CN114762733A - Extracochlear hair cell specific cis-form regulatory element and application thereof - Google Patents
Extracochlear hair cell specific cis-form regulatory element and application thereof Download PDFInfo
- Publication number
- CN114762733A CN114762733A CN202110055458.7A CN202110055458A CN114762733A CN 114762733 A CN114762733 A CN 114762733A CN 202110055458 A CN202110055458 A CN 202110055458A CN 114762733 A CN114762733 A CN 114762733A
- Authority
- CN
- China
- Prior art keywords
- prestin
- expression
- sequence
- regulatory element
- cochlear
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
- 210000002768 hair cell Anatomy 0.000 title claims abstract description 73
- 230000001105 regulatory effect Effects 0.000 title claims abstract description 11
- 108050001617 Prestin Proteins 0.000 claims abstract description 100
- 102000011383 Prestin Human genes 0.000 claims abstract description 100
- 108090000623 proteins and genes Proteins 0.000 claims abstract description 61
- 108091062157 Cis-regulatory element Proteins 0.000 claims abstract description 40
- 210000000114 cochlear outer hair cell Anatomy 0.000 claims abstract description 39
- 239000012636 effector Substances 0.000 claims abstract description 31
- 208000016354 hearing loss disease Diseases 0.000 claims abstract description 31
- 238000000034 method Methods 0.000 claims abstract description 26
- 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 claims abstract description 10
- 125000003275 alpha amino acid group Chemical group 0.000 claims abstract 2
- 230000014509 gene expression Effects 0.000 claims description 71
- 210000004027 cell Anatomy 0.000 claims description 58
- 239000013604 expression vector Substances 0.000 claims description 30
- 210000003477 cochlea Anatomy 0.000 claims description 24
- 239000008194 pharmaceutical composition Substances 0.000 claims description 20
- 239000004480 active ingredient Substances 0.000 claims description 19
- 239000002773 nucleotide Substances 0.000 claims description 18
- 125000003729 nucleotide group Chemical group 0.000 claims description 15
- 239000000203 mixture Substances 0.000 claims description 14
- 230000007850 degeneration Effects 0.000 claims description 13
- 230000006378 damage Effects 0.000 claims description 11
- 108091026890 Coding region Proteins 0.000 claims description 7
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 7
- 230000001172 regenerating effect Effects 0.000 claims description 7
- 239000003937 drug carrier Substances 0.000 claims description 6
- 239000003814 drug Substances 0.000 claims description 5
- 238000002360 preparation method Methods 0.000 claims description 5
- 239000000126 substance Substances 0.000 claims description 4
- 108020005345 3' Untranslated Regions Proteins 0.000 claims description 3
- 108020003589 5' Untranslated Regions Proteins 0.000 claims description 3
- 150000001875 compounds Chemical class 0.000 claims description 3
- 238000009472 formulation Methods 0.000 claims description 3
- 239000000825 pharmaceutical preparation Substances 0.000 claims description 3
- 230000002265 prevention Effects 0.000 claims description 3
- 230000002463 transducing effect Effects 0.000 claims description 3
- 230000008439 repair process Effects 0.000 claims description 2
- 230000008929 regeneration Effects 0.000 claims 1
- 238000011069 regeneration method Methods 0.000 claims 1
- 230000035772 mutation Effects 0.000 abstract description 8
- 241000699670 Mus sp. Species 0.000 description 58
- 241000699666 Mus <mouse, genus> Species 0.000 description 47
- 239000012634 fragment Substances 0.000 description 36
- 108020004414 DNA Proteins 0.000 description 32
- 239000013598 vector Substances 0.000 description 27
- 241000282414 Homo sapiens Species 0.000 description 24
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 22
- 241000700605 Viruses Species 0.000 description 21
- 238000012217 deletion Methods 0.000 description 21
- 230000037430 deletion Effects 0.000 description 21
- 108020004999 messenger RNA Proteins 0.000 description 17
- 210000001519 tissue Anatomy 0.000 description 16
- 239000013603 viral vector Substances 0.000 description 14
- 108091027544 Subgenomic mRNA Proteins 0.000 description 12
- 108091033409 CRISPR Proteins 0.000 description 11
- 241000702421 Dependoparvovirus Species 0.000 description 11
- 238000004458 analytical method Methods 0.000 description 11
- 239000003623 enhancer Substances 0.000 description 11
- 238000002474 experimental method Methods 0.000 description 11
- 230000006870 function Effects 0.000 description 10
- 101001094051 Homo sapiens Prestin Proteins 0.000 description 9
- 150000007523 nucleic acids Chemical group 0.000 description 9
- 108091028043 Nucleic acid sequence Proteins 0.000 description 8
- 102000056771 human SLC26A5 Human genes 0.000 description 8
- 230000010076 replication Effects 0.000 description 8
- 239000007788 liquid Substances 0.000 description 7
- 239000000523 sample Substances 0.000 description 7
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 6
- 241000701022 Cytomegalovirus Species 0.000 description 6
- 206010011878 Deafness Diseases 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 238000010186 staining Methods 0.000 description 6
- 238000011830 transgenic mouse model Methods 0.000 description 6
- 239000013607 AAV vector Substances 0.000 description 5
- 102000007469 Actins Human genes 0.000 description 5
- 108010085238 Actins Proteins 0.000 description 5
- 241000287828 Gallus gallus Species 0.000 description 5
- 241000124008 Mammalia Species 0.000 description 5
- 101001037757 Mus musculus Heat shock 70 kDa protein 1A Proteins 0.000 description 5
- 238000011529 RT qPCR Methods 0.000 description 5
- 108091023040 Transcription factor Proteins 0.000 description 5
- 210000003030 auditory receptor cell Anatomy 0.000 description 5
- 210000003027 ear inner Anatomy 0.000 description 5
- 230000000694 effects Effects 0.000 description 5
- 238000001415 gene therapy Methods 0.000 description 5
- 238000009396 hybridization Methods 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 230000001939 inductive effect Effects 0.000 description 5
- 210000000067 inner hair cell Anatomy 0.000 description 5
- 238000003780 insertion Methods 0.000 description 5
- 230000037431 insertion Effects 0.000 description 5
- 210000004379 membrane Anatomy 0.000 description 5
- 239000012528 membrane Substances 0.000 description 5
- 239000013612 plasmid Substances 0.000 description 5
- 238000011160 research Methods 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- 102100035902 Glutamate decarboxylase 1 Human genes 0.000 description 4
- 208000016621 Hearing disease Diseases 0.000 description 4
- 241001465754 Metazoa Species 0.000 description 4
- 241000699660 Mus musculus Species 0.000 description 4
- 101001094049 Mus musculus Prestin Proteins 0.000 description 4
- 102000018210 Recoverin Human genes 0.000 description 4
- 108010076570 Recoverin Proteins 0.000 description 4
- 101150052863 THY1 gene Proteins 0.000 description 4
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 4
- 108700019146 Transgenes Proteins 0.000 description 4
- 239000013504 Triton X-100 Substances 0.000 description 4
- 229920004890 Triton X-100 Polymers 0.000 description 4
- 102000014823 calbindin Human genes 0.000 description 4
- 108060001061 calbindin Proteins 0.000 description 4
- 231100000895 deafness Toxicity 0.000 description 4
- 230000002950 deficient Effects 0.000 description 4
- 235000013601 eggs Nutrition 0.000 description 4
- 210000004602 germ cell Anatomy 0.000 description 4
- 108010024847 glutamate decarboxylase 1 Proteins 0.000 description 4
- 238000011534 incubation Methods 0.000 description 4
- 238000011813 knockout mouse model Methods 0.000 description 4
- 238000007480 sanger sequencing Methods 0.000 description 4
- 241000701161 unidentified adenovirus Species 0.000 description 4
- 230000003612 virological effect Effects 0.000 description 4
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 241000282412 Homo Species 0.000 description 3
- 241000725303 Human immunodeficiency virus Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 108020004518 RNA Probes Proteins 0.000 description 3
- 239000003391 RNA probe Substances 0.000 description 3
- 101001094050 Rattus norvegicus Prestin Proteins 0.000 description 3
- 101150115433 SLC26A5 gene Proteins 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- AUYYCJSJGJYCDS-LBPRGKRZSA-N Thyrolar Chemical class IC1=CC(C[C@H](N)C(O)=O)=CC(I)=C1OC1=CC=C(O)C(I)=C1 AUYYCJSJGJYCDS-LBPRGKRZSA-N 0.000 description 3
- 102000040945 Transcription factor Human genes 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 238000010276 construction Methods 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 239000005090 green fluorescent protein Substances 0.000 description 3
- 238000010166 immunofluorescence Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 238000004519 manufacturing process Methods 0.000 description 3
- 102000004169 proteins and genes Human genes 0.000 description 3
- 238000003753 real-time PCR Methods 0.000 description 3
- 108010054624 red fluorescent protein Proteins 0.000 description 3
- 230000035945 sensitivity Effects 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 239000005495 thyroid hormone Substances 0.000 description 3
- 229940036555 thyroid hormone Drugs 0.000 description 3
- 238000010354 CRISPR gene editing Methods 0.000 description 2
- 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 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 2
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 2
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 2
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 2
- TWRXJAOTZQYOKJ-UHFFFAOYSA-L Magnesium chloride Chemical compound [Mg+2].[Cl-].[Cl-] TWRXJAOTZQYOKJ-UHFFFAOYSA-L 0.000 description 2
- 108700026244 Open Reading Frames Proteins 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 238000012408 PCR amplification Methods 0.000 description 2
- RJKFOVLPORLFTN-LEKSSAKUSA-N Progesterone Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)C)[C@@]1(C)CC2 RJKFOVLPORLFTN-LEKSSAKUSA-N 0.000 description 2
- 241000713311 Simian immunodeficiency virus Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 102000008579 Transposases Human genes 0.000 description 2
- 108010020764 Transposases Proteins 0.000 description 2
- 150000001413 amino acids Chemical group 0.000 description 2
- 210000000133 brain stem Anatomy 0.000 description 2
- OSGAYBCDTDRGGQ-UHFFFAOYSA-L calcium sulfate Chemical compound [Ca+2].[O-]S([O-])(=O)=O OSGAYBCDTDRGGQ-UHFFFAOYSA-L 0.000 description 2
- 238000007405 data analysis Methods 0.000 description 2
- QONQRTHLHBTMGP-UHFFFAOYSA-N digitoxigenin Natural products CC12CCC(C3(CCC(O)CC3CC3)C)C3C11OC1CC2C1=CC(=O)OC1 QONQRTHLHBTMGP-UHFFFAOYSA-N 0.000 description 2
- 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 2
- 201000010099 disease Diseases 0.000 description 2
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 2
- 239000006185 dispersion Substances 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 210000000981 epithelium Anatomy 0.000 description 2
- -1 etc.) Polymers 0.000 description 2
- 210000003722 extracellular fluid Anatomy 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 238000001502 gel electrophoresis Methods 0.000 description 2
- 239000011521 glass Substances 0.000 description 2
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 2
- 239000010931 gold Substances 0.000 description 2
- 229910052737 gold Inorganic materials 0.000 description 2
- 230000012010 growth Effects 0.000 description 2
- 231100000888 hearing loss Toxicity 0.000 description 2
- 230000010370 hearing loss Effects 0.000 description 2
- 238000007901 in situ hybridization Methods 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000011065 in-situ storage Methods 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 239000003068 molecular probe Substances 0.000 description 2
- 210000002985 organ of corti Anatomy 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 229920005862 polyol Polymers 0.000 description 2
- 150000003077 polyols Chemical class 0.000 description 2
- 230000003389 potentiating effect Effects 0.000 description 2
- 239000000843 powder Substances 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 238000012216 screening Methods 0.000 description 2
- 230000001953 sensory effect Effects 0.000 description 2
- 238000002864 sequence alignment Methods 0.000 description 2
- 238000012163 sequencing technique Methods 0.000 description 2
- 230000004936 stimulating effect Effects 0.000 description 2
- 230000000638 stimulation Effects 0.000 description 2
- 229960001603 tamoxifen Drugs 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 238000013518 transcription Methods 0.000 description 2
- 230000035897 transcription Effects 0.000 description 2
- 230000026683 transduction Effects 0.000 description 2
- 238000010361 transduction Methods 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 2
- DIGQNXIGRZPYDK-WKSCXVIASA-N (2R)-6-amino-2-[[2-[[(2S)-2-[[2-[[(2R)-2-[[(2S)-2-[[(2R,3S)-2-[[2-[[(2S)-2-[[2-[[(2S)-2-[[(2S)-2-[[(2R)-2-[[(2S,3S)-2-[[(2R)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[2-[[(2S)-2-[[(2R)-2-[[2-[[2-[[2-[(2-amino-1-hydroxyethylidene)amino]-3-carboxy-1-hydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxypropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1,5-dihydroxy-5-iminopentylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxybutylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1,3-dihydroxypropylidene]amino]-1-hydroxyethylidene]amino]-1-hydroxy-3-sulfanylpropylidene]amino]-1-hydroxyethylidene]amino]hexanoic acid Chemical compound C[C@@H]([C@@H](C(=N[C@@H](CS)C(=N[C@@H](C)C(=N[C@@H](CO)C(=NCC(=N[C@@H](CCC(=N)O)C(=NC(CS)C(=N[C@H]([C@H](C)O)C(=N[C@H](CS)C(=N[C@H](CO)C(=NCC(=N[C@H](CS)C(=NCC(=N[C@H](CCCCN)C(=O)O)O)O)O)O)O)O)O)O)O)O)O)O)O)N=C([C@H](CS)N=C([C@H](CO)N=C([C@H](CO)N=C([C@H](C)N=C(CN=C([C@H](CO)N=C([C@H](CS)N=C(CN=C(C(CS)N=C(C(CC(=O)O)N=C(CN)O)O)O)O)O)O)O)O)O)O)O)O DIGQNXIGRZPYDK-WKSCXVIASA-N 0.000 description 1
