NZ765592A - Methods and compositions for targeted genetic modifications and methods of use - Google Patents
Methods and compositions for targeted genetic modifications and methods of use Download PDFInfo
- Publication number
- NZ765592A NZ765592A NZ765592A NZ76559215A NZ765592A NZ 765592 A NZ765592 A NZ 765592A NZ 765592 A NZ765592 A NZ 765592A NZ 76559215 A NZ76559215 A NZ 76559215A NZ 765592 A NZ765592 A NZ 765592A
- Authority
- NZ
- New Zealand
- Prior art keywords
- cell
- mouse
- sequence
- cells
- target
- Prior art date
Links
- 238000000034 method Methods 0.000 title claims abstract description 273
- 238000012239 gene modification Methods 0.000 title claims abstract description 124
- 230000005017 genetic modification Effects 0.000 title claims abstract description 122
- 235000013617 genetically modified food Nutrition 0.000 title claims abstract description 122
- 239000000203 mixture Substances 0.000 title claims abstract description 52
- 210000004027 cell Anatomy 0.000 claims abstract description 846
- 101150059736 SRY gene Proteins 0.000 claims abstract description 223
- 241000282414 Homo sapiens Species 0.000 claims abstract description 217
- 210000002593 Y chromosome Anatomy 0.000 claims abstract description 138
- 230000004048 modification Effects 0.000 claims abstract description 77
- 238000012986 modification Methods 0.000 claims abstract description 77
- 238000012258 culturing Methods 0.000 claims abstract description 27
- 102000040430 polynucleotide Human genes 0.000 claims description 215
- 108091033319 polynucleotide Proteins 0.000 claims description 215
- 239000002157 polynucleotide Substances 0.000 claims description 215
- 150000007523 nucleic acids Chemical group 0.000 claims description 164
- 108090000623 proteins and genes Proteins 0.000 claims description 158
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 claims description 155
- 101710163270 Nuclease Proteins 0.000 claims description 149
- 241000699666 Mus <mouse, genus> Species 0.000 claims description 147
- 230000008685 targeting Effects 0.000 claims description 131
- 239000003795 chemical substances by application Substances 0.000 claims description 121
- 108020005004 Guide RNA Proteins 0.000 claims description 118
- 102000004169 proteins and genes Human genes 0.000 claims description 108
- 210000001161 mammalian embryo Anatomy 0.000 claims description 106
- 239000013598 vector Substances 0.000 claims description 104
- 238000012217 deletion Methods 0.000 claims description 96
- 230000037430 deletion Effects 0.000 claims description 96
- 102000039446 nucleic acids Human genes 0.000 claims description 92
- 108020004707 nucleic acids Proteins 0.000 claims description 92
- 239000002773 nucleotide Substances 0.000 claims description 92
- 125000003729 nucleotide group Chemical group 0.000 claims description 87
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 80
- 239000011780 sodium chloride Substances 0.000 claims description 77
- 108700010045 sry Genes Proteins 0.000 claims description 56
- 108091079001 CRISPR RNA Proteins 0.000 claims description 48
- 238000003780 insertion Methods 0.000 claims description 46
- 230000037431 insertion Effects 0.000 claims description 46
- 108091033409 CRISPR Proteins 0.000 claims description 45
- 238000010459 TALEN Methods 0.000 claims description 34
- 230000005782 double-strand break Effects 0.000 claims description 32
- 239000003550 marker Substances 0.000 claims description 25
- 239000013589 supplement Substances 0.000 claims description 25
- 108700008625 Reporter Genes Proteins 0.000 claims description 23
- 241000699670 Mus sp. Species 0.000 claims description 21
- 150000002500 ions Chemical class 0.000 claims description 17
- 230000001568 sexual effect Effects 0.000 claims description 17
- 238000006467 substitution reaction Methods 0.000 claims description 17
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 claims description 16
- 238000011740 C57BL/6 mouse Methods 0.000 claims description 12
- 230000035935 pregnancy Effects 0.000 claims description 10
- 229910000030 sodium bicarbonate Inorganic materials 0.000 claims description 9
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical group [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 claims description 8
- 235000017557 sodium bicarbonate Nutrition 0.000 claims description 8
- 229910052725 zinc Inorganic materials 0.000 claims description 8
- 239000011701 zinc Substances 0.000 claims description 8
- 101100478386 Mus musculus Sry gene Proteins 0.000 claims description 4
- 230000001135 feminizing effect Effects 0.000 claims description 4
- 101100042648 Drosophila melanogaster sing gene Proteins 0.000 claims 2
- 102100029054 Homeobox protein notochord Human genes 0.000 claims 2
- 101000634521 Homo sapiens Homeobox protein notochord Proteins 0.000 claims 2
- 108091028113 Trans-activating crRNA Proteins 0.000 claims 1
- 241000112708 Vates Species 0.000 claims 1
- 230000003213 activating effect Effects 0.000 claims 1
- 230000000694 effects Effects 0.000 abstract description 85
- 238000000338 in vitro Methods 0.000 abstract description 34
- 230000007423 decrease Effects 0.000 abstract description 28
- 238000011161 development Methods 0.000 abstract description 16
- 210000004102 animal cell Anatomy 0.000 abstract description 6
- 239000002609 medium Substances 0.000 description 187
- 235000018102 proteins Nutrition 0.000 description 104
- 108020004414 DNA Proteins 0.000 description 91
- 241000700159 Rattus Species 0.000 description 77
- 241001465754 Metazoa Species 0.000 description 61
- 241000283984 Rodentia Species 0.000 description 58
- 230000014509 gene expression Effects 0.000 description 53
- 210000004962 mammalian cell Anatomy 0.000 description 51
- 230000035772 mutation Effects 0.000 description 49
- 108700028369 Alleles Proteins 0.000 description 44
- 241000124008 Mammalia Species 0.000 description 36
- 241000699800 Cricetinae Species 0.000 description 32
- 230000000295 complement effect Effects 0.000 description 32
- 150000003839 salts Chemical class 0.000 description 31
- BVKZGUZCCUSVTD-UHFFFAOYSA-L Carbonate Chemical compound [O-]C([O-])=O BVKZGUZCCUSVTD-UHFFFAOYSA-L 0.000 description 30
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 30
- 230000001105 regulatory effect Effects 0.000 description 27
- 108090000765 processed proteins & peptides Proteins 0.000 description 26
- 102000004196 processed proteins & peptides Human genes 0.000 description 25
- 238000003776 cleavage reaction Methods 0.000 description 24
- 229920001184 polypeptide Polymers 0.000 description 24
- 230000007017 scission Effects 0.000 description 24
- 239000002253 acid Substances 0.000 description 23
- 210000001236 prokaryotic cell Anatomy 0.000 description 22
- 108010017070 Zinc Finger Nucleases Proteins 0.000 description 21
- 230000027455 binding Effects 0.000 description 20
- 238000003556 assay Methods 0.000 description 19
- 125000005588 carbonic acid salt group Chemical group 0.000 description 19
- 150000001768 cations Chemical class 0.000 description 19
- 210000002257 embryonic structure Anatomy 0.000 description 19
- 229910052751 metal Inorganic materials 0.000 description 19
- 239000002184 metal Substances 0.000 description 19
- 230000003247 decreasing effect Effects 0.000 description 18
- 150000004820 halides Chemical class 0.000 description 18
- 210000000287 oocyte Anatomy 0.000 description 18
- 108091026890 Coding region Proteins 0.000 description 17
- 150000001413 amino acids Chemical class 0.000 description 16
- 210000005260 human cell Anatomy 0.000 description 16
- 235000001014 amino acid Nutrition 0.000 description 15
- 230000018109 developmental process Effects 0.000 description 15
- 239000012634 fragment Substances 0.000 description 15
- 238000009396 hybridization Methods 0.000 description 15
- 230000001939 inductive effect Effects 0.000 description 14
- 230000006798 recombination Effects 0.000 description 14
- 238000005215 recombination Methods 0.000 description 14
- 238000011144 upstream manufacturing Methods 0.000 description 14
- 230000004568 DNA-binding Effects 0.000 description 13
- 108010008532 Deoxyribonuclease I Proteins 0.000 description 13
- 102000007260 Deoxyribonuclease I Human genes 0.000 description 13
- 230000002068 genetic effect Effects 0.000 description 13
- 239000001963 growth medium Substances 0.000 description 13
- 238000002744 homologous recombination Methods 0.000 description 13
- 230000006801 homologous recombination Effects 0.000 description 13
- 210000001519 tissue Anatomy 0.000 description 13
- -1 trachNAs Proteins 0.000 description 13
- 241000282693 Cercopithecidae Species 0.000 description 12
- 229940024606 amino acid Drugs 0.000 description 12
- 230000000670 limiting effect Effects 0.000 description 12
- 230000002829 reductive effect Effects 0.000 description 12
- 239000000523 sample Substances 0.000 description 12
- 241000894007 species Species 0.000 description 12
- 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 11
- 239000008103 glucose Substances 0.000 description 11
- 238000012546 transfer Methods 0.000 description 11
- 238000010453 CRISPR/Cas method Methods 0.000 description 10
- 210000004436 artificial bacterial chromosome Anatomy 0.000 description 10
- 210000000349 chromosome Anatomy 0.000 description 10
- 108020004999 messenger RNA Proteins 0.000 description 10
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 10
- 230000009261 transgenic effect Effects 0.000 description 10
- 230000002759 chromosomal effect Effects 0.000 description 9
- 230000004927 fusion Effects 0.000 description 9
- 229910001507 metal halide Inorganic materials 0.000 description 9
- 210000000472 morula Anatomy 0.000 description 9
- 231100000350 mutagenesis Toxicity 0.000 description 9
- 108020004705 Codon Proteins 0.000 description 8
- 241001494479 Pecora Species 0.000 description 8
- 108010052160 Site-specific recombinase Proteins 0.000 description 8
- 108010073062 Transcription Activator-Like Effectors Proteins 0.000 description 8
- 230000010354 integration Effects 0.000 description 8
- 238000012216 screening Methods 0.000 description 8
- 210000000130 stem cell Anatomy 0.000 description 8
- 238000013518 transcription Methods 0.000 description 8
- 230000035897 transcription Effects 0.000 description 8
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 8
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 7
- 102000040650 (ribonucleotides)n+m Human genes 0.000 description 7
- 241000283690 Bos taurus Species 0.000 description 7
- 102000053602 DNA Human genes 0.000 description 7
- 241000282326 Felis catus Species 0.000 description 7
- 241000193996 Streptococcus pyogenes Species 0.000 description 7
- 238000007792 addition Methods 0.000 description 7
- 230000004075 alteration Effects 0.000 description 7
- 230000001580 bacterial effect Effects 0.000 description 7
- 238000009395 breeding Methods 0.000 description 7
- 230000001488 breeding effect Effects 0.000 description 7
