium
- fetal calf serum Clontech
- Example 4 The toxin protein fusion protein expression vector prepared in (2) and (3) and the toxin substrate protein expression vector were mixed at an appropriate ratio.
- the total amount of DNA used was 2.4 ug / well, and the volume was adjusted to 100 ⁇ l by adding serum-free DMEM.
- 14.4 ⁇ l of SuperFect reagent was added, mixed well, and incubated at room temperature for 5 to 10 minutes.
- 600 // I of DMEM containing 10% serum was added to prepare a transfection solution.
- a total of 714.4 ⁇ l of the gene transfer solution was added per well, and the cells were cultured at 37 ° C. Three hours later, the medium was removed, and 2ral of DMEM containing 10% serum was added, and the culture was continued for 48 hours.
- the following six combinations were used for the expression vectors used to co-express the toxin protein and the toxin action target protein.
- lX Tris-Glycine sample buffer 100 mM Tris (pH 6.8), 2% SDS, 10% glycerol, 0.002% bromophenol blue, 1% beta menolecaptoethanol.
- the blotted filter is immersed in 100% methanol and washed with PBS.
- PBST containing 1% skim milk (Yukijirushi) (0.1% Tween-20) is used.
- PBS containing PBS for 30 minutes at room temperature.
- a chemiluminescence reaction was performed with ECL-plus (manufactured by Amersham Pharmacia Biotech), and exposure was performed on an X-ray film: Hyperfilm-MP (manufactured by Amersham Pharmacia Biotech).
- the antibody used and its dilution ratio were as follows:
- the primary antibody was an anti-human Syntaxin monoclonal antibody: Clone SP6 (Upstate Biotechnology: Catalog # 05-397), l / ig // x HRP-conjugated goat anti-mouse IgG antibody (manufactured by Santa Cruz Biotechnology: Catalog SC-2005) was diluted 5000 times and used as the secondary antibody.
- SNAP25A For detection of SNAP25A, goat anti-human (rat) SNAP25A polyclonal antibody (Santa Cruz Biotechnology: Catalog SC-7'538, SC-7539) 4 ⁇ g / ⁇ 1 was used as the primary antibody at 8,000 HRP-conjugated camel anti-goat IgG antibody (manufactured by Santa Cruz Biotechnology: Catalog SC-2020) was diluted 5000-fold and used as a secondary antibody.
- a heron anti-rat (mouse) VAMP2 polyclonal antibody (provided by Dr. Masami Takahashi of Mitsubishi Chemical Life Science Laboratory, Inc.) diluted 250-fold as the primary antibody was used.
- an HRP-conjugated goat IgG antibody (manufactured by Santa Cruz Biotechnology: Catalog SC-2004) was used after diluting 5000-fold.
- Figure 5 shows the results of analysis of the proteins expressed in these DNA-transfected cells.
- the GABA A a6-rtTA gene-transferred mouse is a genetically modified mouse intended to specifically express the rtTA gene in cerebellar granule cells. Expression analysis was performed to confirm whether the rtTA gene was indeed specifically expressed in cerebellar granule cells in the prepared GABA A a6-rtTA gene transfected mouse.
- Example 3 8 lines (line numbers 612, 613, 615, 620, 621, 626, 628, 606) of the independent 10 lines identified as GABA A a6_rtTA transgenic mice and wild-type mice The expression analysis of the rtTA gene was performed by mRNA Northern blot analysis.
- RNAzol TM B reagent TEL-TEST
- Total RNA was purified by using it according to the procedure manual. Heart, liver, kidney, muscle, and small intestine tissues were also removed, and total RNA was purified as in brain tissue.
- mRNA was purified from total RNA by using Oligotex TM -dT30 Super> reagent (JSR, Roche Diagnostics) according to the attached protocol. Purified raRNA is available at 260 nm, 230 nm, 280 nm, The concentration and purity were determined by measuring the absorbance at a wavelength of 320 nm.