- PRDFBSVERLRRMY-UHFFFAOYSA-N 2'-(4-ethoxyphenyl)-5-(4-methylpiperazin-1-yl)-2,5'-bibenzimidazole Chemical compound C1=CC(OCC)=CC=C1C1=NC2=CC=C(C=3NC4=CC(=CC=C4N=3)N3CCN(C)CC3)C=C2N1 PRDFBSVERLRRMY-UHFFFAOYSA-N 0.000 description 1
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 1
- QRXMUCSWCMTJGU-UHFFFAOYSA-N 5-bromo-4-chloro-3-indolyl phosphate Chemical compound C1=C(Br)C(Cl)=C2C(OP(O)(=O)O)=CNC2=C1 QRXMUCSWCMTJGU-UHFFFAOYSA-N 0.000 description 1
- 241000649047 Adeno-associated virus 12 Species 0.000 description 1
- 108091093088 Amplicon Proteins 0.000 description 1
- 101150002428 Atoh1 gene Proteins 0.000 description 1
- 241000714230 Avian leukemia virus Species 0.000 description 1
- 241000283690 Bos taurus Species 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 101150044789 Cap gene Proteins 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- 108090000565 Capsid Proteins Proteins 0.000 description 1
- 102100023321 Ceruloplasmin Human genes 0.000 description 1
- 102000004420 Creatine Kinase Human genes 0.000 description 1
- 108010042126 Creatine kinase Proteins 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- LTMHDMANZUZIPE-AMTYYWEZSA-N Digoxin Natural products O([C@H]1[C@H](C)O[C@H](O[C@@H]2C[C@@H]3[C@@](C)([C@@H]4[C@H]([C@]5(O)[C@](C)([C@H](O)C4)[C@H](C4=CC(=O)OC4)CC5)CC3)CC2)C[C@@H]1O)[C@H]1O[C@H](C)[C@@H](O[C@H]2O[C@@H](C)[C@H](O)[C@@H](O)C2)[C@@H](O)C1 LTMHDMANZUZIPE-AMTYYWEZSA-N 0.000 description 1
- 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 1
- 108010067770 Endopeptidase K Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 239000001856 Ethyl cellulose Substances 0.000 description 1
- ZZSNKZQZMQGXPY-UHFFFAOYSA-N Ethyl cellulose Chemical compound CCOCC1OC(OC)C(OCC)C(OCC)C1OC1C(O)C(O)C(OC)C(CO)O1 ZZSNKZQZMQGXPY-UHFFFAOYSA-N 0.000 description 1
- 108700024394 Exon Proteins 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 206010064571 Gene mutation Diseases 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 102100031181 Glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- 239000007995 HEPES buffer Substances 0.000 description 1
- 208000032041 Hearing impaired Diseases 0.000 description 1
- 101710113860 Heat shock protein 68 Proteins 0.000 description 1
- 108010004889 Heat-Shock Proteins Proteins 0.000 description 1
- 102000002812 Heat-Shock Proteins Human genes 0.000 description 1
- 101150108210 IX gene 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
- 238000012404 In vitro experiment Methods 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 241000581650 Ivesia Species 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 108010052285 Membrane Proteins Proteins 0.000 description 1
- 102000018697 Membrane Proteins Human genes 0.000 description 1
- 102000003792 Metallothionein Human genes 0.000 description 1
- 108090000157 Metallothionein Proteins 0.000 description 1
- 241000713869 Moloney murine leukemia virus Species 0.000 description 1
- 241000713333 Mouse mammary tumor virus Species 0.000 description 1
- 101100202288 Mus musculus Slc26a5 gene Proteins 0.000 description 1
- 102000003505 Myosin Human genes 0.000 description 1
- 108060008487 Myosin Proteins 0.000 description 1
- 108091092724 Noncoding DNA Proteins 0.000 description 1
- 108020004711 Nucleic Acid Probes Proteins 0.000 description 1
- 241000701945 Parvoviridae Species 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 108091027981 Response element Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 101710137500 T7 RNA polymerase Proteins 0.000 description 1
- 239000008049 TAE buffer Substances 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 108700009124 Transcription Initiation Site Proteins 0.000 description 1
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Triethanolamine Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 1
- 102100031834 Unconventional myosin-VI Human genes 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 1
- DPXJVFZANSGRMM-UHFFFAOYSA-N acetic acid;2,3,4,5,6-pentahydroxyhexanal;sodium Chemical compound [Na].CC(O)=O.OCC(O)C(O)C(O)C(O)C=O DPXJVFZANSGRMM-UHFFFAOYSA-N 0.000 description 1
- HGEVZDLYZYVYHD-UHFFFAOYSA-N acetic acid;2-amino-2-(hydroxymethyl)propane-1,3-diol;2-[2-[bis(carboxymethyl)amino]ethyl-(carboxymethyl)amino]acetic acid Chemical compound CC(O)=O.OCC(N)(CO)CO.OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O HGEVZDLYZYVYHD-UHFFFAOYSA-N 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000012841 animal feeding operation Methods 0.000 description 1
- 238000010171 animal model Methods 0.000 description 1
- 230000000692 anti-sense effect Effects 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 210000003050 axon Anatomy 0.000 description 1
- 230000001580 bacterial effect Effects 0.000 description 1
- 210000002469 basement membrane Anatomy 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000029803 blastocyst development Effects 0.000 description 1
- 230000000903 blocking effect Effects 0.000 description 1
- 230000036760 body temperature Effects 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 235000011148 calcium chloride Nutrition 0.000 description 1
- 210000000234 capsid Anatomy 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000030833 cell death Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 229920002301 cellulose acetate Polymers 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- RNFNDJAIBTYOQL-UHFFFAOYSA-N chloral hydrate Chemical compound OC(O)C(Cl)(Cl)Cl RNFNDJAIBTYOQL-UHFFFAOYSA-N 0.000 description 1
- 229960002327 chloral hydrate Drugs 0.000 description 1
- 238000012411 cloning technique Methods 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 239000002285 corn oil Substances 0.000 description 1
- 235000005687 corn oil Nutrition 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 230000030808 detection of mechanical stimulus involved in sensory perception of sound Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 230000004069 differentiation Effects 0.000 description 1
- LTMHDMANZUZIPE-PUGKRICDSA-N digoxin Chemical compound C1[C@H](O)[C@H](O)[C@@H](C)O[C@H]1O[C@@H]1[C@@H](C)O[C@@H](O[C@@H]2[C@H](O[C@@H](O[C@@H]3C[C@@H]4[C@]([C@@H]5[C@H]([C@]6(CC[C@@H]([C@@]6(C)[C@H](O)C5)C=5COC(=O)C=5)O)CC4)(C)CC3)C[C@@H]2O)C)C[C@@H]1O LTMHDMANZUZIPE-PUGKRICDSA-N 0.000 description 1
- 229960005156 digoxin Drugs 0.000 description 1
- LTMHDMANZUZIPE-UHFFFAOYSA-N digoxine Natural products C1C(O)C(O)C(C)OC1OC1C(C)OC(OC2C(OC(OC3CC4C(C5C(C6(CCC(C6(C)C(O)C5)C=5COC(=O)C=5)O)CC4)(C)CC3)CC2O)C)CC1O LTMHDMANZUZIPE-UHFFFAOYSA-N 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 230000003467 diminishing effect Effects 0.000 description 1
- 238000002224 dissection Methods 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 210000005069 ears Anatomy 0.000 description 1
- 230000005014 ectopic expression Effects 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 235000019325 ethyl cellulose Nutrition 0.000 description 1
- 229920001249 ethyl cellulose Polymers 0.000 description 1
- DEFVIWRASFVYLL-UHFFFAOYSA-N ethylene glycol bis(2-aminoethyl)tetraacetic acid Chemical compound OC(=O)CN(CC(O)=O)CCOCCOCCN(CC(O)=O)CC(O)=O DEFVIWRASFVYLL-UHFFFAOYSA-N 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000000796 flavoring agent Substances 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 235000013355 food flavoring agent Nutrition 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000003209 gene knockout Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 238000010362 genome editing Methods 0.000 description 1
- 239000003862 glucocorticoid Substances 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- 150000003278 haem Chemical class 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 229940094991 herring sperm dna Drugs 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 238000003125 immunofluorescent labeling Methods 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 238000012744 immunostaining Methods 0.000 description 1
- 230000000415 inactivating effect Effects 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 238000009413 insulation Methods 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 210000002977 intracellular fluid Anatomy 0.000 description 1
- 239000007928 intraperitoneal injection Substances 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 239000012669 liquid formulation Substances 0.000 description 1
- 231100000053 low toxicity Toxicity 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 239000012931 lyophilized formulation Substances 0.000 description 1
- 229910001629 magnesium chloride Inorganic materials 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 210000001595 mastoid Anatomy 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 238000010369 molecular cloning Methods 0.000 description 1
- 230000009456 molecular mechanism Effects 0.000 description 1
- 238000010172 mouse model Methods 0.000 description 1
- 108010049787 myosin VI Proteins 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 239000002853 nucleic acid probe Substances 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 102000039446 nucleic acids Human genes 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- 238000001543 one-way ANOVA Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 231100000763 ototoxin Toxicity 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 239000006072 paste Substances 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 239000000186 progesterone Substances 0.000 description 1
- 229960003387 progesterone Drugs 0.000 description 1
- 230000000750 progressive effect Effects 0.000 description 1
- 210000001236 prokaryotic cell Anatomy 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 101150066583 rep gene Proteins 0.000 description 1
- 230000003362 replicative effect Effects 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 230000000630 rising effect Effects 0.000 description 1
- 210000002265 sensory receptor cell Anatomy 0.000 description 1
- 102000027509 sensory receptors Human genes 0.000 description 1
- 108091008691 sensory receptors Proteins 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 235000019812 sodium carboxymethyl cellulose Nutrition 0.000 description 1
- 229920001027 sodium carboxymethylcellulose Polymers 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 210000003582 temporal bone Anatomy 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 229940124597 therapeutic agent Drugs 0.000 description 1
- 230000005030 transcription termination Effects 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 241001529453 unidentified herpesvirus Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 229960005486 vaccine Drugs 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 230000001720 vestibular Effects 0.000 description 1
- 230000029812 viral genome replication Effects 0.000 description 1
- 210000002845 virion Anatomy 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/17—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- A61K38/1703—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- A61K38/1709—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/0008—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
- A61K48/0058—Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/16—Otologicals
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4702—Regulators; Modulating activity
-
- 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
- C12N15/86—Viral vectors
-
- 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
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14141—Use of virus, viral particle or viral elements as a vector
- C12N2750/14143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Organic Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Molecular Biology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Biotechnology (AREA)
- Epidemiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Zoology (AREA)
- Biochemistry (AREA)
- Biomedical Technology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- General Engineering & Computer Science (AREA)
- Biophysics (AREA)
- Wood Science & Technology (AREA)
- Gastroenterology & Hepatology (AREA)
- General Chemical & Material Sciences (AREA)
- Immunology (AREA)
- Virology (AREA)
- Physics & Mathematics (AREA)
- Marine Sciences & Fisheries (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Plant Pathology (AREA)
- Microbiology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Toxicology (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
The invention discloses a specific cis-form regulatory element of cochlear outer hair cells and application thereof. Specifically, the invention discloses a cis-regulatory element specific to cochlear outer hair cells, which comprises the amino acid sequence shown in SEQ ID NO: 1 or SEQ ID NO: 2, can be used for driving effector gene to be specifically expressed in cochlear outer hair cells. The extracochlear hair cell specific cis-regulatory element can drive Prestin specificity to be expressed in extracochlear hair cells, so that hearing impairment caused by Prestin mutation can be treated. AAV viruses constructed by using the cis-regulatory element of the present invention are expected to become a new method for clinical treatment of hearing impairment.