- BVKZGUZCCUSVTD-UHFFFAOYSA-N carbonic acid Chemical compound OC(O)=O BVKZGUZCCUSVTD-UHFFFAOYSA-N 0.000 description 7
- 230000006870 function Effects 0.000 description 7
- 101150066555 lacZ gene Proteins 0.000 description 7
- 230000001404 mediated effect Effects 0.000 description 7
- 210000004940 nucleus Anatomy 0.000 description 7
- 230000005783 single-strand break Effects 0.000 description 7
- 230000002103 transcriptional effect Effects 0.000 description 7
- 241000283707 Capra Species 0.000 description 6
- 241000282994 Cervidae Species 0.000 description 6
- 102000004190 Enzymes Human genes 0.000 description 6
- 108090000790 Enzymes Proteins 0.000 description 6
- 241000287828 Gallus gallus Species 0.000 description 6
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 6
- 102000004058 Leukemia inhibitory factor Human genes 0.000 description 6
- 108090000581 Leukemia inhibitory factor Proteins 0.000 description 6
- 108700026244 Open Reading Frames Proteins 0.000 description 6
- 241000009328 Perro Species 0.000 description 6
- 108700029634 Y-Linked Genes Proteins 0.000 description 6
- 238000004458 analytical method Methods 0.000 description 6
- 230000000692 anti-sense effect Effects 0.000 description 6
- 210000001106 artificial yeast chromosome Anatomy 0.000 description 6
- 235000013330 chicken meat Nutrition 0.000 description 6
- 230000001965 increasing effect Effects 0.000 description 6
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 6
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 6
- 239000000126 substance Substances 0.000 description 6
- 241000271566 Aves Species 0.000 description 5
- 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 5
- 241000282560 Macaca mulatta Species 0.000 description 5
- 241000282341 Mustela putorius furo Species 0.000 description 5
- 108010021466 Mutant Proteins Proteins 0.000 description 5
- 102000008300 Mutant Proteins Human genes 0.000 description 5
- 102000018120 Recombinases Human genes 0.000 description 5
- 108010091086 Recombinases Proteins 0.000 description 5
- 102100022978 Sex-determining region Y protein Human genes 0.000 description 5
- 150000007513 acids Chemical class 0.000 description 5
- 230000035558 fertility Effects 0.000 description 5
- 210000002950 fibroblast Anatomy 0.000 description 5
- 108010021843 fluorescent protein 583 Proteins 0.000 description 5
- UYTPUPDQBNUYGX-UHFFFAOYSA-N guanine Chemical compound O=C1NC(N)=NC2=C1N=CN2 UYTPUPDQBNUYGX-UHFFFAOYSA-N 0.000 description 5
- 238000000520 microinjection Methods 0.000 description 5
- 239000000178 monomer Substances 0.000 description 5
- 239000013612 plasmid Substances 0.000 description 5
- 239000011734 sodium Substances 0.000 description 5
- 229910052708 sodium Inorganic materials 0.000 description 5
- 229940054269 sodium pyruvate Drugs 0.000 description 5
- 239000000758 substrate Substances 0.000 description 5
- 238000001890 transfection Methods 0.000 description 5
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 4
- 101150077884 Eif2s3y gene Proteins 0.000 description 4
- 102000004533 Endonucleases Human genes 0.000 description 4
- 108010042407 Endonucleases Proteins 0.000 description 4
- 108010034791 Heterochromatin Proteins 0.000 description 4
- 108010025815 Kanamycin Kinase Proteins 0.000 description 4
- CSNNHWWHGAXBCP-UHFFFAOYSA-L Magnesium sulfate Chemical compound [Mg+2].[O-][S+2]([O-])([O-])[O-] CSNNHWWHGAXBCP-UHFFFAOYSA-L 0.000 description 4
- 229930193140 Neomycin Natural products 0.000 description 4
- 108010077850 Nuclear Localization Signals Proteins 0.000 description 4
- 241000283973 Oryctolagus cuniculus Species 0.000 description 4
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 4
- 241000288906 Primates Species 0.000 description 4
- 241000282887 Suidae Species 0.000 description 4
- 108010043645 Transcription Activator-Like Effector Nucleases Proteins 0.000 description 4
- 108700019146 Transgenes Proteins 0.000 description 4
- 241000700605 Viruses Species 0.000 description 4
- 210000001766 X chromosome Anatomy 0.000 description 4
- 102000005936 beta-Galactosidase Human genes 0.000 description 4
- 108010005774 beta-Galactosidase Proteins 0.000 description 4
- 230000015572 biosynthetic process Effects 0.000 description 4
- 210000002459 blastocyst Anatomy 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 210000001671 embryonic stem cell Anatomy 0.000 description 4
- 239000012091 fetal bovine serum Substances 0.000 description 4
- 108091006047 fluorescent proteins Proteins 0.000 description 4
- 102000034287 fluorescent proteins Human genes 0.000 description 4
- 210000004602 germ cell Anatomy 0.000 description 4
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 4
- 210000004458 heterochromatin Anatomy 0.000 description 4
- 238000004519 manufacturing process Methods 0.000 description 4
- 241001515942 marmosets Species 0.000 description 4
- 229960004927 neomycin Drugs 0.000 description 4
- 230000008488 polyadenylation Effects 0.000 description 4
- 229920000642 polymer Polymers 0.000 description 4
- 229910052594 sapphire Inorganic materials 0.000 description 4
- 239000010980 sapphire Substances 0.000 description 4
- 238000012163 sequencing technique Methods 0.000 description 4
- 239000000243 solution Substances 0.000 description 4
- 108091005957 yellow fluorescent proteins Proteins 0.000 description 4
- 210000004340 zona pellucida Anatomy 0.000 description 4
- YMHOBZXQZVXHBM-UHFFFAOYSA-N 2,5-dimethoxy-4-bromophenethylamine Chemical compound COC1=CC(CCN)=C(OC)C=C1Br YMHOBZXQZVXHBM-UHFFFAOYSA-N 0.000 description 3
- 108091005944 Cerulean Proteins 0.000 description 3
- 241000579895 Chlorostilbon Species 0.000 description 3
- 108010077544 Chromatin Proteins 0.000 description 3
- 108091005943 CyPet Proteins 0.000 description 3
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 3
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 3
- 238000009015 Human TaqMan MicroRNA Assay kit Methods 0.000 description 3
- 241000699660 Mus musculus Species 0.000 description 3
- 229930182555 Penicillin Natural products 0.000 description 3
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M Potassium chloride Chemical compound [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 3
- 101710188553 Sex-determining region Y protein Proteins 0.000 description 3
- 108091081024 Start codon Proteins 0.000 description 3
- 241000545067 Venus Species 0.000 description 3
- 235000004279 alanine Nutrition 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 230000001413 cellular effect Effects 0.000 description 3
- 210000003483 chromatin Anatomy 0.000 description 3
- 238000013461 design Methods 0.000 description 3
- 201000010099 disease Diseases 0.000 description 3
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 3
- 230000011559 double-strand break repair via nonhomologous end joining Effects 0.000 description 3
- 239000010976 emerald Substances 0.000 description 3
- 229910052876 emerald Inorganic materials 0.000 description 3
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 3
- 239000013613 expression plasmid Substances 0.000 description 3
- 108020001507 fusion proteins Proteins 0.000 description 3
- 102000037865 fusion proteins Human genes 0.000 description 3
- 238000010363 gene targeting Methods 0.000 description 3
- 238000003205 genotyping method Methods 0.000 description 3
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 3
- 210000000688 human artificial chromosome Anatomy 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 230000008948 male sex determination Effects 0.000 description 3
- 210000003470 mitochondria Anatomy 0.000 description 3
- 238000002703 mutagenesis Methods 0.000 description 3
- 230000036961 partial effect Effects 0.000 description 3
- 229940049954 penicillin Drugs 0.000 description 3
- 229950010131 puromycin Drugs 0.000 description 3
- 238000012207 quantitative assay Methods 0.000 description 3
- 238000003753 real-time PCR Methods 0.000 description 3
- 108091008146 restriction endonucleases Proteins 0.000 description 3
- 230000008771 sex reversal Effects 0.000 description 3
- 229960005322 streptomycin Drugs 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 2
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 2
- LRFVTYWOQMYALW-UHFFFAOYSA-N 9H-xanthine Chemical compound O=C1NC(=O)NC2=C1NC=N2 LRFVTYWOQMYALW-UHFFFAOYSA-N 0.000 description 2
- 241000251468 Actinopterygii Species 0.000 description 2
- 102000006822 Agouti Signaling Protein Human genes 0.000 description 2
- 108010072151 Agouti Signaling Protein Proteins 0.000 description 2
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 2
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 2
- 241000272517 Anseriformes Species 0.000 description 2
- 101710201279 Biotin carboxyl carrier protein Proteins 0.000 description 2
- 108700010070 Codon Usage Proteins 0.000 description 2
- 241000484025 Cuniculus Species 0.000 description 2
- 238000001712 DNA sequencing Methods 0.000 description 2
- 102000016911 Deoxyribonucleases Human genes 0.000 description 2
- 108010053770 Deoxyribonucleases Proteins 0.000 description 2
- 241000283073 Equus caballus Species 0.000 description 2
- 108010022894 Euchromatin Proteins 0.000 description 2
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 2
- 102000005720 Glutathione transferase Human genes 0.000 description 2
- 108010070675 Glutathione transferase Proteins 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 101710154606 Hemagglutinin Proteins 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 2
- 229930182816 L-glutamine Natural products 0.000 description 2
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 2
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 2
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 2
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 241001529936 Murinae Species 0.000 description 2
- 241000711408 Murine respirovirus Species 0.000 description 2
- 101710093908 Outer capsid protein VP4 Proteins 0.000 description 2
- 101710135467 Outer capsid protein sigma-1 Proteins 0.000 description 2
- 101710176177 Protein A56 Proteins 0.000 description 2
- AUNGANRZJHBGPY-SCRDCRAPSA-N Riboflavin Chemical compound OC[C@@H](O)[C@@H](O)[C@@H](O)CN1C=2C=C(C)C(C)=CC=2N=C2C1=NC(=O)NC2=O AUNGANRZJHBGPY-SCRDCRAPSA-N 0.000 description 2
- 108091028664 Ribonucleotide Proteins 0.000 description 2
- 108700032475 Sex-Determining Region Y Proteins 0.000 description 2
- 241000700584 Simplexvirus Species 0.000 description 2
- 238000002105 Southern blotting Methods 0.000 description 2
- 241000194020 Streptococcus thermophilus Species 0.000 description 2
- 241000203587 Streptosporangium roseum Species 0.000 description 2
- 102000002933 Thioredoxin Human genes 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine kinase Proteins 0.000 description 2
- 108091023040 Transcription factor Proteins 0.000 description 2
- 102000040945 Transcription factor Human genes 0.000 description 2
- 108090000848 Ubiquitin Proteins 0.000 description 2