- a solution containing 20 mM (IX) MOPS buffer (pH 7.0) and 3.7% formaldehyde as the electrophoresis buffer was used, and a 1% agarose denaturing gel (final concentration 1% in electrophoresis buffer) was used.
- 0.5 ⁇ g / well of the purified mRNA was loaded at 100 V for about 1 hour.
- the gel was treated with 50 mM sodium hydroxide, 10 raM sodium chloride, neutralized with 0.1 M Tris buffer (pH 7.5), equilibrated with 20 X SSC, and then blocked with 20 X SSC.
- Positively charged nylon film (Roche's Diagnostics Co., Ltd.) was used for plotting by the capillary method using a coating solution.
- the membrane was air-dried, and the mRNA transferred to the membrane was fixed by ultraviolet irradiation (120 mj / cm 2 ) (UV Stratalinker TM: manufactured by Stratagene).
- the molecular weight can be determined using 0.24-9.5 kb RNA Ladder (Life Technologies) or DIG-labeled (0.3-6.9 kb) RNA marker (Roche Diagnostics). I went using it.
- a probe for detecting the expression of the rtTA gene was prepared as follows. EcoRI restriction site (g / AATTC) on the 5 'side of the 648b gene fragment from C at base 367 to g at 1014, where A is the ATg of the start codon of the rtTA gene.
- the Hind III restriction enzyme site (A / AgCTT) sequence was added to the 3rd and 3rd sides by PCR, and subcloned into the EcoRI / HindIII region of the pGEM-4Z vector (Promega), and pGEMrtTA (367-1014) Vectors were prepared.
- the pGEMrtTA (369-1014) vector is linearized with EcoRI, purified, and used with the T7 RNA polymerase included in the DIG RNA Labeling Kit (Roche Diagnostics) according to the attached protocol. Digoxigenin-labeled rtTA gene A chisense RNA probe was prepared.
- a probe for detecting the expression of the ⁇ -actin gene was prepared as follows. Oligonucleotide DNA having an antisense sequence for the 70 nucleotide region from 438A to 507T of the nucleotide sequence registered in GenBank Accession No .: 03672 was synthesized. Using a DIG oligonucleotide tiling kit (manufactured by Roche Diagnostics), an antisense oligonucleotide DNA probe for the digoxigenin-labeled / 3-actin gene was prepared according to the attached protocol.
- a probe for detecting the expression of the darichelaldehyde triphosphate dehydrogenase gene was prepared as follows. GenBank Accession No .: Oligonucleotide DNA having an antisense sequence for the 70 base region from 663 C to 732 T of the base sequence registered in M32599 was synthesized. Using the DIG Oligonucleotide Tiling Kit, an antisense oligonucleotide DNA probe of the digoxigenin-labeled dalycelaldehyde triphosphate dehydrogenase gene was prepared according to the attached protocol.
- the blot membrane prepared in Example 5 (2) was prehybridized with a DIG easyhive solution (Roche Diagnostics), and then a digoxigenin-labeled probe for the gene whose expression was to be detected was added. Was carried out.
- the hybridization temperature was 68 ° C when using an RNA probe, and 42 ° C when using an oligonucleotide DNA probe.
- rtTA gene expression signal was approximately 3 Kb, which is the sum of the etason 1 to 8 of GABA A a6 gene, IRES, rtTA gene, and polyA sequence size. It is expected that it will be detected nearby.
- the expression signal of the rtTA gene was detected in the cerebellum (arrow in FIG. 6), but not in other brain tissue regions other than the cerebellum.
- the expression signal of the rtTA gene was not detected at all from the cerebellum and brain tissue regions other than the cerebellum.
- the rtTA gene expression signal was the strongest at line 620.
- mRNA blotting analysis of rtTA gene expression was also performed on organs other than the brain (heart, liver, kidney, muscle, and small intestine). I did it.