Description
Technical Field
The invention relates to the technical field of biology, in particular to a cis-form regulatory element for specificity of cochlear outer hair cells and application thereof.
Background
The cochlea is the auditory organ of mammals, located ventrally of the inner ear, responsible for the detection of sound. The sensory receptors for sound, Hair Cells (HC), are located in the auditory epithelium of the cochlea (also known as the Organ of Corti, Organ of Corti (OC)). There are two types of hair cells, Inner Hair Cells (IHC) and Outer Hair Cells (OHC). These two cell types share many HC markers, such as Myo6, but they also differ in many ways, such as cell morphology, gene expression profile, function, ototoxin susceptibility, and the like.
Extra cochlear hair cells (OHCs) are essential for mammals to perceive sound. OHC has the function of a sound amplifier, depending on the motor protein Prestin expressed on the lateral membrane. Prestin is expressed only in OHC throughout development and adulthood. However, the mechanism of how to achieve specificity of Prestin expression in time and space is not clear.
The gene mutation of the outer hair cell can cause hearing diseases such as deafness and the like, and is closely related to the deletion of Prestin expression. Therefore, there is a need in the art to develop a method for treating hearing diseases such as deafness caused by mutation of outer hair cell gene.
Disclosure of Invention
The invention aims to provide a cis-regulatory element specific to an extracochlear hair cell and application thereof in repairing and/or regenerating the extracochlear hair cell to treat hearing loss.
In a first aspect of the invention, there is provided the use of a combination of active ingredients for the manufacture of a formulation or medicament for: (i) repairing and/or regenerating cochlear outer hair cells; (ii) treating or preventing hearing impairment; and/or (iii) specific expression of effector genes in cochlear outer hair cells;
wherein, the active ingredient combination comprises:
(a) a cochlear outer hair cell-specific cis-regulatory element;
(b) a promoter; and
(c) an effector gene or a coding sequence thereof.
In another preferred example, the hearing impairment is hearing impairment associated with degeneration and/or damage of cochlear Outer Hair Cells (OHC).
In another preferred embodiment, the degeneration of extracochlear hair cells (OHC) is the degeneration caused by mutation of Prestin protein in OHC cells.
In another preferred embodiment, the cochlear outer hair cell is a human or non-human mammalian cochlear outer hair cell.
In another preferred embodiment, the nucleotide sequence of the cochlear outer hair cell-specific cis-regulatory element is as set forth in SEQ ID NO: 1 or SEQ ID NO: 2, respectively.
In another preferred embodiment, the promoter is selected from the group consisting of: hsp68, L7, thy-1, recoverin, calbindin, human CMV, GAD-67, chicken actin, CAG or CBA.
In another preferred embodiment, the promoter is Hsp 68.
In another preferred embodiment, the effector gene is a gene encoding a Prestin protein.
In another preferred embodiment, the cis-regulatory element drives expression of the effector gene specifically in cochlear outer hair cells.
In a second aspect of the invention, there is provided an active ingredient combination comprising:
(a) a cochlear outer hair cell-specific cis-regulatory element;
(b) a promoter; and
(c) an effector gene or coding sequence thereof;
wherein, the nucleotide sequence of the cochlea outer hair cell specificity cis-form regulatory element is shown as SEQ ID NO: 1 or SEQ ID NO: 2, respectively.
In another preferred embodiment, the promoter is selected from the group consisting of: hsp68, L7, thy-1, recoverin, calbindin, human CMV, GAD-67, chicken actin, CAG or CBA.
In another preferred embodiment, the promoter is Hsp 68.
In another preferred embodiment, the effector gene is a gene encoding a Prestin protein.
In another preferred embodiment, the cis regulatory element drives the expression of the effector gene specifically in extracochlear hair cells.
In a third aspect of the present invention, there is provided an expression vector comprising an expression cassette, wherein the expression cassette has a structure of formula I from 5 'end to 3' end:
Z0-Z1-Z2-Z3-Z0’ (I)
in the formula (I), the compound is shown in the specification,
each "-" is independently a chemical bond or a nucleotide linking sequence;
z0 is none, or a 5' UTR sequence;
z1 is the nucleotide sequence of the cis-regulatory element of the specificity of the cochlear outer hair cell;
z2 is a promoter sequence;
z3 is the nucleotide sequence of an effector gene; and
z0 'is a null, or 3' UTR sequence.
In another preferred embodiment, each nucleotide linker sequence is 1 to 30nt, preferably 1 to 15nt, more preferably 3 to 6nt in length.
In another preferred embodiment, the nucleotide linker sequence is derived from a nucleotide linker sequence cleaved by a restriction endonuclease.
In another preferred embodiment, the nucleotide sequence of the cochlear outer hair cell-specific cis-regulatory element is set forth in SEQ ID NO: 1 or SEQ ID NO: 2, respectively.
In another preferred embodiment, the promoter is selected from the group consisting of: hsp68, L7, thy-1, recoverin, calbindin, human CMV, GAD-67, chicken actin, CAG or CBA.
In another preferred embodiment, the promoter is Hsp 68.
In another preferred embodiment, the effector gene is a gene encoding a Prestin protein.
In another preferred embodiment, the expression vector is selected from the group consisting of: plasmids, viral vectors.
In another preferred embodiment, the expression vector is selected from the group consisting of: eukaryotic expression vectors, lentiviral vectors, adenoviral vectors, adeno-associated viral vectors (AAV), or combinations thereof.
In another preferred example, the expression vector is an adeno-associated virus AAV vector.
In another preferred embodiment, the AAV vector is selected from the group consisting of: AAV2.7m8, AAV-ie or AAV-Anc 80.
In another preferred embodiment, said expression vector is used for expressing said effector gene.
In another preferred embodiment, the expression vector is used for expressing Prestin protein.
In another preferred embodiment, the expression vector is used for specifically expressing effector genes in cochlear outer hair cells.
In a fourth aspect of the invention, there is provided a host cell comprising an expression vector according to the third aspect of the invention.
In another preferred embodiment, the host cell is selected from the group consisting of: eukaryotic cells or prokaryotic cells.
In another preferred embodiment, the host cell is a mammalian cell, including human and non-human mammals.
In another preferred embodiment, the host cell is an extracochlear hair cell of a human or non-human mammal.
In a fifth aspect of the invention, there is provided a pharmaceutical formulation comprising: (a) the combination of active ingredients according to the second aspect of the invention, or the expression vector according to the third aspect of the invention, or the cell according to the fourth aspect of the invention, and (b) a pharmaceutically acceptable carrier or excipient.
In another preferred embodiment, the dosage form of the pharmaceutical formulation is selected from the group consisting of: a lyophilized formulation, a liquid formulation, or a combination thereof.
In another preferred embodiment, the carrier is selected from the group consisting of: a lentiviral vector, an adenoviral vector, an adeno-associated viral vector (AAV), or a combination thereof; preferably, the vector is an AAV vector; more preferably AAV2.7m8, AAV-ie or AAV-Anc 80.
In another preferred embodiment, the content of the carrier in the pharmaceutical preparation is 1 × 1011-1×1014Individual virus/ml, preferably 1X 1012-1×1013Individual virus/ml.
In another preferred embodiment, the therapeutically effective amount of the pharmaceutical formulation is 1 × 1011-1×1014A virus, preferably 1X 1012-1×1013And (4) viruses.
In another preferred embodiment, the pharmaceutical formulation is for use in the treatment or prevention of hearing impairment, preferably the treatment of cochlear outer hair cell degeneration and/or damage.
In a sixth aspect of the invention, there is provided the use of a combination of active ingredients according to the second aspect of the invention, or an expression vector according to the third aspect of the invention, or a host cell according to the fourth aspect of the invention, or a pharmaceutical formulation according to the fifth aspect of the invention, in the manufacture of a medicament for the treatment or prevention of hearing loss.
In another preferred example, the hearing impairment is hearing impairment associated with degeneration and/or damage of cochlear outer hair cells.
In a seventh aspect of the invention, there is provided a method of repairing and/or regenerating outer cochlear hair cells, comprising transducing the active ingredient combination of the second aspect of the invention, or the expression vector of the third aspect of the invention, into degenerated or damaged outer cochlear hair cells.
In another preferred embodiment, the Prestin protein in the denatured or damaged cochlear outer hair cells is mutated.
In another preferred embodiment, the mutation comprises an inactivating mutation resulting from deletion, insertion or substitution of a single base or multiple bases in the coding sequence.
In an eighth aspect of the invention, there is provided a method of treating or preventing hearing impairment, the method comprising administering to a subject in need thereof a pharmaceutical formulation according to the fifth aspect of the invention.
In another preferred example, the hearing impairment is hearing impairment associated with degeneration and/or damage of extracochlear hair cells.
In another preferred embodiment, the method is a method of reducing degeneration and/or damage of extracochlear hair cells in a patient suffering from or at risk of developing hearing impairment.
In another preferred embodiment, the subject in need thereof includes humans and non-human mammals.
In another preferred example, the subject in need thereof has hearing impairment associated with degeneration and/or damage of outer cochlear hair cells
In another preferred embodiment, the method comprises directly administering the expression vector to the ear of a subject in need thereof.
In another preferred embodiment, the method comprises directly implanting the host cell into a cochlea of a subject in need thereof.
In another preferred example, the method can repair cochlear outer hair cells of a subject in need of hearing impairment associated with degeneration and/or damage of cochlear outer hair cells.
In another preferred embodiment, the method is such that the auditory function is substantially restored or maintained in the treated ear.
In a ninth aspect of the invention, there is provided a use of a cochlear outer hair cell-specific cis regulatory element, the cis regulatory element comprising an amino acid sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 2 for driving the expression of effector genes specifically in cochlear outer hair cells.