- 102000044159 Ubiquitin Human genes 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 210000004504 adult stem cell Anatomy 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- 230000003115 biocidal effect Effects 0.000 description 2
- 230000004071 biological effect Effects 0.000 description 2
- 244000309464 bull Species 0.000 description 2
- 101150016727 ch25h gene Proteins 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 102000021178 chitin binding proteins Human genes 0.000 description 2
- 108091011157 chitin binding proteins Proteins 0.000 description 2
- 210000003763 chloroplast Anatomy 0.000 description 2
- 239000002299 complementary DNA Substances 0.000 description 2
- 239000003636 conditioned culture medium Substances 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 238000007796 conventional method Methods 0.000 description 2
- 238000001514 detection method Methods 0.000 description 2
- 238000002224 dissection Methods 0.000 description 2
- 239000003814 drug Substances 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000012636 effector Substances 0.000 description 2
- 108010026638 endodeoxyribonuclease FokI Proteins 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 239000003623 enhancer Substances 0.000 description 2
- 230000007159 enucleation Effects 0.000 description 2
- 210000000632 euchromatin Anatomy 0.000 description 2
- 210000003527 eukaryotic cell Anatomy 0.000 description 2
- 238000002474 experimental method Methods 0.000 description 2
- OVBPIULPVIDEAO-LBPRGKRZSA-N folic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 2
- 210000004392 genitalia Anatomy 0.000 description 2
- 239000000185 hemagglutinin Substances 0.000 description 2
- 229910052739 hydrogen Inorganic materials 0.000 description 2
- 239000001257 hydrogen Substances 0.000 description 2
- 230000000415 inactivating effect Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- VCJMYUPGQJHHFU-UHFFFAOYSA-N iron(3+);trinitrate Chemical compound [Fe+3].[O-][N+]([O-])=O.[O-][N+]([O-])=O.[O-][N+]([O-])=O VCJMYUPGQJHHFU-UHFFFAOYSA-N 0.000 description 2
- 229940043355 kinase inhibitor Drugs 0.000 description 2
- 230000004807 localization Effects 0.000 description 2
- 229910052943 magnesium sulfate Inorganic materials 0.000 description 2
- 235000019341 magnesium sulphate Nutrition 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 238000002844 melting Methods 0.000 description 2
- 230000008018 melting Effects 0.000 description 2
- 238000010369 molecular cloning Methods 0.000 description 2
- 238000005457 optimization Methods 0.000 description 2
- 210000001672 ovary Anatomy 0.000 description 2
- 210000003101 oviduct Anatomy 0.000 description 2
- 230000035515 penetration Effects 0.000 description 2
- 230000026731 phosphorylation Effects 0.000 description 2
- 238000006366 phosphorylation reaction Methods 0.000 description 2
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 108010045647 puromycin N-acetyltransferase Proteins 0.000 description 2
- LXNHXLLTXMVWPM-UHFFFAOYSA-N pyridoxine Chemical compound CC1=NC=C(CO)C(CO)=C1O LXNHXLLTXMVWPM-UHFFFAOYSA-N 0.000 description 2
- 108010054624 red fluorescent protein Proteins 0.000 description 2
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 2
- 230000008439 repair process Effects 0.000 description 2
- 238000011160 research Methods 0.000 description 2
- 239000002336 ribonucleotide Substances 0.000 description 2
- 125000002652 ribonucleotide group Chemical group 0.000 description 2
- 230000020509 sex determination Effects 0.000 description 2
- 230000035939 shock Effects 0.000 description 2
- 159000000000 sodium salts Chemical class 0.000 description 2
- 210000001082 somatic cell Anatomy 0.000 description 2
- 125000006850 spacer group Chemical group 0.000 description 2
- 230000002269 spontaneous effect Effects 0.000 description 2
- 238000012453 sprague-dawley rat model Methods 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 238000010381 tandem affinity purification Methods 0.000 description 2
- 238000012360 testing method Methods 0.000 description 2
- 210000001550 testis Anatomy 0.000 description 2
- 108060008226 thioredoxin Proteins 0.000 description 2
- 229940094937 thioredoxin Drugs 0.000 description 2
- 108091006107 transcriptional repressors Proteins 0.000 description 2
- 238000003151 transfection method Methods 0.000 description 2
- 210000005253 yeast cell Anatomy 0.000 description 2
- XDIYNQZUNSSENW-UUBOPVPUSA-N (2R,3S,4R,5R)-2,3,4,5,6-pentahydroxyhexanal Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C=O.OC[C@@H](O)[C@@H](O)[C@H](O)[C@@H](O)C=O XDIYNQZUNSSENW-UUBOPVPUSA-N 0.000 description 1
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 1
- WEEMDRWIKYCTQM-UHFFFAOYSA-N 2,6-dimethoxybenzenecarbothioamide Chemical compound COC1=CC=CC(OC)=C1C(N)=S WEEMDRWIKYCTQM-UHFFFAOYSA-N 0.000 description 1
- KQPKMEYBZUPZGK-UHFFFAOYSA-N 4-[(4-azido-2-nitroanilino)methyl]-5-(hydroxymethyl)-2-methylpyridin-3-ol Chemical compound CC1=NC=C(CO)C(CNC=2C(=CC(=CC=2)N=[N+]=[N-])[N+]([O-])=O)=C1O KQPKMEYBZUPZGK-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- 208000026817 47,XYY syndrome Diseases 0.000 description 1
- 102100033392 ATP-dependent RNA helicase DDX3Y Human genes 0.000 description 1
- 208000037068 Abnormal Karyotype Diseases 0.000 description 1
- 241000007910 Acaryochloris marina Species 0.000 description 1
- 241001135192 Acetohalobium arabaticum Species 0.000 description 1
- 241001464929 Acidithiobacillus caldus Species 0.000 description 1
- 241000605222 Acidithiobacillus ferrooxidans Species 0.000 description 1
- 241000699718 Akodon Species 0.000 description 1
- 102100036475 Alanine aminotransferase 1 Human genes 0.000 description 1
- 244000003363 Allium ursinum Species 0.000 description 1
- 241000190857 Allochromatium vinosum Species 0.000 description 1
- 241000147155 Ammonifex degensii Species 0.000 description 1
- 101100004864 Arabidopsis thaliana ZFN1 gene Proteins 0.000 description 1
- 241000620196 Arthrospira maxima Species 0.000 description 1
- 240000002900 Arthrospira platensis Species 0.000 description 1
- 235000016425 Arthrospira platensis Nutrition 0.000 description 1
- 108091005950 Azurite Proteins 0.000 description 1
- 241000157302 Bison bison athabascae Species 0.000 description 1
- 108010045123 Blasticidin-S deaminase Proteins 0.000 description 1
- 241000823281 Burkholderiales bacterium Species 0.000 description 1
- NLZUEZXRPGMBCV-UHFFFAOYSA-N Butylhydroxytoluene Chemical compound CC1=CC(C(C)(C)C)=C(O)C(C(C)(C)C)=C1 NLZUEZXRPGMBCV-UHFFFAOYSA-N 0.000 description 1
- 101150018129 CSF2 gene Proteins 0.000 description 1
- 101150069031 CSN2 gene Proteins 0.000 description 1
- 101100170173 Caenorhabditis elegans del-1 gene Proteins 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 1
- 102000000584 Calmodulin Human genes 0.000 description 1
- 108010041952 Calmodulin Proteins 0.000 description 1
- 241001496650 Candidatus Desulforudis Species 0.000 description 1
- 102000014914 Carrier Proteins Human genes 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 102000020313 Cell-Penetrating Peptides Human genes 0.000 description 1
- 108010051109 Cell-Penetrating Peptides Proteins 0.000 description 1
- 108091092236 Chimeric RNA Proteins 0.000 description 1
- VEXZGXHMUGYJMC-UHFFFAOYSA-M Chloride anion Chemical compound [Cl-] VEXZGXHMUGYJMC-UHFFFAOYSA-M 0.000 description 1
- 208000031639 Chromosome Deletion Diseases 0.000 description 1
- 108091005960 Citrine Proteins 0.000 description 1
- 241000193403 Clostridium Species 0.000 description 1
- 241000193155 Clostridium botulinum Species 0.000 description 1
- 241000907165 Coleofasciculus chthonoplastes Species 0.000 description 1
- 241000065716 Crocosphaera watsonii Species 0.000 description 1
- 241000159506 Cyanothece Species 0.000 description 1
- 206010011732 Cyst Diseases 0.000 description 1
- AUNGANRZJHBGPY-UHFFFAOYSA-N D-Lyxoflavin Natural products OCC(O)C(O)C(O)CN1C=2C=C(C)C(C)=CC=2N=C2C1=NC(=O)NC2=O AUNGANRZJHBGPY-UHFFFAOYSA-N 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 102000052510 DNA-Binding Proteins Human genes 0.000 description 1
- 108700020911 DNA-Binding Proteins Proteins 0.000 description 1
- 101100239628 Danio rerio myca gene Proteins 0.000 description 1
- 101000889905 Enterobacteria phage RB3 Intron-associated endonuclease 3 Proteins 0.000 description 1
- 101000889904 Enterobacteria phage T4 Defective intron-associated endonuclease 3 Proteins 0.000 description 1
- 101000889900 Enterobacteria phage T4 Intron-associated endonuclease 1 Proteins 0.000 description 1
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- 101001091269 Escherichia coli Hygromycin-B 4-O-kinase Proteins 0.000 description 1
- 241000289695 Eutheria Species 0.000 description 1
- 241001468125 Exiguobacterium Species 0.000 description 1
- 101150112093 FGF9 gene Proteins 0.000 description 1
- 108700007698 Genetic Terminator Regions Proteins 0.000 description 1
- KOSRFJWDECSPRO-WDSKDSINSA-N Glu-Glu Chemical compound OC(=O)CC[C@H](N)C(=O)N[C@@H](CCC(O)=O)C(O)=O KOSRFJWDECSPRO-WDSKDSINSA-N 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 1
- 102000004144 Green Fluorescent Proteins Human genes 0.000 description 1
- 241000700721 Hepatitis B virus Species 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- MAJYPBAJPNUFPV-BQBZGAKWSA-N His-Cys Chemical compound SC[C@@H](C(O)=O)NC(=O)[C@@H](N)CC1=CN=CN1 MAJYPBAJPNUFPV-BQBZGAKWSA-N 0.000 description 1
- 108010074870 Histone Demethylases Proteins 0.000 description 1
- 102000008157 Histone Demethylases Human genes 0.000 description 1
- 102000003893 Histone acetyltransferases Human genes 0.000 description 1
- 108090000246 Histone acetyltransferases Proteins 0.000 description 1
- 101000870664 Homo sapiens ATP-dependent RNA helicase DDX3Y Proteins 0.000 description 1
- 101001139134 Homo sapiens Krueppel-like factor 4 Proteins 0.000 description 1
- 101001088879 Homo sapiens Lysine-specific demethylase 5D Proteins 0.000 description 1
- 101001111984 Homo sapiens N-acylneuraminate-9-phosphatase Proteins 0.000 description 1
- 101000984042 Homo sapiens Protein lin-28 homolog A Proteins 0.000 description 1
- 101000711846 Homo sapiens Transcription factor SOX-9 Proteins 0.000 description 1
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 1
- GRRNUXAQVGOGFE-UHFFFAOYSA-N Hygromycin-B Natural products OC1C(NC)CC(N)C(O)C1OC1C2OC3(C(C(O)C(O)C(C(N)CO)O3)O)OC2C(O)C(CO)O1 GRRNUXAQVGOGFE-UHFFFAOYSA-N 0.000 description 1
- 108010091358 Hypoxanthine Phosphoribosyltransferase Proteins 0.000 description 1
- 102000018251 Hypoxanthine Phosphoribosyltransferase Human genes 0.000 description 1
- XQFRJNBWHJMXHO-RRKCRQDMSA-N IDUR Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(I)=C1 XQFRJNBWHJMXHO-RRKCRQDMSA-N 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 102100020677 Krueppel-like factor 4 Human genes 0.000 description 1
- 241001430080 Ktedonobacter racemifer Species 0.000 description 1
- FFEARJCKVFRZRR-UHFFFAOYSA-N L-Methionine Natural products CSCCC(N)C(O)=O FFEARJCKVFRZRR-UHFFFAOYSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 1