- mRNA expression of the rtTA transgene was specific to the cerebellum, and was not detected at all in brain regions other than the cerebellum or in organs other than the brain. (Fig. 6)
- Example 5 the GABA A a6- rtTA transgenic mice that rtTA gene was expressed specifically in the cerebellum was confirmed by m RA Northern blot analysis. Next, in order to examine in which cells of the cerebellum the rtTA gene was expressed, in situ hybridization was performed by angular analysis.
- in situ hybridization analysis was performed on wild type mice and six lines of the GABA A o 6- rtTA transgenic mice generated (line number 612, 613, 620, 621, 626, 628) (negative control).
- mice were anesthetized with getyl ether, and the brain was quickly removed and divided into appropriate sizes.
- the brain tissues were embedded in Tissue-Tek R OCT- Compound (manufactured by Sakura Finetechnical Co.), the isopentane cooled with dry ice and rapidly frozen. Frozen tissue blocks are stored at 120 to 180 ° C. During sectioning, cryostats with a temperature of -20 to 114 ° C are used to cut sections of 10 ⁇ in thickness. It was prepared and affixed to MAS coated slide glass (Matsunami).
- the sections affixed to the slide glass were fixed with a fixing solution (4% paraformaldehyde, 0.1 PBS), and acetylation was added to 0.25% acetic anhydride, 0.9% in order to suppress nonspecific reaction of the probe.
- % Sodium chloride, 0.1 M triethanolamine [ PH 8.0] solution After rinsing with PBS, degreased and dehydrated with 70%, 80%, 90%, 100%, and 100% ethanol series, dried at 55 ° C for 15 to 60 minutes, and dried for one hour until hybridization. Stored at 80 ° C.
- 0.5 ⁇ g of type I DNA was added to IX Transcription buffer (40 raM Tris [ ⁇ 8.0], 50 mM Sodium chloride, 8 mM magnesium chloride, 2 mM spermidine), 500 ⁇ ATP, 500 / iM GTP, 500 ⁇ TTP, 10 mM DTT, 11.5 U / ⁇ 1 RNase inhibitor (Takarasha), 20 ⁇ M S-CTP, 20 ⁇ [a- 35 S] CTP, ⁇ 7 polymerase (manufactured by Stratagene) at 37 ° C,!
- the reaction was performed for about 2 hours to prepare an antisense RNA probe of the rtTA gene labeled with [a- 35 S] CTP.
- an antisense RNA probe of the rtTA gene labeled with [a- 35 S] CTP.
- DNase Promega
- unreacted mononucleotides and DNA degradation products are removed by Sephadex G-50 (Amersham Pharmacia) and ethanol precipitation, and then 0.5 to 1.0%.
- X 10 8 cpra / ml such that the hybridization buffer (50% Horumuami de, 2 X SSC, 100 mM tris [pH 7.4], 10% dextran sulfate sodium, 0.2% SDS, 1 X Denha belt solution) To prepare a hybridization probe solution.
- a hybridization probe solution of an antisense RNA probe for the rtTA gene was added to the slice on the slide glass dried at 55 ° C, and a hybridization reaction was performed at 60 ° C. 1 After 2 to 24 hours, wash with 2 X SSC, 10 mM in 3 mM mercaptoethanol solution, wash with 60, 2 / ⁇ / 11111 ⁇ 356, 101113 ⁇ 41 Tris [pH 8.0], 1 raM EDTA, 500 mM chloride Unreacted RNA probe was degraded at 37: in the sodium solution. Furthermore, after washing at 60 ° C. in a ⁇ SSC, 10 mM / 3 mercaptoethanol solution, dehydration and drying were performed in an ethanol series.
- the expression signal of the rtTA gene was detected by radioautography using BAS5000 (manufactured by Fuji Film Co., Ltd.), Hyperfilra-i3Max (manufactured by Amersham Pharmacia), and emulsion (NTB3: manufactured by Kodak Company).