In another preferred embodiment, the effector gene is a gene encoding a Prestin protein.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Drawings
FIG. 1: the deletion of the 13kb fragment suppressed expression of Prestin at P6. (A) The 13kb fragment is located in Prestin intron 2. (B) Sanger sequencing results of DNA from rat tail of Prestin-13k/+ mice. (C) Gel electrophoresis of PCR amplification products from mouse rat tail DNA from wild type (Prestin +/+), heterozygote (Prestin-13k/+) and homozygote (Prestin-13k/Prestin-13 k). (D-E) Prestin +/+ (D) and Prestin-13k/Prestin-13k (E) P6 cochlear tissue sample Whole-standard in situ RNA hybridization staining. Prestin mRNA is specifically expressed in wild-type outer hair cells (D), but cannot be detected in homozygous mutants (E). (F-G) immunohistofluorescence detection of Prestin using samples of cochlear tissue from P5 Prestin +/+ (F) and Prestin-13k/Prestin-13k (G). The Prestin protein is specifically expressed in wild-type outer hair cells (F), but no signal of the Prestin protein can be detected in homozygous mutants (G). The panels embedded in (F) are the same panels, but only the signal of the Prestin channel is presented. (H) Schematic flow chart of manual selection of tdTomato positive cells from P5 wild type, heterozygote and homozygote mice. (I-J) the content of Pretsin and Myo6 in cells derived from mice of different genotypes in the (H) picture was determined using the qPCR method. Deletion of the 13kb fragment had an effect on Prestin mRNA and a dose effect, but not Myo6 mRNA. Enrichment of Myo6 in hair cells demonstrated high purity of hair cells (J). P <0.05, p <0.001, scale: 20 μm (E, G).
FIG. 2: deletion of the near 5' 4kb fragment of intron 2 replicates the phenotype of the Prestin-13k mutant. (A) The distribution position of the 4k fragment (blue line, #1) on the Prestin intron-2 is shown. Mice with the other two fragments (#2 and #3) deleted did not observe a phenotype. (B) Morge sequencing of mouse rat tail DNA from Prestin 4k-1/+ mice. (C) Gel electrophoresis of PCR amplification products from mouse rat tail DNA from wild type (Prestin +/+), heterozygote (Prestin-4k/+) and homozygote (Prestin-4k-1/Prestin-4 k-1). (D-E') immunofluorescence Co-staining for Prestin and Myo6 in cochlear tissue samples of Prestin (+/+) and Prestin-4k-1/Prestin-4k-1 at P5. The Prestin protein is specifically expressed in wild-type outer hair cells (D-D '), but no signal of the Prestin protein can be detected in homozygous mutants (E-E'). (F-I) NLC measurements were performed on OHC's of 5 different genotypes at P14.5(F) and P60(G), respectively. The NLC curve of the Prestin-4k-1 mutant overlaps with that of the Prestin-13k mutant. Notably, no difference was found in the total Prestin content between Prestin-13k/+ and Prestin-4k-1/+ at P14.5 and P60 (H). Again, there was no difference between P14.5 and P60(I), Prestin-13k/Prestin-13k and Prestin-4k-1/Prestin-4 k-1.
FIG. 3: preset expression of Prestin requires a 1.4k fragment. (A-C) schematically illustrates how random deletions were induced in the 4k fragment (blue line) using multiple sgRNAs, of which 7 more potent sgRNAs have been previously tested for efficiency. Sgrnas were divided into two subgroups: group 1 at the 5' end, includes sg1, 5, 6, 7 and 8; group 2 includes sgrnas 8, 9, 10, 11, and 12 at the 3' end. Cas9 mRNA was injected into one cell phase zygote along with 5 sgrnas of group 1 (B) and group 2 (C), yielding 32 and 43F 0 mice, respectively. (D) Shows sanger sequencing of 1/32F0 mouse rat tail DNA from group 1 sgrnas (b). Sequencing results show that the mutant is possibly a homozygous mutant with 1429bp (1.4k) fragment deletion between sgRNA-1 and sgRNA-7. (E-F') immunofluorescence co-staining of Prestin and Myo6 in mouse cochlear specimens of P9 (D). The images are acquired at a low resolution (E-E ') and a high resolution (F-F'), respectively. A heterogeneous expression pattern of Prestin can be observed. Outer hair cells with high Prestin should be of wild type genotype (arrow in F-F '), and outer hair cells with very low or undetectable Prestin should be homozygous deleted for 1.4k (asterisk in F-F'). In conclusion, the mouse should be a chimera. The tail cells are mostly homozygous mutations, resulting in the inability to amplify the wild-type PCR product. Scale bar: 100 μm (E'); 20 μm (F').
FIG. 4: the 1.4kb fragment of mouse Presin and the 398bp fragment of human Prestin were sufficient to drive the expression of specific green fluorescent protein in cochlear OHC. (A) The transgenic mice were obtained by the Piggybac method with a 1.4kb fragment (as enhancer) and Hsp68 promoter to control EGFP. A unique transgene insertion pattern was present for each F0, but could be analyzed directly without further propagation. (B-B') immunofluorescent co-staining of EGFP and Myo6 was performed on cochlear tissue samples of F0 mice from P14. Viewing the image of the insert in (B-B') under a high power lens, EGFP expression was specific for outer hair cells, but not all outer hair cells were EGFP positive, indicating that the transgene insert in F0 mice was chimeric. (C-C') A cochlear tissue sample from another F0 mouse was subjected to immunofluorescence co-staining for EGFP and Myo6 at P21. EGFP signals were expressed in all outer hair cells, indicating that transgene insertion was not chimeric in this mouse. (D-F') human Prestin and mouse Prestin have 398bp highly homologous sequences. The sequence of 398bp is utilized to express EGFP in a mouse cochlea OHC specifically. Scale bar: 200 μm (B ') and 20 μm (C ', F ').
FIG. 5: the 1.4kb fragment of mouse Presin is aligned with the 398bp fragment of human Prestin for detailed similarity.
FIG. 6: the AAV virus made from mouse 1.4kb sequence and human 398bp sequence is used to specifically infect cochlear OHC. (A) AAV was produced using a 1.4kb sequence of the mouse Prestin gene and injected into the cochlea of a mouse. (B-B') P17 analysis of mouse cochlea, EGFP is mainly expressed in the extracochlear hair cells. (C) The 398bp sequence of the human Prestin gene is used to make AAV virus, which is injected into the cochlea of mouse. (D-D') P17 analysis of mouse cochlea, EGFP is also predominantly expressed on the extracochlear hair cells.
Detailed Description
The inventors of the present invention have conducted extensive and intensive studies and have surprisingly screened, for the first time, an extracochlear hair cell-specific cis regulatory element that can drive effector genes to be specifically expressed in extracochlear hair cells. Experiments prove that the extracochlear hair cell specificity cis-regulatory element regulates the expression of Prestin in extracochlear hair cells, and is a necessary condition for timely starting the expression of Prestin in OHC. And the AAV expression vector constructed by using the extracochlear hair cell specific cis-regulatory element can drive the specific expression of effector genes in the expression vector in the extracochlear hair cells.
On the basis of this, the present invention has been completed.
Term(s) for
In order that the disclosure may be more readily understood, certain terms are first defined. As used in this application, each of the following terms shall have the meaning given below, unless explicitly specified otherwise herein. Other definitions are set forth throughout the application.
The term "about" can refer to a value or composition that is within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
As used herein, the term "comprising" or "includes" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of …," or "consisting of ….
Sequence identity (homology) is determined by comparing two aligned sequences along a predetermined comparison window (which may be 50%, 60%, 70%, 80%, 90%, 95% or 100% of the length of the reference nucleotide sequence or protein) and determining the number of positions at which identical residues occur. Typically, this is expressed as a percentage. The measurement of sequence identity of nucleotide sequences is a method well known to those skilled in the art.
As used herein, the terms "subject", "subject in need thereof" refer to any mammal or non-mammal. Mammals include, but are not limited to, humans, vertebrates such as rodents, non-human primates, cows, horses, dogs, cats, pigs, sheep, goats.
As used herein, the terms "hearing impaired," "hearing disorder," and "hearing impairment" refer to a hearing dysfunction resulting from degeneration and/or damage of extracochlear hair cells.
Cis regulatory elements
Gene expression is dependent on two key components: one is a cis-regulatory element (CRE) consisting of a Transcription Factor (TF) binding site and other non-coding DNA sufficient to drive gene expression in a controlled temporal and spatial manner; the other is a trans-acting factor, such as TF that binds CRE. Promoters and enhancers are two major types of CRE. Promoters are usually close to the transcription start site, but promoters alone can only produce a minimal level of mRNA expression (Brown and Feder, 2005). Enhancers are widely distributed upstream, downstream, introns, or distal to individual genes and are critical for CRE differentiation. For example, one of the Atoh1 gene enhancers, located about 2 kb downstream of its 3' -UTR, is thought to be involved in determining the expression level of Atoh 1. Only cochlear sensory precursor cells expressing high levels of Atoh1 eventually develop into HC. Furthermore, gene expression is often controlled by multiple enhancers. Each enhancer may affect the expression pattern of a gene in a particular cell type or at a particular developmental stage.
Thyroid Hormone (TH) is a trans-acting factor, and a TH Response Element (TRE) plays a crucial role in regulating the expression of rat Prestin, and is presumed to be distributed in long intron 2 (. about.20 kbp) between exon 2 and exon 3 of rat Prestin. However, genetic evidence in vivo has not proven the importance of intron 2 sequences. In addition, it is not clear which part is the critical CRE, because 20 kbbp is too long to act as a driver to activate ectopic expression of specific genes in OHCs, finding a critical and short CRE provides a deeper understanding of the molecular mechanisms that reveal specific Prestin expression patterns in OHCs, and finding an outer hair cell specific CRE is of great clinical significance for gene therapy of deafness.
The invention provides a specific cis-regulatory element of an outer hair cell of a cochlea, wherein the cis-regulatory element comprises the amino acid sequence shown as SEQ ID NO: 1 or SEQ ID NO: 2 for driving the expression of effector genes specifically in cochlear outer hair cells. The cis-regulatory element is a 1.4kb sequence (SEQ ID NO: 1) located in mouse Prestin intron 2, which has about 80% sequence identity to a 398bp sequence (SEQ ID NO: 2) in human Prestin intron by sequence alignment. In the present example, AAV viruses constructed with the mouse 1.4kb sequence and the human 398bp sequence were able to express effector genes specifically in extracochlear hair cells.
Effector gene
As used herein, the term "effector gene" refers to any gene capable of being expressed in the outer hair cells of the cochlea. The sequence of the effector gene may be a sequence derived from the coding region (CDS) or open reading frame, or a sequence having at least 85% sequence identity with the sequence of the coding region (CDS) or open reading frame.
In another preferred embodiment, the "effector gene" refers to a gene that can be expressed and exert efficacy in extracochlear hair cells. Including but not limited to, those related to morphology, function of outer hair cells of the cochlea. For example, the efficacy of maintaining the sound sensitivity of the outer hair cells of the cochlea. In a preferred embodiment of the present invention, the effector gene is a gene encoding a Prestin protein.
Prestin
Prestin is a motor-type membrane protein encoded by the Slc26a5 gene and expressed specifically and persistently in the outer hair cells of the cochlea. The main function of Prestin protein is to help outer hair cells to stretch their own length when sound comes, thereby increasing the sensitivity of human or animal to sound. Both Prestin-/-mice and the population carrying the Prestin point mutation had a marked reduction in hearing capacity, and the outer hair cells of Prestin-/-mice developed progressive cell death from the high to low frequency region with age.
Prestin's information such as https:// www.ncbi.nlm.nih.gov/protein/? the information of the Slc26a5 gene, shown in term of Prestin, is https:// www.ncbi.nlm.nih.gov/gene/? term — Slc26a 5. As used herein, the term "gene encoding a Prestin protein" refers to the Slc26a5 gene. In preferred embodiments herein, the Prestin protein is preferably a human or mouse Prestin protein.
Adeno-associated virus
Adeno-associated virus (AAV), also called adeno-associated virus, belongs to the genus dependovirus of the family parvoviridae, is the simplest single-stranded DNA-deficient virus of the currently discovered class, and requires a helper virus (usually adenovirus) to participate in replication. It encodes the cap and rep genes in inverted repeats (ITRs) at both ends. ITRs are crucial for replication and packaging of viruses. The cap gene encodes the viral capsid protein, and the rep gene is involved in viral replication and integration. AAV can infect a variety of cells.