- 229930182844 L-isoleucine Natural products 0.000 description 1
- 239000004395 L-leucine Substances 0.000 description 1
- 235000019454 L-leucine Nutrition 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- 229930195722 L-methionine Natural products 0.000 description 1
- 241000186673 Lactobacillus delbrueckii Species 0.000 description 1
- 241000186869 Lactobacillus salivarius Species 0.000 description 1
- 241001134698 Lyngbya Species 0.000 description 1
- 102100033143 Lysine-specific demethylase 5D Human genes 0.000 description 1
- 101150039798 MYC gene Proteins 0.000 description 1
- 241000501784 Marinobacter sp. Species 0.000 description 1
- 102100025169 Max-binding protein MNT Human genes 0.000 description 1
- 241000204639 Methanohalobium Species 0.000 description 1
- 108060004795 Methyltransferase Proteins 0.000 description 1
- 241000192710 Microcystis aeruginosa Species 0.000 description 1
- 241000190905 Microscilla Species 0.000 description 1
- 241000699709 Microtus Species 0.000 description 1
- 241000108056 Monas Species 0.000 description 1
- 108010085220 Multiprotein Complexes Proteins 0.000 description 1
- 102000007474 Multiprotein Complexes Human genes 0.000 description 1
- 101100494762 Mus musculus Nedd9 gene Proteins 0.000 description 1
- 241001232540 Myopus Species 0.000 description 1
- OVBPIULPVIDEAO-UHFFFAOYSA-N N-Pteroyl-L-glutaminsaeure Natural products C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-UHFFFAOYSA-N 0.000 description 1
- 102100023906 N-acylneuraminate-9-phosphatase Human genes 0.000 description 1
- 241000167285 Natranaerobius thermophilus Species 0.000 description 1
- 101100385413 Neurospora crassa (strain ATCC 24698 / 74-OR23-1A / CBS 708.71 / DSM 1257 / FGSC 987) csm-3 gene Proteins 0.000 description 1
- DFPAKSUCGFBDDF-UHFFFAOYSA-N Nicotinamide Chemical compound NC(=O)C1=CC=CN=C1 DFPAKSUCGFBDDF-UHFFFAOYSA-N 0.000 description 1
- 241000919925 Nitrosococcus halophilus Species 0.000 description 1
- 241001515112 Nitrosococcus watsonii Species 0.000 description 1
- 241000203619 Nocardiopsis dassonvillei Species 0.000 description 1
- 241001223105 Nodularia spumigena Species 0.000 description 1
- 241000192673 Nostoc sp. Species 0.000 description 1
- 102000011931 Nucleoproteins Human genes 0.000 description 1
- 108010061100 Nucleoproteins Proteins 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 241000192520 Oscillatoria sp. Species 0.000 description 1
- 108091081548 Palindromic sequence Proteins 0.000 description 1
- 241000142651 Pelotomaculum thermopropionicum Species 0.000 description 1
- 239000004107 Penicillin G sodium Substances 0.000 description 1
- 241001135648 Petrotoga Species 0.000 description 1
- 241000286209 Phasianidae Species 0.000 description 1
- BELBBZDIHDAJOR-UHFFFAOYSA-N Phenolsulfonephthalein Chemical compound C1=CC(O)=CC=C1C1(C=2C=CC(O)=CC=2)C2=CC=CC=C2S(=O)(=O)O1 BELBBZDIHDAJOR-UHFFFAOYSA-N 0.000 description 1
- 241001599925 Polaromonas naphthalenivorans Species 0.000 description 1
- 108091036407 Polyadenylation Proteins 0.000 description 1
- 239000004698 Polyethylene Substances 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920002873 Polyethylenimine Polymers 0.000 description 1
- 101710149951 Protein Tat Proteins 0.000 description 1
- 102100026827 Protein associated with UVRAG as autophagy enhancer Human genes 0.000 description 1
- 101710102978 Protein associated with UVRAG as autophagy enhancer Proteins 0.000 description 1
- 102100025460 Protein lin-28 homolog A Human genes 0.000 description 1
- 241000590028 Pseudoalteromonas haloplanktis Species 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 102000014450 RNA Polymerase III Human genes 0.000 description 1
- 108010078067 RNA Polymerase III Proteins 0.000 description 1
- 230000004570 RNA-binding Effects 0.000 description 1
- 241000700157 Rattus norvegicus Species 0.000 description 1
- 101100247004 Rattus norvegicus Qsox1 gene Proteins 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 102000006382 Ribonucleases Human genes 0.000 description 1
- 108010083644 Ribonucleases Proteins 0.000 description 1
- 108020004422 Riboswitch Proteins 0.000 description 1
- 101150086694 SLC22A3 gene Proteins 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 238000012300 Sequence Analysis Methods 0.000 description 1
- 241000191967 Staphylococcus aureus Species 0.000 description 1
- 241000194022 Streptococcus sp. Species 0.000 description 1
- 241000187747 Streptomyces Species 0.000 description 1
- 101001091268 Streptomyces hygroscopicus Hygromycin-B 7''-O-kinase Proteins 0.000 description 1
- 241001518258 Streptomyces pristinaespiralis Species 0.000 description 1
- 241000272534 Struthio camelus Species 0.000 description 1
- 241000192560 Synechococcus sp. Species 0.000 description 1
- 101710137500 T7 RNA polymerase Proteins 0.000 description 1
- 241000288716 Talpa Species 0.000 description 1
- 101710192266 Tegument protein VP22 Proteins 0.000 description 1
- 108091046869 Telomeric non-coding RNA Proteins 0.000 description 1
- 241000206213 Thermosipho africanus Species 0.000 description 1
- JZRWCGZRTZMZEH-UHFFFAOYSA-N Thiamine Natural products CC1=C(CCO)SC=[N+]1CC1=CN=C(C)N=C1N JZRWCGZRTZMZEH-UHFFFAOYSA-N 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 102100034204 Transcription factor SOX-9 Human genes 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 241000078013 Trichormus variabilis Species 0.000 description 1
- 108010064978 Type II Site-Specific Deoxyribonucleases Proteins 0.000 description 1
- 108010067022 Type III Site-Specific Deoxyribonucleases Proteins 0.000 description 1
- 102000018390 Ubiquitin-Specific Proteases Human genes 0.000 description 1
- 108010066496 Ubiquitin-Specific Proteases Proteins 0.000 description 1
- 208000037206 XYY Karyotype Diseases 0.000 description 1
- 108010027570 Xanthine phosphoribosyltransferase Proteins 0.000 description 1
- 101100459258 Xenopus laevis myc-a gene Proteins 0.000 description 1
- 241001673106 [Bacillus] selenitireducens Species 0.000 description 1
- 210000001015 abdomen Anatomy 0.000 description 1
- 230000001594 aberrant effect Effects 0.000 description 1
- 230000009471 action Effects 0.000 description 1
- 125000003295 alanine group Chemical group N[C@@H](C)C(=O)* 0.000 description 1
- KOSRFJWDECSPRO-UHFFFAOYSA-N alpha-L-glutamyl-L-glutamic acid Natural products OC(=O)CCC(N)C(=O)NC(CCC(O)=O)C(O)=O KOSRFJWDECSPRO-UHFFFAOYSA-N 0.000 description 1
- 208000036878 aneuploidy Diseases 0.000 description 1
- 231100001075 aneuploidy Toxicity 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 229940011019 arthrospira platensis Drugs 0.000 description 1
- 229940009098 aspartate Drugs 0.000 description 1
- 238000002869 basic local alignment search tool Methods 0.000 description 1
- FCPVYOBCFFNJFS-LQDWTQKMSA-M benzylpenicillin sodium Chemical group [Na+].N([C@H]1[C@H]2SC([C@@H](N2C1=O)C([O-])=O)(C)C)C(=O)CC1=CC=CC=C1 FCPVYOBCFFNJFS-LQDWTQKMSA-M 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 235000008429 bread Nutrition 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- FAPWYRCQGJNNSJ-UBKPKTQASA-L calcium D-pantothenic acid Chemical compound [Ca+2].OCC(C)(C)[C@@H](O)C(=O)NCCC([O-])=O.OCC(C)(C)[C@@H](O)C(=O)NCCC([O-])=O FAPWYRCQGJNNSJ-UBKPKTQASA-L 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 244000309466 calf Species 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 229920006317 cationic polymer Polymers 0.000 description 1
- 210000004671 cell-free system Anatomy 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000013611 chromosomal DNA Substances 0.000 description 1
- ZDLBNXXKDMLZMF-UHFFFAOYSA-N cinitapride Chemical compound CCOC1=CC(N)=C([N+]([O-])=O)C=C1C(=O)NC1CCN(CC2CC=CCC2)CC1 ZDLBNXXKDMLZMF-UHFFFAOYSA-N 0.000 description 1
- 239000011035 citrine Substances 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 239000002131 composite material Substances 0.000 description 1
- 238000012790 confirmation Methods 0.000 description 1
- 101150055601 cops2 gene Proteins 0.000 description 1
- 230000001186 cumulative effect Effects 0.000 description 1
- 238000005520 cutting process Methods 0.000 description 1
- 108010082025 cyan fluorescent protein Proteins 0.000 description 1
- 208000031513 cyst Diseases 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 235000013365 dairy product Nutrition 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 230000001079 digestive effect Effects 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 230000002124 endocrine Effects 0.000 description 1
- 108010050663 endodeoxyribonuclease CreI Proteins 0.000 description 1
- 230000002255 enzymatic effect Effects 0.000 description 1
- 230000004049 epigenetic modification Effects 0.000 description 1
- 238000010195 expression analysis Methods 0.000 description 1
- 230000006408 female gonad development Effects 0.000 description 1
- 239000007850 fluorescent dye Substances 0.000 description 1
- 229960000304 folic acid Drugs 0.000 description 1
- 235000019152 folic acid Nutrition 0.000 description 1
- 239000011724 folic acid Substances 0.000 description 1
- 230000004077 genetic alteration Effects 0.000 description 1
- 231100000118 genetic alteration Toxicity 0.000 description 1
- 102000054766 genetic haplotypes Human genes 0.000 description 1
- 238000010362 genome editing Methods 0.000 description 1
- 108010055341 glutamyl-glutamic acid Proteins 0.000 description 1
- 239000005090 green fluorescent protein Substances 0.000 description 1
- 230000007062 hydrolysis Effects 0.000 description 1
- 238000006460 hydrolysis reaction Methods 0.000 description 1
- GRRNUXAQVGOGFE-NZSRVPFOSA-N hygromycin B Chemical compound O[C@@H]1[C@@H](NC)C[C@@H](N)[C@H](O)[C@H]1O[C@H]1[C@H]2O[C@@]3([C@@H]([C@@H](O)[C@@H](O)[C@@H](C(N)CO)O3)O)O[C@H]2[C@@H](O)[C@@H](CO)O1 GRRNUXAQVGOGFE-NZSRVPFOSA-N 0.000 description 1
- 229940097277 hygromycin b Drugs 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000007901 in situ hybridization Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 208000000509 infertility Diseases 0.000 description 1
- 231100000535 infertility Toxicity 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- CDAISMWEOUEBRE-GPIVLXJGSA-N inositol Chemical compound O[C@H]1[C@H](O)[C@@H](O)[C@H](O)[C@H](O)[C@@H]1O CDAISMWEOUEBRE-GPIVLXJGSA-N 0.000 description 1
- 229960000310 isoleucine Drugs 0.000 description 1
- 238000005304 joining Methods 0.000 description 1
- 229960003136 leucine Drugs 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 210000004324 lymphatic system Anatomy 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 239000000463 material Substances 0.000 description 1
- 230000013011 mating Effects 0.000 description 1
- 238000005259 measurement Methods 0.000 description 1
- 239000012577 media supplement Substances 0.000 description 1
- 229910021645 metal ion Inorganic materials 0.000 description 1
- 229960004452 methionine Drugs 0.000 description 1
- 108091070501 miRNA Proteins 0.000 description 1