- GABA A a6-rtTA transgenic in situ hybridization analysis of results tested GABA A a6-rtTA transgenic mice 6 lines all at Mausu, only in cerebellar granule cells expression signals rtTA is detected, the cerebellum It was not detected in Purkinje cells, cells in the molecular layer (the layer where parallel fibers that are axons of granule cells are present), white matter, and cerebellar nuclei. In addition, it contains the lower olive nucleus, which is the origin of the climbing fiber. In other areas of the eye brain, expression signals rtTA were detected intensity of expression signals (Fig.
- mice The treBoNT. Adl, treBoNT. Bdl, treBoNT. Cldl, treBoNT. Edl, and treTeNTdl mice obtained in Example 3 are collectively referred to as treToxin mice.
- GABA A a6-rtTA transgenic mice and treToxin transgenic mice were weaned at about 4 weeks of age and genotyped by genomic Southern blot analysis. If you want to induce neurotoxin protein expression, drink doxycycline (Sigma) in drinking water (2 mg / ml doxycycline, 10% sucrose) and pellet diet
- mice (6 mg / g doxycycline, manufactured by BIO-SERV) and given to mice. If you do not want to induce the expression of the neurotoxin protein, or if you want to stop the induction of the expression, the drinking water and food were changed to those that did not contain doxycycline.
- Example 9 Detection of induction and extinction of neurotoxin protein expression in mice transfected with the GABA, a6-rtTA gene and treToxin gene double transfection gene:
- BIO-RAD DC Protein Assay reagent
- the brain tissue protein lysates subjected to 40 M g protein / lane load to SDS-PAGE, having conducted the blotted to PVDF membranes.
- To detect the expression of neurotoxin protein tag the N-terminal end of neurotoxin protein Using a polyclonal antibody against EGFP (Molecular Probe: Catalog A6455) fused as a primary antibody, using an HRP-conjugated goat anti-Peagle IgG antibody (manufactured by Santa Cruz Biotechnology) as a secondary antibody, and using ECL-plus Was performed. It is expected from the calculated amino acid composition that the expression signal of the neurotoxin protein will be detected at around 84 KDa.
- FIG. 8 shows the results of preparing a protein lysate from the cerebellum of a heavy transgenic mouse (Genotype: + / +, D0X: +), and examining the induction of neurotoxin protein expression. An expression signal was detected near 84 KDa only in the cerebellum of the double transgenic mouse (Genotype: + / +, D0X: +) to which doxycycline was administered for one week (arrow in FIG. 8).
- Example 9 the induction of neurotoxin protein expression was analyzed by Western blotting.
- a double transgenic mouse (Genotype: + / +, D0X: +) to which doxycycline was administered for one week, Toxin protein expression It was confirmed that it was induced.
- Toxin protein expression It was confirmed that it was induced.
- the mouse is anesthetized with getyl ether, thoracotomy, and from the left ventricle of the heart, using a peristaltic pump for 2 minutes with PBS and a fixative (phosphate buffer [pH 7.3] containing 4% paraformaldehyde) for 1 min. Perfused at a flow rate of 10 ml / miri for 0 to 15 minutes. After excision of the brain, it was divided into appropriate sizes, immersed in fixative, and permeabilized at 4 ° C for 12 to 48 hours. The fixed brain tissue was transferred to PBS containing 30% sucrose and 0.05% sodium azide, and cryoprotection was performed at 4 ° C to suppress tissue damage caused by ice crystallization that occurs during tissue freezing.
- a fixative phosphate buffer [pH 7.3] containing 4% paraformaldehyde
- Sections were washed with washing buffer (PBS containing 0. 1% Tri ton R X- 100), antibodies diluent for (0. 1% Triton R X- 100 , 2% normal turbocharger formic serum (Vector Lab Incubate for 1 hour at room temperature in PBS containing 0.05% sodium azide) to block nonspecific antigen-antibody reactions.