The recombinant adeno-associated virus (rAAV) is derived from non-pathogenic wild adeno-associated virus, is considered to be one of the most promising gene transfer vectors due to the characteristics of good safety, wide host cell range (divided and non-divided cells), low immunogenicity, long time for expressing foreign genes in vivo and the like, and is widely applied to gene therapy and vaccine research in the world. Over 10 years of research, the biological properties of recombinant adeno-associated viruses have been well understood, and many data have been accumulated on the application effects of recombinant adeno-associated viruses in various cells, tissues and in vivo experiments. In medical research, rAAV is used in the study of gene therapy for a variety of diseases (including in vivo, in vitro experiments); meanwhile, the gene transfer vector is used as a characteristic gene transfer vector and is widely applied to the aspects of gene function research, disease model construction, gene knock-out mouse preparation and the like.
Expression vectors and host cells
The invention provides an expression vector, which comprises an expression cassette, wherein the expression cassette has a structure shown in a formula I from 5 'end to 3' end:
Z0-Z1-Z2-Z3-Z0’ (I)
in the formula (I), the compound is shown in the specification,
each "-" is independently a chemical bond or a nucleotide linking sequence;
z0 is none, or a 5' UTR sequence;
z1 is the nucleotide sequence of the cis-regulatory element of the specificity of the cochlear outer hair cell;
z2 is a promoter sequence;
z3 is the nucleotide sequence of an effector gene; and
z0 'is a null, or 3' UTR sequence.
With the sequence information provided, the skilled artisan can use available cloning techniques to generate nucleic acid sequences or vectors suitable for transduction into cells.
Preferably, the nucleic acid sequence encoding the Prestin protein is provided as a vector, preferably as an expression vector. Preferably, it may be provided as a gene therapy vector, preferably suitable for transduction and expression in a target cell (e.g. a cochlear support cell). The vector may be viral or non-viral (e.g., a plasmid). Viral vectors include those derived from adenovirus, adeno-associated virus (AAV), including mutated forms, retroviruses, lentiviruses, herpes viruses, vaccinia virus, MMLV, GaLV, Simian Immunodeficiency Virus (SIV), HIV, poxviruses, and SV 40. Preferably, the viral vector is replication defective, or may be replication deficient, replication capable or conditionally replicating. Viral vectors can generally remain extrachromosomal without integrating into the genome of the target cell. A preferred viral vector for introducing an expression cassette of the invention into a target cell is an AAV vector. Selective targeting can be achieved using specific AAV serotypes (AAV serotype 2 through AAV serotype 12) or modified versions of any of these serotypes. In a preferred embodiment of the invention, the AAV vector is preferably AAV2.7m8, AAV-ie or AAV-Anc 80.
The viral vector may be modified to delete any non-essential sequences. For example, in AAV the virus may be modified to delete all or part of the IX gene, Ela and/or Elb gene. Replication is very inefficient for wild type AAV, without the presence of helper viruses such as adenovirus. For recombinant adeno-associated viruses, preferably, the replication and capsid genes are provided in trans (in the pRep/Cap plasmid), and only the 2 ITRs of the AAV genome are retained and packaged into virions, while the desired adenoviral genes are provided by adenovirus or another plasmid. Similar modifications can be made to lentiviral vectors.
Viral vectors have the ability to enter cells. However, non-viral vectors such as plasmids may be complexed with agents to facilitate uptake of the viral vector by the target cell. Such agents include polycationic agents. Alternatively, a delivery system such as a liposome-based delivery system may be used. The vector for use in the present invention is preferably suitable for use in vivo or in vitro, and preferably for use in humans.
The vector will preferably comprise one or more regulatory sequences to direct expression of the nucleic acid sequence in the target cell. Regulatory sequences may include promoters, enhancers, transcription termination signals, polyadenylation sequences, origins of replication, nucleic acid restriction sites, and homologous recombination sites, operably linked to a nucleic acid sequence. The vector may also include a selectable marker, e.g., to determine expression of the vector in a growth system (e.g., a bacterial cell) or in a target cell.
By "operably linked" is meant that the nucleic acid sequences are functionally related to the sequences to which they are operably linked, such that they are linked in a manner that affects the expression or function of each other. For example, a nucleic acid sequence operably linked to a promoter will have an expression pattern that is affected by the promoter.
The promoter mediates expression of the nucleic acid sequence linked thereto. Promoters may be constitutive or may be inducible. The promoter may direct the expression of the gene ubiquitously in cochlear cells, or specifically in cochlear cells. In the latter case, the promoter may direct cell type specific expression, such as an extracochlear hair cell. Suitable promoters will be known to those skilled in the art. For example, suitable promoters may be selected from the group consisting of: hsp68, L7, thy-1, recoverin, calbindin, human CMV, GAD-67, chicken actin, CAG, CBA and the like, but not limited to the group. Furthermore, gene expression targeting specific cells can also be achieved using cell-specific promoters.
In a preferred embodiment of the invention, an example of a suitable promoter is the Hsp68 promoter. In addition, one example of a suitable promoter is the immediate early Cytomegalovirus (CMV) promoter sequence. The promoter sequence is a strong constitutive promoter sequence capable of driving high level expression of any polynucleotide sequence operably linked thereto. Another example of a suitable promoter is elongation growth factor-1 α (EF-1 α). However, other constitutive promoter sequences may also be used, including, but not limited to, the simian virus 40(SV40) early promoter, the Mouse Mammary Tumor Virus (MMTV), the Human Immunodeficiency Virus (HIV) Long Terminal Repeat (LTR) promoter, the MoMuLV promoter, the avian leukemia virus promoter, the Epstein-Barr (Epstein-Barr) virus immediate early promoter, the rous sarcoma virus promoter, and human gene promoters, such as, but not limited to, the actin promoter, myosin promoter, heme promoter, and creatine kinase promoter. Further, the present invention should not be limited to the use of constitutive promoters. Inducible promoters are also contemplated as part of the invention. The use of an inducible promoter provides a molecular switch that is capable of turning on expression of a polynucleotide sequence operably linked to the inducible promoter when expression is desired, or turning off expression when expression is desired to be turned off. Examples of inducible promoters include, but are not limited to, the metallothionein promoter, the glucocorticoid promoter, the progesterone promoter, and the tetracycline promoter.
The invention also provides a host cell for expressing the Prestin protein. Preferably, the host cell is a mammalian cell (preferably a human, more preferably a human cochlear outer hair cell).
Pharmaceutical formulations and compositions
The present invention provides a pharmaceutical formulation or composition comprising (a) the combination of active ingredients according to the second aspect of the invention, or the expression vector according to the third aspect of the invention, or the host cell according to the fourth aspect of the invention, and (b) a pharmaceutically acceptable carrier or excipient.
In another preferred embodiment, the pharmaceutical formulation is for the treatment of hearing impairment.
In another preferred embodiment, the pharmaceutical formulation is for use in treating degeneration and/or damage of cochlear outer hair cells.
The "active ingredient" in the pharmaceutical preparation of the present invention refers to a vector (vector) of the present invention, such as a viral vector (including adeno-associated viral vectors). The "active ingredients", formulations and/or compositions described herein may be used to treat hearing impairment. "safe and effective amount" means: the amount of active ingredient is sufficient to significantly ameliorate the condition or symptom without causing serious side effects. "pharmaceutically acceptable carrier or excipient (excipient)" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of being combined with the active ingredients of the present invention and with each other without significantly diminishing the efficacy of the active ingredient.
The composition may be a liquid or a solid, such as a powder, gel or paste. Preferably, the composition is a liquid, preferably an injectable liquid. Suitable excipients will be known to those skilled in the art.
In the present invention, the vector may be administered to the subject by direct administration to the ear or cochlea. In either mode of administration, preferably, the carrier is provided as an injectable liquid. Preferably, the injectable liquid is provided as a capsule or syringe.
In the present invention, the host cell according to the present invention may be implanted into a subject for cochlear administration. Preferably, the host cell is provided as an injectable liquid.
Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g. sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g. stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g. soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g. propylene glycol, glycerol, mannitol, sorbitol, etc.), emulsifiers (e.g. tween, etc.)) Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
The compositions may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Method of treatment
The present invention provides a method of repairing and/or regenerating cochlear outer hair cells, the method comprising transducing the active ingredient combination of the second aspect of the invention, or the expression vector of the third aspect of the invention, into degenerated or damaged cochlear outer hair cells.
The present invention provides a pharmaceutical formulation or composition comprising the active ingredient combination of claim 3, or the expression vector of claim 4, or a host cell containing said expression vector, and a pharmaceutically acceptable carrier or excipient for use in a method of treating hearing impairment by repairing and/or regenerating cochlear outer hair cells. The pharmaceutical formulation or composition of the invention may be administered alone or in combination with (e.g. formulated in the same pharmaceutical composition as) other therapeutic agents.
The invention also provides a method of treating hearing impairment, comprising administering to a subject a pharmaceutical formulation or composition of the invention. The hearing impairment is hearing impairment associated with degeneration and/or damage of outer cochlear hair cells. The method comprises administering the expression vector of the invention directly to the ear (cochlea) of a subject in need thereof. The method further comprises directly implanting the host cell of the invention into a cochlea of a subject in need thereof.
As used herein, repairing and/or regenerating an extracochlear hair cell means a cell that has not previously possessed, or whose extracochlear hair cell capacity has been completely or partially degraded, and which, upon expression of a foreign nucleic acid sequence therein using an expression vector according to the present invention, becomes a cell possessing the characterization and function of the extracochlear hair cell.
The main advantages of the invention are:
(1) the cis-regulatory element provided by the invention drives Prestin expression in the cochlear outer hair cells specifically, and the AAV constructed by using the cis-regulatory element only enables effector genes contained in a virus vector to be expressed in the cochlear outer hair cells specifically.
(2) The cis-regulatory element of the invention has short sequence, only 1.4kb, even only 398bp, low difficulty in constructing the vector and compatibility with the vector with small molecular weight.
(3) The cis-form regulatory element has application value in the aspect of clinical gene therapy of hearing diseases such as deafness and the like caused by cochlear outer hair cell mutation.
The invention is further illustrated with reference to specific embodiments. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally followed by conventional conditions, such as Sambrook et al, molecular cloning: conditions described in a Laboratory Manual (New York: Cold Spring Harbor Laboratory Press,1989), or according to the manufacturer's recommendations. Unless otherwise indicated, percentages and parts are by weight.
Methods and materials
Animal feeding
All mice were housed in the animal room according to a conventional 12 hour light/dark alternating schedule. Both male and female mice were used in the experiment. All animal feeding operations were performed according to the guidelines for laboratory animal Care and use of the national institutes of health and approved by the IACUC guidelines of the institute of neuroscience, Chinese academy of sciences (NA-032-2019).
Selection of high efficiency sgrnas
All sgrnas used herein were pretested in mouse one-cell stage zygotes. For testing, each sgRNA was injected into mouse fertilized eggs at a concentration of 50ng/ul together with Cas9 mRNA (100ng/ul), cultured for 3 days until blastocyst development stage, DNA was extracted, PCR amplified and Sanger (Sanger) sequenced. Each sgRNA was tested with 6 zygotes, and the criterion for effective sgrnas was considered to be that at least 3/6 zygotes were cleaved at the position targeted by the sgRNA.
The sgRNA sequences used herein are shown in the following table:
name (R) | Sequence 5 '-3' | SEQ ID NO: |
sgRNA-1 | |
3 |
sgRNA-2 | |
4 |
sgRNA-3 | |
5 |
sgRNA-4 | |
6 |
sgRNA-5 | |
7 |
sgRNA-6 | |
8 |
sgRNA-7 | |
9 |
sgRNA-8 | |
10 |
sgRNA-9 | |
11 |
sgRNA-10 | GTGAACTCCAGTTTCTCATA | 12 |
sgRNA-11 | |
13 |
Construction of different Prestin intron mutant mice by using CRISPR/Cas9 system
The sgRNA-1 and the sgRNA-4 are used for constructing a Prestin-13k intron knockout mouse, the sgRNA-1 and the sgRNA-2 are used for constructing a Prestin-4k-1 intron knockout mouse, the sgRNA-2 and the sgRNA-3 are used for constructing a Prestin-5k intron knockout mouse, and the sgRNA-3 and the sgRNA-4 are used for constructing a Prestin-4k-2 intron knockout mouse. Cas9 mRNA (100 ng/. mu.l) and two different combinations of sgRNAs (50 ng/. mu.l each) were co-injected into one-cell fertilized eggs of mice and then implanted into pseudopregnant females. Performing mouse tail PCR identification on the F0 mouse, and carrying out passage after crossing the positive mouse and the wild mouse.