- 239000002679 microRNA Substances 0.000 description 1
- 230000025608 mitochondrion localization Effects 0.000 description 1
- 108091005601 modified peptides Proteins 0.000 description 1
- 230000003387 muscular Effects 0.000 description 1
- 239000002105 nanoparticle Substances 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 229960003966 nicotinamide Drugs 0.000 description 1
- 235000005152 nicotinamide Nutrition 0.000 description 1
- 239000011570 nicotinamide Substances 0.000 description 1
- 208000012978 nondisjunction Diseases 0.000 description 1
- 125000000371 nucleobase group Chemical group 0.000 description 1
- 229940099990 ogen Drugs 0.000 description 1
- 230000003287 optical effect Effects 0.000 description 1
- 210000003463 organelle Anatomy 0.000 description 1
- 230000003204 osmotic effect Effects 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000003415 peat Substances 0.000 description 1
- 230000000149 penetrating effect Effects 0.000 description 1
- 235000019369 penicillin G sodium Nutrition 0.000 description 1
- 229940056360 penicillin g Drugs 0.000 description 1
- 229960003531 phenolsulfonphthalein Drugs 0.000 description 1
- 229960005190 phenylalanine Drugs 0.000 description 1
- 230000004962 physiological condition Effects 0.000 description 1
- 239000013600 plasmid vector Substances 0.000 description 1
- 108010011110 polyarginine Proteins 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 229920001223 polyethylene glycol Polymers 0.000 description 1
- 102000054765 polymorphisms of proteins Human genes 0.000 description 1
- 239000011148 porous material Substances 0.000 description 1
- 230000029279 positive regulation of transcription, DNA-dependent Effects 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- 235000011164 potassium chloride Nutrition 0.000 description 1
- 244000144977 poultry Species 0.000 description 1
- 235000013594 poultry meat Nutrition 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 238000004321 preservation Methods 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000012545 processing Methods 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 230000007026 protein scission Effects 0.000 description 1
- 230000004850 protein–protein interaction Effects 0.000 description 1
- 235000008160 pyridoxine Nutrition 0.000 description 1
- 239000011677 pyridoxine Substances 0.000 description 1
- 150000004728 pyruvic acid derivatives Chemical class 0.000 description 1
- 230000008707 rearrangement Effects 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 102000037983 regulatory factors Human genes 0.000 description 1
- 108091008025 regulatory factors Proteins 0.000 description 1
- 108091035233 repetitive DNA sequence Proteins 0.000 description 1
- 102000053632 repetitive DNA sequence Human genes 0.000 description 1
- 230000003252 repetitive effect Effects 0.000 description 1
- 210000002345 respiratory system Anatomy 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 229960002477 riboflavin Drugs 0.000 description 1
- 235000019192 riboflavin Nutrition 0.000 description 1
- 239000002151 riboflavin Substances 0.000 description 1
- CDAISMWEOUEBRE-UHFFFAOYSA-N scyllo-inosotol Natural products OC1C(O)C(O)C(O)C(O)C1O CDAISMWEOUEBRE-UHFFFAOYSA-N 0.000 description 1
- 238000005204 segregation Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 229960001153 serine Drugs 0.000 description 1
- 230000003584 silencer Effects 0.000 description 1
- 239000007858 starting material Substances 0.000 description 1
- 229960002385 streptomycin sulfate Drugs 0.000 description 1
- 230000004960 subcellular localization Effects 0.000 description 1
- 235000019157 thiamine Nutrition 0.000 description 1
- KYMBYSLLVAOCFI-UHFFFAOYSA-N thiamine Chemical compound CC1=C(CCO)SCN1CC1=CN=C(C)N=C1N KYMBYSLLVAOCFI-UHFFFAOYSA-N 0.000 description 1
- 229960003495 thiamine Drugs 0.000 description 1
- 239000011721 thiamine Substances 0.000 description 1
- 229960002898 threonine Drugs 0.000 description 1
- 230000005026 transcription initiation Effects 0.000 description 1
- 108091006106 transcriptional activators Proteins 0.000 description 1
- 238000003146 transient transfection Methods 0.000 description 1
- 230000007704 transition Effects 0.000 description 1
- GWBUNZLLLLDXMD-UHFFFAOYSA-H tricopper;dicarbonate;dihydroxide Chemical compound [OH-].[OH-].[Cu+2].[Cu+2].[Cu+2].[O-]C([O-])=O.[O-]C([O-])=O GWBUNZLLLLDXMD-UHFFFAOYSA-H 0.000 description 1
- 229960004441 tyrosine Drugs 0.000 description 1
- 230000002485 urinary effect Effects 0.000 description 1
- 230000003612 virological effect Effects 0.000 description 1
- 229940011671 vitamin b6 Drugs 0.000 description 1
- 229940075420 xanthine Drugs 0.000 description 1
- NWONKYPBYAMBJT-UHFFFAOYSA-L zinc sulfate Chemical group [Zn+2].[O-]S([O-])(=O)=O NWONKYPBYAMBJT-UHFFFAOYSA-L 0.000 description 1
Landscapes
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Methods and compositions are provided for generating targeted genetic modifications on the Y chromosome or a challenging target locus. Compositions include an in vitro culture comprising an XY pluripotent and/or totipotent animal cell (i.e., XY ES cells or XY iPS cells) having a modification that decreases the level and/or activity of an Sry protein; and, culturing these cells in a medium that promotes development of XY F0 fertile females. Such compositions find use in various methods for making a fertile female XY non-human mammal in an F0 generation.
Description
METHODS AND COMPOSITIONS FOR TARGETED
GENETIC MODIFICATIONS AND METHODS OF USE
This application is a onal application from New Zealand Patent Application Number
728561, the entire disclosures of which are incorporated herein by reference.
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No.
,582, filed June 26, 2014, and of U.S. Provisional Application No. 62/017,627, filed
June 26, 2014, each of which is hereby incorporated herein in its entirety by reference.
REFERENCE TO A CE LISTING SUBMITTED
AS A TEXT FILE VIA EFS WEB
The official copy of the sequence listing is submitted electronically via EFS-Web
as an ASCII formatted ce listing with a file named 463545SEQLIST.TXT, created on
June 25, 2015, and having a size of 14 kilobytes, and is filed concurrently with the
ication. The sequence listing contained in this ASCII formatted nt is part of the
ication and is herein incorporated by reference in its entirety.
FIELD
The invention relates to the methods and compositions for maintaining or
ing pluripotent and/or totipotent cells and methods and compositions for generating cell
populations and transgenic animals.
BACKGROUND
Perhaps due to unique structural features of the Y chromosome, conventional gene
targeting strategies in mouse embryonic stem cells to generate mutations on the Y-linked
genes have had limited success. Therefore, often the understanding of the ons of
murine Y-linked genes is limited to ts gained from studies of mice that carry
spontaneous deletions, random gene traps insertions or autosomal transgenes. Methods are
needed to improve the ability to target a genomic locus on the Y chromosome.
The Sry protein (sex-determining region Y) is the key regulator of male sex
ination in placental mammals. The Sry gene, also known as the Testis Determining
Factor (TDF), resides on the Y chromosome. Sry is thought to be a transcription factor that
binds DNA through its High Mobility Group (HMG) domain. Expression of the mouse Sry
gene is restricted to the genital ridge in a narrow time window around day 11 of embryonic
development; both Sry mRNA and n are ed. Sufficient Sry must be made within
this time window to convert the ntial genital ridge toward the male testis forming
program while inhibiting the female program of ovary development. In adult testes a circular
Sry transcript is detected but not the Sry protein. Mutations in the Sry gene that cause the
production of an inactive Sry protein or that alter the timing and th of gene expression
can cause male to female sex reversal, resulting in animals that have an X and a Y
chromosome but are anatomically female. So-called XY females are often sterile or have a
low fertility. Being able to control sex determination by regulation of the Sry would have
great value in the production of cally modified animals.
SUMMARY
A method for making an XY embryonic stem (ES) cell line capable of producing a
fertile XY female non-human mammal in an F0 generation is provided. The method
comprises: (a) modifying a non-human mammalian XY embryonic stem (ES) cell to have a
modification that decreases the level and/or activity of an Sry protein; and, (b) culturing the
ed ES cell line under conditions that allow for making an ES cell line e of
producing a fertile XY female non-human mammal in an F0 generation.
A method for making a fertile XY female non-human mammal in an F0
generation is also provided. The method comprises: (a) ucing the man
mammalian XY ES cell made by the above method having a modification that decreases the
level and/or activity of an Sry protein into a host embryo; (b) gestating the host embryo; and,
(c) ing an F0 XY female non-human mammal, wherein upon attaining sexual maturity
the F0 XY female non-human mammal is fertile. In one embodiment, the female XY F0 non-
human mammal is fertile when crossed to a wild type mouse. In specific embodiments, the
wild type mouse is C57BL/6.
In one ment, the non-human mammalian XY ES cell is from a rodent. In a
specific embodiment, the rodent is a mouse. In one embodiment, the mouse XY ES cell is
derived from a 129 strain. In one embodiment, the mouse XY ES cell is a VGFl mouse ES
cell. In one embodiment, the mouse XY ES cell comprises a Y chromosome derived from the
129 strain. In one embodiment, the mouse XY ES cell is from a C57BL/6 strain. In another
embodiment the rodent is a rat or a hamster.
In some embodiments, the decreased level and/or activity of the Sry protein
results from a genetic modification in the Sry gene. In some such methods, the genetic
WO 00805
modification in the Sry gene comprises an insertion of one or more nucleotides, a deletion of
one or more nucleotides, a tution of one or more nucleotides, a knockout, a knockin, a
replacement of an endogenous c acid ce with a homologous, heterologous, or
orthologous nucleic acid sequence, or a combination thereof.
In the methods provided herein, the targeted genetic modification can comprises
an insertion, a deletion, a knockout, a n, a point mutation, or a combination thereof. In
another ment, the targeted genetic modification is on an autosome.
In some embodiments, the modification of the Sry gene comprises an insertion of
a selectable marker and/or a reporter gene operably linked to a promoter active in the non-
human mammalian ES cell. In some embodiments, the the modification of the Sry gene
comprises an insertion of a reporter gene operably linked to the nous Sry promoter. In
a specific embodiment, the reporter gene encodes the reporter protein LacZ.