- washing buffer PBS containing 0. 1% Tri ton R X- 100
- antibodies diluent for (0. 1% Triton R X- 100
- 2% normal turbocharger formic serum Vector Lab Incubate for 1 hour at room temperature in PBS containing 0.05% sodium azide
- Primary antibodies include anti-EGFP ⁇ sagi polyclonal antibody (Molecular Probe: Catalog A6455), anti-CalbindinD-28K mouse monoclonal antibody (Sigma: Catalog C9848), anti-Parvalbumin mouse monoclonal antibody (Sigma: Catalog P3088) was diluted with an antibody diluent.
- Alexa Fluor R 488 goat anti-money IgG (H + L) antibody and Alexa Fluor "594 goat anti-mouse IgG (H + L) antibody were used as secondary antibodies in the antibody diluent.
- the sections were attached to slide glass, sealed with VectaShield (Vector Lab.), And observed with a laser scanning microscope system (LSM 510 ETA: Zeiss).
- Fig. 9 A, B, C, and D are wild-type mice (6 weeks old) treated with doxycycline for 1 week
- Fig. 9 A Genotype:-/-, D0X: +
- Fig. 9B Genotype: + / +, D0X:-
- double transgenic mice (6 weeks old) to which doxycycline was administered for 1 week
- Fig. 9C Genotype: + / +, D0X: +)
- doxycycline was administered for 2 weeks, and then doxycycline was removed for 3 weeks (10 weeks old).
- Genotype: + / +, D0X This figure shows the results obtained by performing the primary antibody reaction using the anti-EGFP antibody and the secondary antibody and subsequent antibodies using the ABC method on the cerebellar slices (+ ⁇ -).
- Fig. 9C Genotype: + / +, D0X: +
- the molecular layer of the cerebellum parallel fibers that are axons of granule cells have high density
- Signal in the granule cell layer layer in which the cell body of granule cells exists at high density
- a reaction signal with the anti-EGFP antibody indicating the presence of neurotoxin protein was detected in the purkinje cell layer. There was no response signal, so it was white and it turned out that there was no neurotoxin protein.
- FIGS. 9A, B, and D no reaction signal was detected.
- the red fluorescence signal ( Figure 9F) indicating the presence of Calbindin D-28K is detected in Purkinje cells
- the red fluorescence signal (Fig. 9I) indicating the presence of Parvalbumin is detected in Purkinje cells, sterate cells, and basket cells.
- the respective fluorescent signals do not overlap. It was confirmed that they were completely separated. This indicates that the inducible expression of the neurotoxin protein is restricted to cerebellar granule cells and not in Purkinje cells, stellate cells, or basket cells.
- the expression signal of the rtTA gene is restricted to the granule cells existing in the granule cell layer of the cerebellum, and is expressed in white matter, Purkinje cell layer, and molecular layer. was not detected.
- the expression signal of the neurotoxin protein was detected in the granular cell layer (the layer where the cell bodies of the granular cells exist at a high density) and the molecular layer according to the results of Example 10.
- the molecular layer is composed of parallel fibers, axons of granule cells that exist at an overwhelming density, dendrites of Purkinje cells, stellate cells, basket cells, and the like.
- the expression of neurotoxin protein is not observed in dendritic protrusion of Purkinje cells, stellate cells, and basket cells (FIG. 9 E, F, G, H, I, J). Therefore, the expression signal of the neurotoxin protein detected in the molecular layer is transmitted to the parallel fibers that are the axons of the granule cells. It is considered to be derived.
- mice transfected with the GABA A a6-rtTA gene and treToxin gene the rtTA gene is specifically expressed in cerebellar granule cells, and the expression of neurotoxin protein is reduced in cerebellar granule cell by administration of doxycycline.
- the neurotoxin protein was transported from the cell body of cerebellar granule cells to the axon parallel fiber (the site of action of the neurotoxin protein), and was widely distributed.