Additionally, 7 effective sgRNAs were selected at approximately 500 base intervals within the Prestin-4k-1 intron, and random DNA deletions were generated in the left segment of the Prestin-4k intron by co-injection of Cas9 mRNA (100 ng/. mu.l) and the first set of sgRNAs (sgRNAs-1, 5, 6, 7, 8, 50 ng/. mu.l) in one-cell mouse zygotes; the random DNA deletion of the right segment of the Prestin-4k intron was generated by co-injection of Cas9 mRNA (100 ng/. mu.l) and a second set of sgRNAs (sgRNAs-8, 9, 10, 11, 2, 50 ng/. mu.l) into single-cell mouse zygotes. All F0 mice were analyzed for F0 generation homozygotes using rat tail DNA PCR screening and Sanger sequencing analysis, and the resulting F0 generation heterozygotes were cross-passaged with wild mice.
Construction of Prestin 1.4k-hsp68-EGFP + transgenic mice and Virus preparation
A Prestin 1.4k bp fragment is obtained by PCR by taking mouse genome DNA as a template and is subcloned into a vector containing PiggyBac 5 'homologous arm and 3' homologous arm sequences, a heat shock protein 68(hsp68) mini promoter and EGFP. Using the T7 promoter and mMACHINETMThe T7 super transcription kit (Cat #: AM1345, Thermo Fisher Scientific) transcribes PiggyBac transposase mRNA in vitro. Purified Prestin 1.4k-hsp68-EGFP + vector (50 ng/. mu.l) was co-injected with PiggyBac transposase mRNA (100 ng/. mu.l) into small miceThe fertilized eggs are implanted into a pseudopregnant female mouse in the first cell stage of the mouse. F0 mice first underwent rat tail PCR screening, and identified transgenic insert positive mice directly analyzed phenotype or with wild type mice hybridization, establishment of stable mouse strains. AAV virus production is completed in the gene editing platform of neuroscience research institute of Chinese academy of sciences.
Sample preparation and immunofluorescence staining
Inner ear tissue from P5 mice was harvested and fixed overnight at 4 ℃ in 4% PFA. Mice of P14, P21, P30 and P60 were perfused with 1xPBS and 4% PFA, respectively, and the inner ear was removed and fixed in 4% PFA overnight at 4 ℃. The fixed tissue was washed 3 times with 1xPBS and then treated with EDTA (120mM) decalcification (P14 and P21 tissues decalcification overnight, P30 and P60 tissues decalcification for 2-3 days). Whole-mount staining, cochlear tissue was first incubated with 1% Triton X-100(X-100, Sigma) and 5% BSA (Cat #: BP1605, Fisher Scientific) in blocking solution for 1 hour at room temperature, followed by primary antibody incubation with 0.1% Triton X-100 and 5% BSA at 4 ℃ overnight.
The primary antibodies used in this study were anti-Prestin (goal, 1:500, Cat #: sc-22692, Santa Cruz Biotechnology), anti-Prestin (rabbit,1:50000, ab242128, Abcam, produced by the laboratory in cooperation with Abcam), anti-myostatin-VI (rabbit,1:500, Cat #:25-6791, protein Bioscience), anti-GFP (chicken,1:1000, Cat #: ab13970, Abcam).
The following day, 3 washes with 1xPBS containing 0.1% Triton X-100, followed by incubation with secondary antibody Alexa 488Donkey anti-goat IgG (Cat #: # A11055, Molecular Probes) for 2 hours at room temperature; alexa568donkey anti-rabbit IgG (Molecular Probes, # A10042); alexa 488donkey anti-chicken IgG (Cat #: 703-. After the secondary antibody had been incubated, it was washed three times with 1xPBS containing 0.1% Triton X-100, and the sample was counterstained with Hoechst33342(1:1000, Cat #:62249, Thermo Scientific) to visualize the nuclei. The stained sample was mounted with Prolong gold mounting tape (Cat #: P36930, Thermo Scientific). The specimens were scanned and photographed by a Nikon C2 confocal microscope, and the photographed images were analyzed by Image-J (NIH) software.
Whole tissue RNA in situ hybridization
The Prestin-specific RNA probe covers exons 4-6 of Prestin, is 384 bases in length, and is subcloned into the pBluescript-II vector containing the T3 promoter. The final vector was linearized with NotI (Cat #: R3189s, New England BioLabs) and transcribed using T7 RNA polymerase (Cat #: P2075, Promega). The nucleic acid probes were labeled using Digoxigenin (DIG) kit (Cat #:11277073910, Roche).
The inner ear tissue of P5 mice was dissected out and fixed in 4% DEPC-PFA overnight at 4 ℃. The cochlear shell is partially knocked down in order to allow better binding of the RNA probe to the sensory epithelium. The first day of the experiment, DEPC-PBS and DEPC-PBS containing 0.1% tween-20 (P1379, Sigma) were washed three times, then incubated with 5 μ g/ml proteinase K (Cat #:25530049, Thermo fisher) for 10 minutes at room temperature, followed by a secondary fixation with DEPC-4% PFA for 10 minutes at room temperature, followed by a10 minute incubation with 0.1M rnase-free triethanolamine (Cat #: V900257, Sigma) at room temperature. Then prehybridization was carried out for 2 hours with 1ml of a hybridization solution containing 50% formamide (Cat #:15515026, Thermo fisher), 250. mu.g/ml yeast RNA (Cat #:10109495001, Roche) and 500. mu.g/ml herring sperm DNA (Cat #:15634017, Thermo fisher), and hybridization was carried out overnight at 65 ℃ with a hybridization solution containing 0.5 ng/. mu.l of digoxin-labeled Prestin antisense strand RNA probe after completion of the prehybridization. The following day, the tissues were rinsed with 1XTBST and further electrophoresed in TAE buffer at 60V for 2 hours, followed by incubation with anti-DIG-AP antibody (Cat #:11093274910, Roche) overnight at 4 ℃. On the third day, a color reaction was performed using NBT/BCIP kit (Cat #:11681451001, Roche), and the color-developed sample was mounted with Prolong gold mounting plate (Cat #: P36930, Thermo Scientific), photographed with Olympus VS120 microscope, and subjected to subsequent data analysis using ImageJ (NIH) software.
Manual cell picking and qPCR analysis
Mouse models of the following three genotypes were used for the experiments:
1)Atoh1-CreER+;Ai9/+
2)Atoh1-CreER+;Ai9/+;Prestin-13k/+
3)Atoh1-CreER+;Ai9/+;Prestin-13k/Prestin-13k,
these three models were used to select wild-type hair cells, heterozygous knockout hair cells for the Prestin-13k intron sequence, and homozygous knockout hair cells for the Prestin-13k intron sequence, respectively. All mice were induced at P0, P1 at 3mg/40g mouse weight with tamoxifen (Cat #: T5648, Sigma), dissolved in corn oil (Cat #: C8267, Sigma), with inner and outer hair cells marked red by tdTomato, and tdTomato positive cells were manually selected at P5. This experiment was repeated three times, with each set of experiments containing three genotypes, with approximately 20-25 hair cells selected for each genotype. Dissection of the inner ear, manual cell selection and qPCR experimental procedures were as described in the previous article (Li et al, 2020 a; Li et al, 2018; Li et al, 2020 b). Total RNA inverted cDNA of P11 cochlear tissue was used as a normalization reference, and Prestin, Myosin-VI and GAPDH (internal reference gene) primers were selected by calculating amplification efficiency from a standard curve.
Testing of Auditory Brainstem Response (ABR)
Mice at 14.5 days, 21 days, 30 days and 60 days after taking out are divided into a homozygous group, a heterozygous group and a wild type group, at least 6 mice in each group are subjected to Auditory Brainstem Response (ABR) test, and 7 frequencies are detected in total: 4kHz, 5.6kHz, 8kHz, 11.3kHz, 16kHz, 22.6kHz and 32 kHz. The hearing data were analyzed using Excel and Graphpad prism 8 software.
The specific steps of the ABR test are as follows:
1) mice were weighed on an electronic balance and anesthetized by intraperitoneal injection with chloral hydrate (Cat #:23100, Sigma,480 mg/kg).
2) The anesthetized mouse is placed in a sound insulation chamber, a body temperature controller (Cat #:55-7020, Harvard Apparatus) is turned on, a temperature probe is placed below the body of the mouse, and the constant temperature of 37 ℃ is set.
3) Placing an electrode: electrodes were inserted subcutaneously at the cranial crown (recording electrode), right mastoid (reference electrode) and left shoulder (ground electrode), respectively. The preamplifier was turned on and the resistance was confirmed to not exceed 1 k.
4) ABR testing was performed using biosignz software under an open sound field.
A loudspeaker (MF1, Tucker-Davis Technologies, Inc, Alachua, FL, USA) is placed 10cm away from the two ears of the mouse, a 3 ms-long Toneburst short pure tone signal is adopted as a stimulating sound, the time length of voltage rising and voltage falling is 1ms, the short pure tone stimulating frequency is 20 times/second, the recording time is 15ms, and the average superposition is 400 times. The maximum stimulation sound intensity is 90dB SPL, the short pure tone intensity range is from 0 to 90dB SPL, the minimum stimulation intensity capable of distinguishing the ABR waveform is determined as a threshold value every 5dB step and is repeated for 2 times. Detection was performed in the order of 32kHz, 22.6kHz, 16kHz, 11.3kHz, 8kHz, 5.6kHz, 4 kHz.
5) After the detection, the mice are kept at the constant temperature of 37 ℃ until the mice are completely recovered.
Outer hair Cell Whole Cell Patch Clamp recordings (wheel Cell Patch Clamp Recording)
The temporal bone of the mouse was placed in extracellular fluid and the apical ring basement membrane was dissected out for further patch clamp experiments. All patch ranges correspond to hearing ranges between about 8kHz and 11 kHz. An AXON patch clamp system was used for the experiments, including an AXON200B patch clamp amplifier and a Digidata 1440B digital to analog converter. A glass microelectrode with an inner core (Cat #:1B150F-4, World Precision Instruments) was drawn to a tip impedance between 4-6M using a PC-100(Narige) glass microelectrode drawing machine. All patch clamp recordings and data analyses were performed using jClamp software. In recording the nonlinear membrane capacitance (NLC) of outer hair cells, the composition of the extracellular fluid is as follows: 136mM NaCl (Cat #: S7653, Sigma),1mM CaCl2(Cat #: C5080, Sigma),1mM MgCl2(Cat #: M2670, Sigma), and 10mM HEPES (Cat #: H3375, Sigma), pH was adjusted to 7.2 to 7.4 using NaOH (Cat #: S8045, Sigma) solution, and osmotic pressure was adjusted to 300mOsm using D-Glucose (Cat #: G8270, Sigma). The composition of the intracellular fluid was the same as that of the external fluid, but 10mM EGTA (Cat #: E4378, Sigma) was added. A10 mV continuous high-resolution bi-sinusoidal (390.6 Hz and 781.2Hz, respectively in frequency) stimulus was applied to a ramp voltage ranging from +150mV to-150 mV for 300 ms. The first derivative of the two-state Boltzmann function (two-state Boltzmann function) is used to fit the resulting data:
wherein
In the above formula, Qmax is the maximum value of nonlinear charge transfer, Vpkcm is the voltage value corresponding to the peak value of the membrane capacitance, and is also equal to half of the maximum charge transfer amount, Vm is the membrane potential, z is the ionic valence number, Clin is the linear membrane capacitance, e is the electronic capacity, k is the Boltzmann constant, and T is the absolute temperature value. The parameter reports were taken in the form of Mean ± SEM and were counted using one-way anova with GraphPad Prism 8.0 software (GraphPad, San Diego, US).