In one embodiment, the culturing step comprises ing the non-human
mammalian XY ES cell in a medium comprising a base medium and supplements suitable for
maintaining the non-human mammalian ES cell in culture, n the medium is a low-
osmolality medium. In one embodiment, the low-osmolality medium exhibits an osmolality
from about 200 mOsm/kg to less than about 329 mOsm/kg. In other embodiments the low-
osmolality medium exhibits one or more of the following characteristic: a conductivity of
about 11 mS/cm to about 13 mS/cm; a salt of an alkaline metal and a halide in a
concentration of about 50 mM to about 110 mM; a carbonic acid salt concentration of about
l7mM to about 30 mM; a total alkaline metal halide salt and carbonic acid salt concentration
of about 85mM to about 130 mM; and/or a combination of any two or more thereof.
In some ments, upon introduction of the non-human mammalian XY ES
cells into a host embryo and following gestation of the host embryo, at least 80% at least
85%, at least 90%, or at least 95% of the F0 non-human mammals are XY females which
upon attaining sexual maturity the F0 XY female non-human mammal is fertile.
In one embodiment, the man mammalian XY ES cell comprises a target
genomic locus on the Y chromosome comprising a recognition site for a se agent, and
wherein the nuclease agent induces a nick or double-strand break at the recognition site. Such
a method can r comprise exposing the ES cell to the nuclease agent in the ce of a
targeting vector comprising an insert polynucleotide, wherein following exposure to the
nuclease agent and the targeting vector, the ES cell is modified to contain the insert
polynucleotide. In one embodiment, the nuclease agent is an mRNA encoding a nuclease. In
specific embodiments, the se agent is (a) a zinc finger nuclease (ZEN); (b) is a
2015/038001
Transcription Activator-Like Effector se (TALEN); or (C) a meganuclease. In other
embodiments, the nuclease agent comprises a Clustered Regularly Interspaced Short
Palindromic Repeats (CRISPR)-associated (Cas) protein and a guide RNA (gRNA). In such
methods, the guide RNA (gRNA) ses (a) a Clustered Regularly Interspaced Short
Palindromic Repeats (CRISPR) RNA (chNA) that targets the first recognition site; and (b) a
trans-activating CRISPR RNA (trachNA). In some cases, the recognition site is immediately
flanked by a Protospacer Adjacent Motif (PAM) sequence. In one embodiment, the Cas
protein is Cas9.
Also provided is an in vitro culture comprising the non-human mammalian XY ES
cell line according to any of the methods provided herein.
An in vitro culture is provided and comprises (a) a non-human mammalian XY
embryonic stem (ES) cell having a modification that decreases the level and/or activity of an
Sry n; and, (b) a medium comprising a base medium and supplements suitable for
maintaining the non-human mammalian ES cell in culture. In one embodiment, the base
medium ts an osmolality from about 200 mOsm/kg to less than about 329 mOsm/kg. In
other embodiments, the base medium ts one or more of the following characteristic: a
conductivity of about 11 mS/cm to about 13 mS/cm; a salt of an alkaline metal and a halide in
a concentration of about 50 mM to about 110 mM; a carbonic acid salt concentration of about
l7mM to about 30 mM; a total alkaline metal halide salt and carbonic acid salt concentration
of about 85mM to about 130 mM; and/or a ation of any two or more thereof. In one
embodiment, the non-human mammalian XY ES cell is from a rodent. In one embodiment,
the rodent is a mouse or a rat. In one embodiment, the mouse XY ES cell is a VGFl mouse
ES cell. In one embodiment, the rodent is a rat or a hamster. In one embodiment, the
decreased level and/or activity of the Sry protein is from a genetic cation in the Sry
gene. In one embodiment, the c modification in the Sry gene comprises an insertion of
one or more tides, a on of one or more nucleotides, a substitution of one or more
nucleotides, a knockout, a knockin, a replacement of an endogenous nucleic acid sequence
with a heterologous nucleic acid sequence or a ation thereof. In one embodiment, the
non-human mammalian ES cell comprises one, two, three or more targeted genetic
modifications. In one embodiment, the targeted genetic modification comprises an insertion,
a deletion, a knockout, a knockin, a point on, or a combination thereof. In one
embodiment, the targeted genetic modification comprises at least one ion of a
heterologous polynucleotide into the genome of the XY ES cell. In one ment, the
targeted genetic modification is on an autosome. In one embodiment, the base medium
exhibits 50 i 5 mM NaCl, 26 i 5 mM carbonate, and 218 i 22 mOsm/kg. In one
embodiment, the base medium ts about 3 mg/mL NaCl, 2.2 mg/mL sodium
bicarbonate, and 218 g. In one embodiment, the base medium exhibits 87 i 5 mM
NaCl, 18 i 5 mM carbonate, and 261 i 26 mOsm/kg. In one embodiment, the base medium
exhibits about 5.1 mg/mL NaCl, 1.5 mg/mL sodium bicarbonate, and 261 mOsm/kg. In one
embodiment, the base medium ts 110 i 5 mM NaCl, 18 i 5 mM carbonate, and 294 i
29 mOsm/kg. In one embodiment, the base medium exhibits about 6.4 mg/mL NaCl, 1.5
mg/mL sodium bicarbonate, and 294 mOsm/kg. In one embodiment, the base medium
exhibits 87 i 5 mM NaCl, 26 i 5 mM carbonate, and 270 i 27 mOsm/kg. In one
embodiment, the base medium ts about 5.1 mg/mL NaCl, 2.2 mg/mL sodium
bicarbonate, and 270 mOsm/kg. In one embodiment, the base medium exhibits 87 i 5 mM
NaCl, 26 i 5 mM carbonate, 86 i 5 mM e, and 322 i 32 mOsm/kg. In one
embodiment, the base medium exhibits about 5.1 mg/mL NaCl, 2.2 mg/mL sodium
bicarbonate, 15.5 mg/mL glucose, and 322 mOsm/kg. In one ment, upon introduction
of the non-human mammalian XY ES cells into a host embryo and following gestation of the
host embryo, at least 80% of the F0 non-human mammals are XY females which upon
ing sexual maturity the F0 XY female non-human mammal is fertile.
Further provided is a method for making a fertile female XY non-human mammal
in an F0 generation, comprising: (a) culturing a donor non-human mammalian XY
embryonic stem (ES) cell having a modification that decreases the level and/or activity of an
Sry protein in a medium comprising a base medium and supplements suitable for maintaining
the non-human mammalian ES cell in culture, (b) introducing the donor XY non-human
ian ES cell into a host ; (c) gestating the host embryo; and, (d) obtaining an
F0 XY female non-human mammal, wherein upon attaining sexual maturity the F0 XY
female non-human mammal is fertile. In one embodiment, the medium exhibits an osmolality
from about 200 mOsm/kg to less than about 329 mOsm/kg. In other embodiments, the
medium exhibits a characteristic comprising one or more of the following: a tivity of
about 11 mS/cm to about 13 mS/cm; a salt of an alkaline metal and a halide in a
tration of about 50mM to about 110 mM; a carbonic acid salt concentration of about
17 mM to about 30 mM; a total alkaline metal halide salt and carbonic acid salt concentration
of about 85 mM to about 130 mM; and/or a ation of any two or more thereof; In one
embodiment, the non-human mammalian XY ES cell is from a rodent. In one embodiment,
the rodent is a mouse or a rat. In one embodiment, the mouse XY ES cell is a VGFl mouse
ES cell. In one embodiment, the rodent is a rat or a hamster. In one embodiment, the
decreased level and/or activity of the Sry protein is from a genetic modification in the Sry
gene. In one embodiment, the genetic modification in the Sry gene comprises an ion of
one or more nucleotides, a deletion of one or more nucleotides, a tution of one or more
nucleotides, a knockout, a knockin, a replacement of an endogenous nucleic acid sequence
with a heterologous c acid sequence or a combination f. In one embodiment, the
non-human mammalian ES cell comprises one, two, three or more targeted genetic
modifications. In one embodiment, the targeted c modification comprises an insertion,
a deletion, a knockout, a knockin, a point on, or a combination thereof. In one
embodiment, the targeted genetic modification comprises at least one insertion of a
heterologous polynucleotide into a genome of the XY ES cell. In one embodiment, the
targeted genetic modification is on an autosome. In one embodiment, the base medium
exhibits 50 i 5 mM NaCl, 26 i 5 mM carbonate, and 218 i 22 g. In one
embodiment, the base medium exhibits about 3 mg/mL NaCl, 2.2 mg/mL sodium
bicarbonate, and 218 mOsm/kg. In one embodiment, the base medium exhibits 87 i 5 mM
NaCl, 18 i 5 mM carbonate, and 261 i 26 mOsm/kg. In one embodiment, the base medium
exhibits about 5.1 mg/mL NaCl, 1.5 mg/mL sodium bicarbonate, and 261 mOsm/kg. In one
embodiment, the base medium exhibits 110 i 5 mM NaCl, 18 i 5 mM carbonate, and 294 i
29 mOsm/kg. In one embodiment, the base medium ts about 6.4 mg/mL NaCl, 1.5
mg/mL sodium bicarbonate, and 294 mOsm/kg. In one embodiment, the base medium
exhibits 87 i 5 mM NaCl, 26 i 5 mM carbonate, and 270 i 27 mOsm/kg. In one
embodiment, the base medium exhibits about 5.1 mg/mL NaCl, 2.2 mg/mL sodium
bicarbonate, and 270 mOsm/kg. In one ment, wherein the base medium exhibits 87 i
mM NaCl, 26 i 5 mM carbonate, 86 i 5 mM glucose, and 322 i 32 mOsm/kg. In one
embodiment, n the base medium exhibits about 5.1 mg/mL NaCl, 2.2 mg/mL sodium
bicarbonate, 15.5 mg/mL glucose, and 322 mOsm/kg.
Further provided are methods of producing a enic non-human mammal
homozygous for a targeted genetic mutation in the F1 generation comprising: (a) crossing an
F0 XY fertile female having a decreased level and/or activity of the Sry protein with a cohort
clonal sibling, derived from the same ES cell clone, F0 XY male non-human mammal,
wherein the F0 XY fertile female non-human mammal and the F0 XY male non-human
mammal each is heterozygous for the genetic mutation; and,(b) obtaining an F1 progeny
mouse that is homozygous for the c modification.
A method for modifying a target genomic locus on the Y chromosome in a cell is
also ed and comprises (a) providing a cell comprising a target genomic locus on the Y
some comprising a recognition site for a nuclease agent, (b) introducing into the cell
(i) the nuclease agent, wherein the nuclease agent induces a nick or double-strand break at
the first recognition site; and, (ii) a first targeting vector comprising a first insert
polynucleotide flanked by a first and a second homology arm corresponding to a first and a
second target site located in sufficient proximity to the first recognition site; and, (c)
identifying at least one cell comprising in its genome the first insert polynucleotide ated
at the target genomic locus. In one ment, a sum total of the first homology arm and
the second homology arm is at least 4kb but less than 150kb. In one ment, the length
of the first homology arm and/or the second homology arm is at least 400 bp but less than
1000 bp. In another embodiment, the length of the first homology arm and/or the second
homology arm is from about 700 bp to about 800 bp.