- the GABA A a6-rtTA gene and the treToxin gene double transgenic mice exhibited a locomotor behavior similar to that of wild-type mice during walking.
- the wild-type mouse has a disorder of coordination, such as slowing down, wobbling, walking on the stomach, and walking. Phenotypes that are distinguished from.
- wild-type mice mice having only the transgene GABA A a6-rtTA gene, the Dokishisaitarin in mice with only treToxin gene as a transgene throw Even when given, the above phenotype was not shown, and it was indistinguishable from wild-type mice not administered with doxycycline.
- GABA A oc6- rtTA gene in treToxin gene double transgenic mice, by 1-2 weeks of doxycycline (neurotoxin protein derived specifically expressed in cerebellar granule cells), the incoordination After eliciting the phenotype of Drosophila, the patient was returned to the condition without doxycycline and observed 3 weeks later (the neurotoxin protein was no longer present in the cerebellar granule cells). It returned to a state indistinguishable from wild-type mice not treated with doxycitalin.
- the phenotype of incoordination is the individual mouse, GABA A a6- rtTA gene, it two of TreToxin gene present simple or not for side effects of doxycycline, GABA A a6- rtTA gene, TreToxin It was concluded that the gene transfection mouse was caused by the expression and disappearance of neurotoxin protein induced in cerebellar granule cells by administration and non-administration of doxycitalin.
- the mouse was mounted on a rod (UG0 BASILE) moving at a rotation speed of 15 r.p.m. (rotation / min) and 30 r.p.m.
- the mouse performs a cooperative walking movement in accordance with the rotation of the mouth to prevent the mouse from falling from the mouth.
- a mouse that cannot keep up with the rotation of the mouth (coordination is impaired)
- the force that falls from the rod The clinging to the rotating rod, the mouse rotates together.
- the maximum time for one test was 60 seconds. Even if the test rod could stay on the rotating rod for more than 60 seconds, the test was stopped and the score was set to 60 seconds.
- mice wild-type mice (-/-), only the GABA A a6-rtTA gene Introduced mice (+ / _), transgenic mice with only treToxin gene (-/ +), GABA A a6-rtTA gene, treToxin gene Double transgenic mice (+ / +) were placed on a rotating rod ( ( Figure 10).
- Figure 10 On the first day of the test, the mouse is unfamiliar with it, and cannot perform a cooperative walking movement on the rotating rod, and rotates together as if it were wound around the rotating mouth. Or fall and the residence time on the rods was short for all genotypes.
- mice of all genotypes are good at performing cooperative walking on a rotating rod, and the residence time on the rod is long.
- Natsuta when doxycycline was administered to mice transfected with the GABA A a6-rtTA gene and the treToxin gene, the dwelling time on rods decreased, indicating a phenotype that impaired coordinated walking. became.
- mice transfected with only GABA A a6-rtTA gene (+/-) mice transfected with only treToxin gene (+/-), the same level as before administration was obtained. The results showed that the dwell time was shown and that the coordinated walking movement was not impaired.
- mice were placed on wooden rods 30 mm wide, 15 mm, and 6 mm wide lying 40 cm above the floor.
- the mouse dislikes high places, and is a safety cage 60 era away from the place where it was placed (a paper box with a floor width of 20 cm, a floor depth of 16.5 cm, and a height of 10.5 era.
- the entrance is 8 cras wide and 10.5 cm high, and the floor of the safety cage is walked with a stick to escape into the wooden flooring chips normally used for breeding cages.
- the mouse can cross the rod and escape to the safety cage if it can maintain a good balance of physical properties and perform a coordinated walking movement.
- the graph in Fig. 11 shows the ratio of the number of “Fail” times to the number of times tested with the bar width changed to 30 ram, 15 mm, and 6 mm on the vertical axis.
- a width of 30 mm crossing over this rod was not too difficult for any genotyped mouse and no "Fail” was seen.
- 15 mm and 6 mm mice that failed were observed.