Example 1
Deletion of 13k bp fragment of Prestin intron 2 inhibits Prestin expression
The DNA sequence similarity of intron 2 between rat (. about.20 kbp) and mouse (. about.14.5 kbp) was 82.9%. Intron 2 of rat Prestin is considered to be critical for the specific expression of Prestin. The 14.5kb DNA fragment was first deleted by CRISPR/Cas9 method to determine its importance in mouse Prestin expression (FIGS. 1A-C). Two previously tested effective sgrnas (sgRNA-1 and sgRNA-4) were located at the 5 'and 3' ends of intron 2, respectively (fig. 1A). A mouse strain lacking a DNA fragment 6(13503bp, about 13kbp) between the sgRNA-1 and the sgRNA-4 was designated as a 13kb deletion mutant of Prestin intron 2 (Prestin-13 k/+). Both the Prestin-13k/+ heterozygote and the Prestin-13k/Prestin-13k homozygote mutant mice were healthy fertile mice. RNA in situ hybridization analysis showed that although Prestin was specifically detected in OHC (n ═ 3) of wild-type P6, Prestin was not detected in OHC of P6 Prestin-13k/Prestin-13k mutant mice (n ═ 3) (FIGS. 1D-E). Similarly, Prestin protein was clearly detected in all OHCs of wild type (n ═ 3), but Prestin was completely absent in OHCs of Prestin-13k/Prestin-13k mice (n ═ 3) at P5 (fig. 1F-G). Therefore, a 13 kbbp DNA fragment in intron 2 is considered to be necessary for the prompt initiation of Prestin.
It was next determined whether the transcription of intracochlear Prestin of heterozygous Prestin-13k/+ of P5 was affected, since previous results indicated that the Prestin mRNA in the heterozygous Prestin +/-was approximately half that in the wild type cochlea. However, the immunostaining and in situ RNA analysis described above did not show significant differences between wild-type and heterozygotes, partly because of their lower sensitivity. Thus, in three different models, a more sensitive qPCR analysis was applied to Tdtomato gene-tagged HCs (IHC and OHC): 1) atoh1-CreER +; rosa26-CAG-LSL-Tdtomato (Ai9)/+ (wild type HC); 2) atoh1-CreER +; ai 9/+; prestin-13k/+ (heterozygous mutant HC); 3) atoh1-CreER +; ai 9/+; prestin-13k/Prestin-13k (homozygous mutant HC). All mice were dosed with tamoxifen at P0 and P1. The cochlea was digested, Tdtomato + HC was manually picked, and RNA extraction and q-PCR analysis were performed at P5 (FIG. 1H). Three replicates were performed in each model, each replicate containing approximately 20 HCs.
q-PCR analysis showed that Prestin mRNA in heterozygote mutant HCs was statistically lower than that of wild-type HCs (FIG. 1I). As expected, the difference in Prestin was further significant between wild-type and homozygous mutant HCs (. about.. p <0.001) (fig. 1I). In contrast, no statistical differences were detected for Myo6 mRNA in HCs of the three genotypes (FIG. 1J). Note that qPCR analysis should underestimate the differences in Prestin mRNA in different models, since at present it is not technically guaranteed that all Tdtomato + cells are OHCs. Nonetheless, in agreement with previous reports, q-PCR data showed that deletion of 13kbp reduced expression of Prestin mRNA in a dose-dependent manner, but did not affect expression of other HC genes (e.g., Myo 6).
Example 2
Deletion of the near 5' 4k bp fragment of Prestin intron 2 repeats the phenotype of the Prestin-13k mutant
It was further determined which of the 13k bp fragments was necessary for Prestin expression. Using two additional sgRNA-2 and sgRNA-3, 13 kbbp previously tested for Prestin, were successfully divided into three approximately equal-length fractions (FIG. 2A). Through the combination between sgRNA-1 and 2 (# 4254bp), sgRNA-2 and 3 (# 5177bp), and sgRNA-3 and 4 (# 4080bp), three new mutant mice were generated, which were named Prestin-4k-1 (# 1 in FIG. 2A), Prestin-5k (#2 in FIG. 2A), and Prestin-4k-2 (# 3 in FIG. 2A), respectively. It was demonstrated that the mutant of Prestin-4k-1, but not the mutants of Prestin-5k and Prestin-4k-2, replicated the phenotype of the above mutant of Prestin-13 k. Thus, only the characteristics of the Prestin-4k-1 mutant are reported here (FIG. 2B). Both heterozygous Prestin-4k-1/+ and homozygous Prestin-4k-1/Prestin-4k-1 mutant mice were healthy and fertile and were easily identified by tail PCR (FIG. 2C).
Prestin is specifically expressed in wild-type OHC (FIG. 2D-D '), but not detected in OHC's of homozygous Prestin-4k-1/Prestin-4k-1 mutants (FIG. 2E-E '). And OHC of Prestin-4k-1/Prestin-4k-1 mice gradually restored Prestin expression at P14 and P60. The wild type, heterozygote and homozygote Prestin-4k-1 mutants, P14.5 and P60, were further subjected to NLC assay (FIGS. 2F and G). The Prestin-4k-1 mutant is similar to or overlaps with the OHC's corresponding to the Prestin-13k mutant. More importantly, no statistical differences were detected between Prestin-4k-1/+ and Prestin-13k/+ as well as between Prestin-4k-1/Prestin-4k-1 and Prestin-13k/Prestin 13k at P14.5 and P60 in terms of the number of Prestins (FIGS. 2H and I). In addition, there was no statistical difference in cell size and Prestin density. In conclusion, the loss of the 4k bp fragment near the 5' end of intron 2 has a very similar phenotype to the loss of the entire 13k bp fragment.
Example 3
Timely expression of Prestin requires a 1.4kb fragment of Intron 2
Next, it was planned to identify a CRE fragment smaller than 1.5 kbp in the 4 kbp range, which meets two criteria: 1) it is necessary for the timely expression of Prestin; 2) when bound to a mini-promoter, it can efficiently drive EGFP expression in OHCs, but not in IHCs or other cell types. 7 more potent sgrnas were pre-selected (# 5 to #11), which were distributed approximately every 500bp between sgRNA-1 and 2 (fig. 3A). Multiple sgRNAs and Cas9 may produce homozygous mutant F0 mice in single-cell fertilized eggs because of random DNA homozygous deletions between sgRNAs. Thus, these sgrnas are divided into two subgroups: 1) group 1 includes the 5 sgrnas on the left (sgrnas-1, 5, 6, 7, and 8); 2) group 2 includes the 5 sgrnas on the right (sgrnas-8, 9, 10, 11, and 2). Group 1 sgrnas (fig. 3B) or group 2 sgrnas (fig. 3C) were injected into single-cell stage zygotes along with Cas9 mRNA. Tail DNA PCR and Sanger sequencing were performed on 32 founder0(F0) mice from group 1. 12/32 mice clearly showed 7 different types of DNA deletions. Failure to generate clean PCR amplicons in other mice may be due to random insertion of large DNA fragments. More importantly, 1/32 mice appeared to be homozygous deletions of 1429bp (simply 1.4 kbbp) between sgRNA-1 and 7 (fig. 3D). Cochlear samples from this F0 mouse were immediately co-stained with Prestin and Myo6 (fig. 3E-F'). OHC's expressing higher Prestin were wild type genotypes (arrow in FIG. 3F-F'), while OHC's expressing lower or undetectable Prestin were homozygous deletion genotypes of 1.4 kbp (asterisks in FIG. 3F-F'). The chimeric expression pattern of Prestin showed that this 1.4kb chimeric homozygote deletion occurred in the cochlea, while the chimeric pattern of the tail tissue was much less.
In addition to the chimeric homozygous F0 mice analyzed above, other F0 mutant mice were also mated with wild type mice to reproduce germ cell stable mutant mice. Heterozygote-deficient mice from group 1 sgrnas were analyzed collectively: 522bp (Prestin-522/+) and 910bp (Prestin-910/+). The 522bp and 910bp fragments together occupy a 1.4 kbp region. Briefly, Prestin was absent from OHC's of homozygous Prestin-910/Prestin-910 mice of P5. No Prestin was detected in OHC from P5 mice homozygous for Prestin-522/Prestin-522. Of the 43F 0 mice derived from only group 2 sgrnas, only 2/43 mice clearly showed a hybrid DNA deletion of 2329bp and 2013bp, respectively. 2329bp deleted F0 mice were selected for germ cell passage. The 2329bp homology deletion had no effect on Prestin expression. Overall, deletion of 1.4 kbp or a portion (522bp or 910bp) inhibited Prestin expression in OHC of P5. Thus, 1.4 kbp between sgRNA-1 and sgRNA-7 was considered to be a key CRE element for Prestin expression. Whether the 1.4kb fragment is a potentially critical Prestin enhancer will be verified by transgenic mouse methods.
Example 4
The 1.4k fragment of mouse Presin and the 398bp fragment of human Prestin can specifically drive the expression of green fluorescent protein in cochlear OHC
By using the PiggyBac method, a transgenic mouse strain (abbreviated Prestin-1.4k-EGFP +) was constructed in which EGFP was driven by a mini-promoter from the mouse heat shock protein 68kDa (Hsp68) and a 1.4kb fragment (FIG. 4A). If 1.4 kbp is used as Prestin enhancer, then the expression of EGFP will be unique in OHC. A total of 21 positive transgenic mice were obtained. First, 4/21 mice were randomly selected for direct analysis at P6, P14, P21 and P30, respectively. EGFP was strongly and specifically expressed in cochlear OHCs of all 4F 0 mice, but not in all aged IHCs. In addition, the expression characteristics of EGFP in either chimeric (FIGS. 4B-B ") or homogeneous (FIGS. 4C-C") form were observed in F0 mice.
6/21 mice were randomly selected for germ cell stable passage, of which 4/6 mice produced stable Prestin-1.4k-EGFP + progeny. 2/6 mice failed to produce stable Prestin-1.4k-EGFP + progeny, probably due to the absence of transgene insertion into germ cells. EGFP was expressed in all cochlear OHC's, but not in IHC or vestibular HC's of P6-stabilized Prestin-1.4k-EGFP + mice, as expected. Specific EGFP expression in OHCs was maintained at P21. In general, the 1.4kb fragment is an enhancer of Prestin.
Meanwhile, the homologous sequence alignment of the mouse 1.4k bp fragment is carried out, and the result shows that the 398bp fragment can be found to be homologous with the mouse 1.4k bp fragment of Prestin in the human Prestin genome, and the total similarity is 77% (FIG. 4D and FIG. 5). Similarly, a transgenic mouse line was constructed in which expression of EGFP was driven by the human Prestin 398bp and Hsp68 mini-promoter (FIG. 4E). As expected, EGFP was specifically expressed in cochlear OHC of F0 mice (fig. 4F-F "). Thus, both mouse 1.4kb and human 398bp Prestin intron fragments were sufficient to drive the expression of specific EGFP in cochlear OHC.
Example 5
AAV produced by using mouse 1.4k bp sequence and human 398bp sequence specifically infects cochlear OHC
AAV viruses were next planned to be generated for specific transfection of OHCs. AAV2.7m8 was chosen as viral expression vector. Aav2.7m8, similar to Anc80L65, is a powerful viral vector capable of efficiently infecting a variety of cell types, including cochlear IHC, OHC and SC. To confer the ability of virus-specific transfection of OHCs, the original CAG promoter in aav2.7m8 was replaced with the mouse 1.4kb Prestin enhancer (fig. 6A) or human Prestin 398bp (fig. 6C) and Hsp68 mini-promoter which together controlled the expression pattern of reporter EGFP.