Further provided is a method for modifying a target genomic locus on the Y
chromosome in a cell is provided and ses: (a) providing a cell comprising a target
genomic locus on the Y chromosome comprising a recognition site for a nuclease agent, (b)
introducing into the cell a first targeting vector comprising a first insert polynucleotide
flanked by a first and a second gy arm corresponding to a first and a second target
site; and, (c) identifying at least one cell comprising in its genome the first insert
polynucleotide integrated at the target genomic locus. In one embodiment, the length of the
first gy arm and/or the second homology arm is at least 400 bp but less than 1000 bp.
In another embodiment, the length of the first homology arm and/or the second homology
arm is from about 700 bp to about 800 bp. In one embodiment, the cell is a mammalian cell.
In one embodiment, the ian cell is a non-human cell. In one embodiment, the
mammalian cell is from a rodent. In one embodiment, the rodent is a rat, a mouse or a
hamster. In one embodiment, the cell is a pluripotent cell. In one embodiment, the
mammalian cell is an induced pluripotent stem (iPS) cell. In one embodiment, the pluripotent
cell is a non-human embryonic stem (ES) cell. In one embodiment, the pluripotent cell is a
rodent embryonic stem (ES) cell, a mouse nic stem (ES) cell or a rat embryonic stem
(ES) cell. In one embodiment, the nuclease agent is an mRNA encoding a nuclease. In one
embodiment, the nuclease agent is a zinc finger se (ZFN). In one embodiment, the
nuclease agent is a Transcription Activator-Like Effector se (TALEN). In one
embodiment, the se agent is a meganuclease. In some embodiments, the nuclease
agent comprises a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-
associated (Cas) protein and a guide RNA (gRNA). In such a method the guide RNA (gRNA)
can comprise (a) a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)
WO 00805
RNA (chNA) that targets the first recognition site; and (b) a trans-activating CRISPR RNA
(trachNA). In one embodiment, the first or the second recognition sites are immediately
flanked by a Protospacer Adjacent Motif (PAM) sequence. In some embodiments, the Cas
protein is Cas9.
In some embodiments, the cation comprises a deletion of an nous
nucleic acid sequence. In some embodiments, the deletion ranges from about 5 kb to about 10
kb, from about 10 kb to about 20 kb, from about 20 kb to about 40 kb, from about 40 kb to
about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to about 100 kb, from about
100 kb to about 150 kb, or from about 150 kb to about 200 kb, from about 200 kb to about
300 kb, from about 300 kb to about 400 kb, from about 400 kb to about 500 kb, from about
500 kb to about 1 Mb, from about 1 Mb to about 1.5 Mb, from about 1.5 Mb to about 2 Mb,
from about 2 Mb to about 2.5 Mb, or from about 2.5 Mb to about 3 Mb. In a specific
embodiment, the deletion is at least 500 kb. In one embodiment, the cell is a mammalian cell.
In one embodiment, the mammalian cell is a non-human cell. In one embodiment, the
ian cell is from a rodent. In one embodiment, the rodent is a rat, a mouse or a
hamster. In one embodiment, the cell is a pluripotent cell. In one embodiment, the
mammalian cell is an induced pluripotent stem (iPS) cell. In one embodiment, the otent
cell is a non-human embryonic stem (ES) cell. In one ment, the pluripotent cell is a
rodent embryonic stem (ES) cell, a mouse embryonic stem (ES) cell or a rat embryonic stem
(ES) cell. In some embodiments, the nuclease agent comprises a Clustered Regularly
Interspaced Short Palindromic s (CRISPR)-associated (Cas) protein and a guide RNA
(gRNA). In such a method the guide RNA (gRNA) can comprise (a) a Clustered Regularly
Interspaced Short Palindromic Repeats (CRISPR) RNA (chNA) that s the first
recognition site; and (b) a trans-activating CRISPR RNA (trachNA). In one embodiment,
the first or the second recognition sites are immediately flanked by a Protospacer Adjacent
Motif (PAM) ce. In some embodiments, the Cas protein is Cas9. In one embodiment,
the nuclease agent is a zinc finger se (ZFN). In one embodiment, the nuclease agent is
a Transcription Activator-Like Effector Nuclease (TALEN). In one embodiment, the nuclease
agent is a meganuclease.
Methods for modifying the Y chromosome comprising exposing the Y
chromosome to a Cas protein and a CRISPR RNA in the presence of a large targeting vector
(LTVEC) comprising a nucleic acid sequence of at least 10 kb and comprises following
exposure to the Cas protein, the CRISPR RNA, and the LTVEC, the Y chromosome is
modified to contain at least 10 kb nucleic acid sequence. The LTVEC can comprise a nucleic
2015/038001
acid sequence of at least 20 kb, at least 30 kb, at least 40 kb, at least 50 kb, at least 60 kb, at
least 70 kb, at least 80 kb, or at least 90 kb. In other embodiments, the LTVEC comprises a
nucleic acid sequence of at least 100 kb, at least 150 kb, or at least 200 kb.
Further ed is a method for modifying a target genomic locus on the Y
chromosome, comprising: (a) ing a mammalian cell comprising the target genomic
locus on the Y chromosome, wherein the target genomic locus comprises a guide RNA
(gRNA) target sequence; (b) introducing into the ian cell: (i) a large targeting vector
(LTVEC) comprising a first nucleic acid flanked with targeting arms homologous to the
target genomic locus, wherein the LTVEC is at least 10 kb; (ii) a first expression construct
comprising a first promoter operably linked to a second nucleic acid ng a Cas n,
and (iii) a second expression construct comprising a second promoter operably linked to a
third nucleic acid encoding a guide RNA (gRNA) sing a nucleotide sequence that
hybridizes to the gRNA target sequence and a trans-activating CRISPR RNA (trachNA),
wherein the first and the second promoters are active in the ian cell; and (c)
identifying a modified mammalian cell comprising a targeted genetic cation at the
target genomic locus on the Y chromosome. In other embodiments, the LTVEC is at least 15
kb, at least 20 kb, at least 30kb, at least 40 kb, at least 50 kb, at least 60 kb, at least 70 kb, at
least 80 kb, or at least 90 kb. In other embodiments, the LTVEC is at least 100 kb, at least
150 kb, or at least 200 kb. In one embodiment, the mammalian cell is a non-human
mammalian cell. In one embodiment, the mammalian cell is a fibroblast cell. In one
embodiment, the mammalian cell is from a rodent. In one embodiment, the rodent is a rat, a
mouse, or a hamster. In one embodiment, the mammalian cell is a pluripotent cell. In one
embodiment, the pluripotent cell is an induced pluripotent stem (iPS) cell. In one
embodiment, the otent cell is a mouse embryonic stem (ES) cell or a rat embryonic
stem (ES) cell. In one embodiment, the pluripotent cell is a developmentally restricted
human progenitor cell. In one embodiment, the Cas protein is a Cas9 protein. In one
embodiment, the gRNA target sequence is ately flanked by a Protospacer Adjacent
Motif (PAM) sequence. In one embodiment, the sum total of 5’ and 3’ homology arms of the
LTVEC is from about 10 kb to about 150 kb. In one embodiment, the sum total of the 5’ and
the 3’ homology arms of the LTVEC is from about 10 kb to about 20 kb, from about 20 kb to
about 40 kb, from about 40 kb to about 60 kb, from about 60 kb to about 80 kb, from about
80 kb to about 100 kb, from about 100 kb to about 120 kb, or from about 120 kb to 150 kb.
In one embodiment, the targeted genetic modification comprises: (a) a replacement of an
endogenous nucleic acid sequence with a homologous or an orthologous nucleic acid
sequence; (b) a deletion of an endogenous nucleic acid sequence; (c) a deletion of an
endogenous nucleic acid sequence, wherein the deletion ranges from about 5 kb to about 10
kb, from about 10 kb to about 20 kb, from about 20 kb to about 40 kb, from about 40 kb to
about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to about 100 kb, from about
100 kb to about 150 kb, or from about 150 kb to about 200 kb, from about 200 kb to about
300 kb, from about 300 kb to about 400 kb, from about 400 kb to about 500 kb, from about
500 kb to about 1 Mb, from about 1 Mb to about 1.5 Mb, from about 1.5 Mb to about 2 Mb,
from about 2 Mb to about 2.5 Mb, or from about 2.5 Mb to about 3 Mb; (d) insertion of an
exogenous nucleic acid sequence; (e) insertion of an ous c acid ce
ranging from about 5kb to about 10kb, from about 10 kb to about 20 kb, from about 20 kb to
about 40 kb, from about 40 kb to about 60 kb, from about 60 kb to about 80 kb, from about
80 kb to about 100 kb, from about 100 kb to about 150 kb, from about 150 kb to about 200
kb, from about 200 kb to about 250 kb, from about 250 kb to about 300 kb, from about 300
kb to about 350 kb, or from about 350 kb to about 400 kb; (f) insertion of an exogenous
nucleic acid sequence comprising a homologous or an ogous nucleic acid ce; (g)
insertion of a chimeric nucleic acid sequence comprising a human and a non-human nucleic
acid sequence; (h) ion of a conditional allele flanked with site-specific recombinase
target sequences; (i) insertion of a selectable marker or a reporter gene operably linked to a
third promoter active in the mammalian cell; or (1') a combination thereof. In one
embodiment, the target genomic locus comprises (i) a 5’ target sequence that is homologous
to a 5’ gy arm; and (ii) a 3’ target sequence that is homologous to a 3’ homology arm.
In one embodiment, the 5’ target sequence and the 3’ target sequence is separated by at least
kb but less than 3 Mb. In one embodiment, the 5’ target sequence and the 3’ target
sequence is separated by at least 5 kb but less than 10 kb, at least 10 kb but less than 20 kb, at
least 20 kb but less than 40 kb, at least 40 kb but less than 60 kb, at least 60 kb but less than
80 kb, at least about 80 kb but less than 100 kb, at least 100 kb but less than 150 kb, or at
least 150 kb but less than 200 kb, at least about 200 kb but less than about 300 kb, at least
about 300 kb but less than about 400 kb, at least about 400 kb but less than about 500 kb, at
least about 500 kb but less than about le, at least about 1 Mb but less than about 1.5 Mb, at
least about 1.5 Mb but less than about 2 Mb, at least about 2 Mb but less than about 2.5 Mb,
or at least about 2.5 Mb but less than about 3 Mb. In one embodiment, the first and the
second expression constructs are on a single nucleic acid molecule. In one embodiment, the
target genomic locus comprises the Sry locus.
Further provided is a method for targeted genetic modification on the Y
chromosome of a non-human animal, comprising: (a) modifying a genomic locus of interest
on the Y chromosome of a non-human pluripotent cell according to the methods described
herein, y producing a genetically modified non-human pluripotent cell comprising a
targeted genetic modification on the Y some; (b) introducing the ed non-
human pluripotent cell of (a) into a non-human host embryo; and gestating the non-human
host embryo comprising the modified pluripotent cell in a surrogate mother, wherein the
surrogate mother produces F0 progeny comprising the ed genetic modification, wherein
the ed genetic modification is e of being transmitted through the germline. In
one embodiment, the genomic locus of interest comprises the Sry locus.
Methods and compositions are provided for generating targeted genetic
modifications on the Y chromosome. Compositions include an in vitro culture comprising an
XY pluripotent and/or totipotent animal cell (i.e., XY ES cells or XY iPS cells) having a
modification that decreases the level and/or activity of an Sry protein; and, culturing these
cells in a medium that promotes development of XY F0 fertile s. Such compositions
find use in various methods for making a fertile female XY non-human s in an F0
generation.