- the width was 6 mm, doxycycline-administered GABA A a6-rtTA gene, treToxin
- the transgenic mice have failed 80% or more in the test. This failure rate is a significant increase compared to around 10% for other genotypes treated with doxycycline.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Biomedical Technology (AREA)
- Chemical & Material Sciences (AREA)
- Molecular Biology (AREA)
- Biotechnology (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Zoology (AREA)
- Physics & Mathematics (AREA)
- Microbiology (AREA)
- General Health & Medical Sciences (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Plant Pathology (AREA)
- Hematology (AREA)
- Immunology (AREA)
- Urology & Nephrology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Cell Biology (AREA)
- Food Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- Analytical Chemistry (AREA)
- General Physics & Mathematics (AREA)
- Pathology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Description
Claims
Priority Applications (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US10/474,529 US20040234974A1 (en) | 2001-04-09 | 2002-04-09 | Gene expression controlling unit and utilization thereof |
EP02713310A EP1386968A4 (en) | 2001-04-09 | 2002-04-09 | UNIT TO CONTROL AND USE GENE EXPRESSION |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2001109445 | 2001-04-09 | ||
JP2001-109445 | 2002-04-09 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2002086131A1 true WO2002086131A1 (fr) | 2002-10-31 |
Family
ID=18961385
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2002/003537 WO2002086131A1 (fr) | 2001-04-09 | 2002-04-09 | Unite de regulation de l'expression genetique et utilisation de cette unite |
Country Status (3)
Country | Link |
---|---|
US (1) | US20040234974A1 (ja) |
EP (1) | EP1386968A4 (ja) |
WO (1) | WO2002086131A1 (ja) |
Families Citing this family (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
AU2012203048A1 (en) * | 2011-05-24 | 2012-12-13 | Agency For Science, Technology And Research | IRES mediated multicistronic vectors |
DE202018006693U1 (de) * | 2017-03-14 | 2022-03-08 | Nanotag Biotechnologies Gmbh | Target-Detektion mit einem monovalenten Antikörper |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1995032738A1 (en) * | 1994-05-31 | 1995-12-07 | Allergan, Inc. | Modification of clostridial toxins for use as transport proteins |
WO1997017369A2 (en) * | 1995-11-09 | 1997-05-15 | Trustees Of Boston University | Dna comprising a neuron-specific transcriptional promoter and its use in a gene therapy vector |
JP2000316583A (ja) * | 1999-05-14 | 2000-11-21 | Eisai Co Ltd | 脳で特異的に発現するベクター |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5912411A (en) * | 1993-06-14 | 1999-06-15 | University Of Heidelberg | Mice transgenic for a tetracycline-inducible transcriptional activator |
GB2337519A (en) * | 1998-05-19 | 1999-11-24 | Univ Bristol | Transgenic mammal comprising specific means for regulation of transgene expression |
CN1352694A (zh) * | 1998-11-09 | 2002-06-05 | 阿文蒂斯药物股份有限公司 | 转基因表达调控的新系统 |
-
2002
- 2002-04-09 EP EP02713310A patent/EP1386968A4/en not_active Withdrawn
- 2002-04-09 US US10/474,529 patent/US20040234974A1/en not_active Abandoned
- 2002-04-09 WO PCT/JP2002/003537 patent/WO2002086131A1/ja not_active Application Discontinuation
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1995032738A1 (en) * | 1994-05-31 | 1995-12-07 | Allergan, Inc. | Modification of clostridial toxins for use as transport proteins |
WO1997017369A2 (en) * | 1995-11-09 | 1997-05-15 | Trustees Of Boston University | Dna comprising a neuron-specific transcriptional promoter and its use in a gene therapy vector |