Aav2.7m8-1.4k (mouse) and aav2.7m8-398bp (human) viruses were injected into the cochlea of a neonatal mouse (P3) through the round window and analyzed 14 days later. It is exciting that EGFP was specifically highly expressed in cochlear OHC of P17 mice by injection of aav2.7m8-1.4k (fig. 6B-B ') or aav2.7m8-398bp virus (fig. 6D-D'). Thus, both aav2.7m8-1.4k and aav2.7m8-398bp viruses are specifically and efficiently targeted to OHCs.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.
Sequence listing
<110> China academy of sciences brain science and intelligent technology prominent innovation center
<120> cochlear outer hair cell specific cis-regulatory element and application thereof
<130> P2020-2885
<160> 13
<170> PatentIn version 3.5
<210> 1
<211> 1429
<212> DNA
<213> mouse (Mus musculus)
<400> 1
accacgtatg gagatttcta ttgtgcttgt attacctagg attctacctt aaaataaaag 60
cattagtttt aaagcaaata tttttagctc attggcttgg taacccagat atctctagtt 120
ttgaaaagtt ccttgaagaa agactagtgt tcggtcaaaa tagattacca agaactgacc 180
tgagaccctg tccttgtgaa cagaaacaaa gtcctcaaaa cttccctcat acacagccag 240
cagcagcccc acgctcacca aaggctcaca tgatgcacac gcatctctca gcagtggtcc 300
aggccgggct tcagtgtatc caggcacagg actgtgctgt gcctcctctc tactcccaca 360
atgctttgct gcgttctcag cttcccatct cctttgatgg gtgaaactct cgcctatctg 420
tgttgatcag ctgttctgaa aggccccagg agatgatagc atctcttctc tgggtatcca 480
tgccagtgtt taatagcctc ttgaaactgc cgagaagtgc tcagcaattg tgtggtgggg 540
aaggacggtg tcagcaagga ataaagatga tctctcgcct cgacggtagc tgaaatgtga 600
gtttctgtgt ccaggctcag ataaaaagtg atcctcacac ctttctcaag tagccagaga 660
cgattgaatt tctggaccag ggcaggaaaa caagagcatg ttgctaacct tgcttagtgc 720
tggggacctg atcttcttgc cattacattg tcatttctat cattctcatt cttgaaaaat 780
tatcctttga tttagagata acttatgaca atgtgactct ttatcttcat tttctttact 840
cctgtcttca tatcctaaat tacctataat atttaaaaaa aaagtgtaga tagtagactg 900
tcaggttagt acaagaatga aagacttgag ctgagaaaga aagaggtggt gagcgagtgg 960
caaggagcca tatttaattt cttcccccca cccccctttg gttcatcctc cagcttccca 1020
tcccacctgt attggtaaaa atgtacctgt gactcaggat ccttgtgttt tttgtggact 1080
tccatacagt gtgtagagca gcagctccat gggtgacctg gtgacctaga ttcacctgat 1140
ctgacccaga tggccaaccc cagggtctta gttaattctc gagaatgtct tgagctaggg 1200
gattctgaga ccagaaacct aagacccaca ggacctgttc tcctccaccc ttccagccac 1260
cattaaagac aactctaata ctggactcat ttgacaagct tattgctaat cataagatgt 1320
cacgcttttg agcataagta tacgaaagta aatgattctt taccaactgc ttacagtgaa 1380
tcaagagttg aacaaaggca ctagaaagat ggctttatca tcataactt 1429
<210> 2
<211> 398
<212> DNA
<213> Intelligent (Homo sapiens)
<400> 2
ggtgtaattc tgcctatcca ggatgatcag ctgttcctaa aggtccccag agacgataac 60
atctctgctc tgagtatcca tgccagtgtt taatagcctc ttgaaatttt ctccttgaag 120
tgatctgtga cttgttgagg tggggaagaa ggtgtcagca ggggataagc atgatcaact 180
tgatcgtagc tgaaatgtga actttagtgg ccaaactgag ataaaaaatg atcctcacac 240
cttctcaagt agttgaagac tattgagtta ccagaactgg gagaggagaa aaagagcaca 300
ttgtttacct taattatttc ttgggaccta atcatgttgc cattacattg taaattctgt 360
agattttcac acttggaaaa ttcttcttaa atttagag 398
<210> 3
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 3
<210> 4
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 4
<210> 5
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 5
<210> 6
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 6
<210> 7
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 7
agaagtgctc agcaattgtg 20
<210> 8
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 8
taaaaatgta cctgtgactc 20
<210> 9
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 9
ctttatcatc ataactttat 20
<210> 10
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 10
<210> 11
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 11
<210> 12
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 12
<210> 13
<211> 20
<212> DNA
<213> Artificial sequence (Artificial sequence)
<400> 13
Claims (10)
1. Use of a combination of active principles for the preparation of a formulation or medicament for: (i) repair and/or regeneration of extra-cochlear hair cells (OHC); (ii) treating or preventing hearing impairment; and/or (iii) specific expression of effector genes in cochlear outer hair cells;
wherein the active ingredient combination comprises:
(a) a cochlear outer hair cell-specific cis-regulatory element;
(b) a promoter; and
(c) an effector gene or a coding sequence thereof.
2. The use according to claim 1, wherein the hearing impairment is hearing impairment associated with degeneration and/or damage of extracochlear hair cells.
3. The use according to claim 1, wherein the extracochlear hair cell-specific cis regulatory element has a nucleotide sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 2, and the effector gene is a gene for coding Prestin protein.
4. An active ingredient combination, characterized in that said combination comprises:
(a) a cochlear outer hair cell-specific cis-regulatory element;
(b) a promoter; and
(c) an effector gene or coding sequence thereof;
wherein, the nucleotide sequence of the cochlea outer hair cell specificity cis-form regulatory element is shown as SEQ ID NO: 1 or SEQ ID NO: 2, respectively.
5. An expression vector comprising an expression cassette, wherein said expression cassette has the structure of formula I from 5 'to 3':
Z0-Z1-Z2-Z3-Z0’ (I)
in the formula (I), the compound is shown in the specification,
each "-" is independently a chemical bond or a nucleotide linking sequence;
z0 is none, or a 5' UTR sequence;
z1 is the nucleotide sequence of the cis-regulatory element of the specificity of the cochlear outer hair cell;
z2 is a promoter sequence;
z3 is the nucleotide sequence of an effector gene; and
z0 'is a null, or 3' UTR sequence.
6. A host cell comprising the expression vector of claim 5.
7. A pharmaceutical formulation, comprising: (a) the active ingredient combination according to claim 4, or the expression vector according to claim 5, or the host cell according to claim 6, and (b) a pharmaceutically acceptable carrier or excipient.
8. Use of an active ingredient combination according to claim 4, an expression vector according to claim 5, a host cell according to claim 6, or a pharmaceutical preparation according to claim 7 for the preparation of a medicament for the treatment or prevention of hearing impairment.
9. A method of repairing and/or regenerating outer hair cells of the cochlea comprising transducing the active ingredient combination of claim 4, or the expression vector of claim 5, into denatured or damaged outer hair cells of the cochlea.
10. Use of an extracochlear hair cell-specific cis regulatory element, wherein the cis regulatory element comprises the amino acid sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 2 for driving the expression of effector genes specifically in cochlear outer hair cells.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202110055458.7A CN114762733A (en) | 2021-01-15 | 2021-01-15 | Extracochlear hair cell specific cis-form regulatory element and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN202110055458.7A CN114762733A (en) | 2021-01-15 | 2021-01-15 | Extracochlear hair cell specific cis-form regulatory element and application thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
CN114762733A true CN114762733A (en) | 2022-07-19 |
Family
ID=82363816
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN202110055458.7A Pending CN114762733A (en) | 2021-01-15 | 2021-01-15 | Extracochlear hair cell specific cis-form regulatory element and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN114762733A (en) |
Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104042616A (en) * | 2014-02-20 | 2014-09-17 | 复旦大学附属眼耳鼻喉科医院 | Use of lysine-specific demethylase 1 (LSD1) inhibitor |
WO2018204226A1 (en) * | 2017-05-03 | 2018-11-08 | St. Jude Children's Research Hospital | Compositions and methods for prevention and treatment of hearing loss |
CN111698975A (en) * | 2017-02-06 | 2020-09-22 | 儿童医疗中心有限公司 | Materials and methods for delivery of nucleic acids to cochlear and vestibular cells |
-
2021
- 2021-01-15 CN CN202110055458.7A patent/CN114762733A/en active Pending
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104042616A (en) * | 2014-02-20 | 2014-09-17 | 复旦大学附属眼耳鼻喉科医院 | Use of lysine-specific demethylase 1 (LSD1) inhibitor |
CN111698975A (en) * | 2017-02-06 | 2020-09-22 | 儿童医疗中心有限公司 | Materials and methods for delivery of nucleic acids to cochlear and vestibular cells |
WO2018204226A1 (en) * | 2017-05-03 | 2018-11-08 | St. Jude Children's Research Hospital | Compositions and methods for prevention and treatment of hearing loss |
Non-Patent Citations (1)
Title |
---|
于利;汤浩;: "哺乳动物耳蜗外毛细胞的马达蛋白:Prestin", 生理科学进展, no. 01, 25 January 2008 (2008-01-25), pages 55 - 58 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN117821509A (en) | Tissue-selective transgene expression | |
Xue et al. | Gene editing in a Myo6 semi-dominant mouse model rescues auditory function | |
CA2375106C (en) | Compositions and methods for the therapeutic use of an atonal-associated sequence for deafness, osteoarthritis, and abnormal cell proliferation | |
CN111683688A (en) | Gene therapy treatment of hearing loss | |
JP2020509786A (en) | Gene therapy compositions and methods for the treatment of hearing loss | |
US11459584B2 (en) | Gene sequence of recombinant human type II mitochondrial dynein-like GTPase and uses thereof | |
US20210330814A1 (en) | Methods of treating non-syndromic sensorineural hearing loss | |
CN115029360B (en) | Transgenic expression cassettes of type IIIA for the treatment of mucopolysaccharidosis | |
Fiore et al. | Apparent normal phenotype of Fgf6-/-mice. | |
JP2024100980A (en) | In vivo homology-directed repair in heart, skeletal muscle, and muscle stem cells | |
US20220112504A1 (en) | Methods and compositions for allele specific gene editing | |
Qi et al. | Preclinical Efficacy And Safety Evaluation of AAV‐OTOF in DFNB9 Mouse Model And Nonhuman Primate | |
Peters et al. | Rescue of hearing by adenine base editing in a humanized mouse model of Usher syndrome type 1F | |
CN110628814A (en) | Method for increasing SMN protein expression based on gene editing technology and application of method in SMA treatment | |
US20220000972A1 (en) | A method for treating an auditory neuropathy spectrum disorder | |
WO2023143366A1 (en) | Variant adeno-associated virus and application thereof in disease treatment | |
CN114369600B (en) | For repairing Klhl18 lowf CRISPR/Cas9 gene editing system of mutant gene and application | |
CN114762733A (en) | Extracochlear hair cell specific cis-form regulatory element and application thereof | |
CN113444722A (en) | Application of single base editing mediated splicing repair in preparation of drugs for treating spinal muscular atrophy | |
JP2024518552A (en) | Gene therapy constructs and methods for treating hearing loss - Patents.com | |
TWI856028B (en) | A method for treating an auditory neuropathy spectrum disorder | |
WO2022147891A1 (en) | Cochlear outer hair cell regenerated by ectopic joint overexpression of atoh1 and ikzf2 and application thereof | |
Manfredsson et al. | 59. rAAV-Mediated Nigral Parkin Over-Expression Is Neuroprotective in the 6-OHDA Rat Model of Parkinson's Disease | |
Koya et al. | GENE THERAPY FOR INFECTIONS AND VACCINES | |
LLADO SANTAEULARIA | THERAPEUTIC GENOME EDITING IN RETINA AND LIVER |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PB01 | Publication | ||
PB01 | Publication | ||
SE01 | Entry into force of request for substantive examination | ||
SE01 | Entry into force of request for substantive examination |