JP2000316583A (ja) * | 1999-05-14 | 2000-11-21 | Eisai Co Ltd | 脳で特異的に発現するベクター |
Non-Patent Citations (4)
Title |
---|
RIBAS A.V. ET AL.: "High-level expression of tetanus toxin fragment C-thioredoxin fusion protein in escherichia coli", BIOTECHNOL. APPL. BIOCHEM., vol. 260, no. PT. 2, 2000, pages 91 - 94, XP000978729 * |
See also references of EP1386968A4 * |
SOULIER S. ET AL.: "Use of doxycycline-controlled gene expression to reversibly alter milk-protein composition in transgenic mice", EUR. J. BIOCHEM., vol. 260, no. 2, 1999, pages 533 - 539, XP002952504 * |
YAMAMOTO A. ET AL.: "Reversal of neuropathology and motor dysfunction in a conditional model of Huntington's disease", CELL, vol. 101, no. 1, 31 March 2000 (2000-03-31), pages 57 - 66, XP002952503 * |
Also Published As
Publication number | Publication date |
---|---|
EP1386968A1 (en) | 2004-02-04 |
EP1386968A4 (en) | 2004-12-22 |
US20040234974A1 (en) | 2004-11-25 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Kuang et al. | Merosin-deficient congenital muscular dystrophy. Partial genetic correction in two mouse models. | |
Taniguchi et al. | Disruption of semaphorin III/D gene causes severe abnormality in peripheral nerve projection | |
JP6443811B2 (ja) | Mrap2ノックアウト | |
US9161520B2 (en) | Transgenic animal expressing Alzheimer's tau protein | |
Moeller et al. | Evaluation of a New Tool for Exploring Podocyte Biology: Mouse: Nphs1: 5′ Flanking Region Drives LacZ Expression in Podocytes | |
JP2014507136A (ja) | NaV1.7ノックアウトマウスとそれらの使用 | |
JP6593595B2 (ja) | Atp可視化動物およびその用途 | |
US10765093B2 (en) | Humanized transgenic animal | |
Eulenburg et al. | GlyT1 determines the glycinergic phenotype of amacrine cells in the mouse retina | |
US10568974B2 (en) | Animal models of atherosclerosis | |
US8487087B2 (en) | Model animal in which state of disease condition is observable in real time, gene construct for achieving the same and use of the same | |
JP2003524420A (ja) | 単一の性の子孫を生産する哺乳動物の生産 | |
US8383880B2 (en) | Infertility control of genetically modified fish | |
US6630612B2 (en) | 5-HT3 receptor assay using transgenic mammal | |
WO2002086131A1 (fr) | Unite de regulation de l'expression genetique et utilisation de cette unite | |
US20040177389A1 (en) | Methods | |
Lahiri et al. | Nephropathy and defective spermatogenesis in mice transgenic for a single isoform of the Wilms' tumour suppressor protein, WT1− KTS, together with one disrupted Wt1 Allele | |
JP2003009888A (ja) | 遺伝子発現制御ユニットおよびその利用 | |
JP2002058486A (ja) | エストロゲン高感受性メダカ | |
JP2002142610A (ja) | Alsモデルラット | |
US7445904B2 (en) | Cysteine string protein and its role in neurodegenerative diseases | |
JP2002518056A (ja) | アッセイにおけるPeg3遺伝子の使用、並びに肥満、体温調節および行動障害に関連する産物 | |
Kim | Improvement and establishment of the tTA-dependent inducible system in the mouse brain | |
JPWO2018012497A1 (ja) | 疾患モデル動物および疾患治療剤 | |
JPWO2008062904A1 (ja) | トランスジーンの安定的発現を可能にする方法 |
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 ES FI GB GD GE GH GM HR HU ID IL IN IS KE KG KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A1 Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN 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: 2002713310 Country of ref document: EP |
|
WWP | Wipo information: published in national office |
Ref document number: 2002713310 Country of ref document: EP |
|
REG | Reference to national code |
Ref country code: DE Ref legal event code: 8642 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 10474529 Country of ref document: US |
|
WWW | Wipo information: withdrawn in national office |
Ref document number: 2002713310 Country of ref document: EP |