BRIEF DESCRIPTION OF THE FIGURES
provides a schematic of the CRISPR Cas9/gRNA targeting the mouse Sry
gene. VG-l (SEQ ID NO:10); VG-2 (SEQ ID NO:1 l); VG-3 (SEQ ID NO:12). The primers
and probes indicated in are provided in SEQ ID NOS: 13-29.
provides a schematic of ing the Sry gene with TALEN and CRISPR
using a lacZ reporter gene. The Sry gene was targeted with both a LTVEC and a armed
vector (smallTVEC) having homology arms smaller than a LTVEC in order to avoid
nging loci on the Y chromosome.
rates LacZ sion in embryos.
provides a schematic of a large deletion of greater than 500 kb on the Y
chromosome mediated by ZFNs or by CRISPR guide RNAs in combination with Cas9 DNA
endonuclease.
A, B, and C provides the sequencing confirmation of the large Y
chromosome on in various clones. A is the sequencing result for clone l-D5. The
Kdm5 Up and Uspy9 down sequence is provided in SEQ ID NO:30; l-D5 l500F (SEQ ID
NO:3l); l-D5 1000R (SEQ ID NO:32); is the sequencing result for clone 5-C4. The
2015/038001
Kdm5 Up and Uspy9 down sequence is provided in SEQ ID NO:33; l500F (SEQ ID NO:34);
1000R (SEQ ID NO:35); 1000F (SEQ ID N036); and is the sequencing result for
clone 6-Al2. The Kde Up and Uspy9 down sequence is provided in SEQ ID NO:37; l500F
(SEQ ID N038); 1000R (SEQ ID NO:39); 1000F (SEQ ID NO:40); 1500R (SEQ ID
. The boxed regions in and represent micro-homology regions.
DETAILED DESCRIPTION
DEFINITIONS
The terms “protein,” “polypeptide,” and “peptide,” used interchangeably herein,
e polymeric forms of amino acids of any length, including coded and non-coded amino
acids and chemically or biochemically modified or derivatized amino acids. The terms also
include polymers that have been modified, such as polypeptides having modified peptide
backbones.
The terms “nucleic acid” and “polynucleotide,” used interchangeably herein,
include ric forms of tides of any length, including ribonucleotides,
deoxyribonucleotides, or analogs or modified versions thereof. They e single-, double-
and multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, and
polymers comprising purine bases, pyrimidine bases, or other natural, chemically modified,
biochemically ed, non-natural, or derivatized nucleotide bases. For simplicity, nucleic
acid size may be referred to in bp whether the nucleic acid is in double-or single-stranded
form, in the latter case, the bp being those formed if and when the single-stranded nucleic
acid is duplexed with its exactly complementary strand.
“Codon optimization” generally includes a process of modifying a nucleic acid
sequence for enhanced expression in particular host cells by replacing at least one codon of
the native sequence with a codon that is more frequently or most frequently used in the genes
of the host cell while maintaining the native amino acid ce. For example, a nucleic
acid encoding a Cas protein can be modified to substitute codons having a higher frequency
of usage in a given prokaryotic or otic cell, including a bacterial cell, a yeast cell, a
human cell, a non-human cell, a mammalian cell, a rodent cell, a mouse cell, a rat cell, a
hamster cell, or any other host cell, as compared to the naturally occurring nucleic acid
ce. Codon usage tables are readily available, for e, at the “Codon Usage
se.” These tables can be adapted in a number of ways. See Nakamura et a1. (2000)
Nucleic Acids Research 28:292. Computer thms for codon optimization of a particular
sequence for expression in a particular host are also available (see, e. g., Gene Forge).
“Operable linkage” or being “operably linked” includes juxtaposition of two or
more components (e.g., a promoter and another sequence element) such that both ents
function normally and allow the ility that at least one of the components can mediate a
function that is exerted upon at least one of the other components. For example, a promoter
can be operably linked to a coding sequence if the promoter controls the level of transcription
of the coding sequence in response to the ce or absence of one or more transcriptional
regulatory factors.
“Complementarity” of nucleic acids means that a nucleotide sequence in one
strand of c acid, due to orientation of its nucleobase groups, forms hydrogen bonds with
another sequence on an opposing nucleic acid strand. The complementary bases in DNA are
typically A with T and C with G. In RNA, they are typically C with G and U with A.
mentarity can be t or substantial/sufficient. Perfect complementarity between
two c acids means that the two nucleic acids can form a duplex in which every base in
the duplex is bonded to a complementary base by Watson-Crick pairing. "Substantial" or
"sufficient" complementary means that a sequence in one strand is not tely and/or
perfectly complementary to a sequence in an opposing strand, but that sufficient bonding
occurs between bases on the two strands to form a stable hybrid complex in set of
hybridization conditions (e.g., salt concentration and temperature). Such conditions can be
predicted by using the sequences and standard mathematical calculations to t the Tm of
ized strands, or by empirical determination of Tm by using routine methods. Tm
includes the temperature at which a population of hybridization complexes formed between
two c acid strands are 50% denatured. At a temperature below the Tm, ion of a
hybridization complex is favored, whereas at a temperature above the Tm, melting or
separation of the strands in the hybridization complex is favored. Tm may be estimated for a
nucleic acid having a known G+C t in an aqueous l M NaCl solution by using, e. g.,
Tm=81.5+0.4l(% G+C), although other known Tm computations take into account nucleic
acid structural characteristics.
"Hybridization condition" es the cumulative environment in which one
nucleic acid strand bonds to a second nucleic acid strand by complementary strand
interactions and hydrogen bonding to produce a hybridization complex. Such conditions
include the chemical components and their concentrations (e. g., salts, chelating agents,
formamide) of an aqueous or organic solution containing the nucleic acids, and the
temperature of the mixture. Other s, such as the length of tion time or reaction
chamber dimensions may contribute to the environment. See, e. g., Sambrook et al.,
Molecular Cloning, A Laboratory Manual, 2.sup.nd ed., pp. 1.90-1.91, 9.47-9.51, 1 1.47-
1157 (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989).
Hybridization requires that the two nucleic acids contain complementary
ces, although mismatches between bases are le. The conditions appropriate for
hybridization between two nucleic acids depend on the length of the nucleic acids and the
degree of complementation, variables well known in the art. The greater the degree of
complementation between two nucleotide sequences, the greater the value of the melting
temperature (Tm) for hybrids of nucleic acids having those sequences. For hybridizations
between nucleic acids with short stretches of complementarity (e.g. complementarity over 35
or fewer, 30 or fewer, 25 or fewer, 22 or fewer, 20 or fewer, or 18 or fewer nucleotides) the
on of ches becomes important (see Sambrook et al., supra, 11.7-11.8).
Typically, the length for a izable nucleic acid is at least about 10 nucleotides.
Illustrative minimum lengths for a hybridizable c acid include at least about 15
nucleotides, at least about 20 nucleotides, at least about 22 nucleotides, at least about 25
nucleotides, and at least about 30 nucleotides. Furthermore, the temperature and wash
solution salt concentration may be adjusted as ary according to factors such as length
of the region of complementation and the degree of complementation.
The sequence of polynucleotide need not be 100% complementary to that of its
target nucleic acid to be specifically hybridizable. Moreover, a polynucleotide may hybridize
over one or more segments such that ening or adjacent segments are not involved in the
hybridization event (e. g., a loop structure or hairpin structure). A polynucleotide (e. g.,
gRNA) can comprise at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or
100% sequence complementarity to a target region within the target nucleic acid sequence to
which they are targeted. For e, a gRNA in which 18 of 20 nucleotides are
complementary to a target region, and would therefore specifically hybridize, would
represent 90% complementarity. In this example, the remaining noncomplementary
nucleotides may be clustered or persed with mentary nucleotides and need not be
contiguous to each other or to complementary nucleotides.
Percent complementarity between particular stretches of nucleic acid sequences
within c acids can be determined routinely using BLAST programs (basic local
alignment search tools) and PowerBLAST programs known in the art (Altschul et a1. (1990)
J. M01. Biol. 215:403-410; Zhang and Madden (1997) Genome Res. 656) or by using
the Gap m (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics
Computer Group, University Research Park, n Wis.), using default settings, which
uses the algorithm of Smith and Waterman (Adv. Appl. Math., 1981, 2, 482-489).
The methods and compositions provided herein employ a variety of different
components. It is recognized hout the description that some components can have
active variants and fragments. Such components include, for example, Cas proteins, CRISPR
RNAs, trachNAs, and guide RNAs. Biological activity for each of these components is
described elsewhere herein.
nce identity" or "identity" in the context of two polynucleotides or
polypeptide sequences makes reference to the residues in the two sequences that are the same
when aligned for maximum correspondence over a specified comparison window. When
percentage of sequence identity is used in nce to proteins it is recognized that residue
positions which are not identical often differ by conservative amino acid substitutions, where
amino acid residues are substituted for other amino acid es with similar chemical
properties (e. g., charge or hydrophobicity) and therefore do not change the functional
properties of the molecule. When ces differ in vative substitutions, the percent
sequence identity may be adjusted upwards to correct for the conservative nature of the
substitution. Sequences that differ by such conservative substitutions are said to have
nce similarity" or "similarity." Means for making this adjustment are well known to
those of skill in the art. Typically, this involves scoring a conservative substitution as a
partial rather than a full mismatch, thereby increasing the percentage sequence identity.
Thus, for example, where an identical amino acid is given a score of 1 and a non-conservative
substitution is given a score of zero, a conservative substitution is given a score between zero
and 1. The scoring of conservative substitutions is calculated, e. g., as implemented in the
program PC/GENE (Intelligenetics, Mountain View, California).
"Percentage of sequence identity" includes the value determined by comparing
two lly aligned ces over a comparison , wherein the portion of the
polynucleotide sequence in the comparison window may comprise additions or deletions (i.e.,
gaps) as ed to the reference sequence (which does not se additions or deletions)
for optimal alignment of the two sequences. The percentage is calculated by determining the
number of positions at which the identical nucleic acid base or amino acid residue occurs in
both ces to yield the number of matched positions, dividing the number of matched
positions by the total number of positions in the window of comparison, and multiplying the
result by 100 to yield the percentage of ce identity.
2015/038001
Unless otherwise stated, sequence ty/similarity values include the value
obtained using GAP Version 10 using the following parameters: % identity and % similarity
for a nucleotide sequence using GAP Weight of 50 and Length Weight of 3, and the
nwsgapdna.cmp scoring matrix; % identity and % similarity for an amino acid sequence
using GAP Weight of 8 and Length Weight of 2, and the BLOSUM62 scoring matrix; or any
equivalent program f. "Equivalent program" includes any sequence comparison
m that, for any two ces in question, generates an alignment having identical
nucleotide or amino acid residue matches and an identical percent sequence identity when
compared to the corresponding alignment generated by GAP Version 10.
Compositions or methods “comprising” or “including” one or more recited
ts may include other elements not specifically recited. For example, a composition
that “comprises” or “includes” a protein may contain the protein alone or in combination with
other ingredients.
Designation of a range of values includes all integers within or defining the range,
and all subranges defined by integers within the range.
Unless ise apparent from the context, the term “about” encompasses values
within a standard margin of error of measurement (e. g., SEM) of a stated value.
The singular forms of the articles “a,” “an,” and “the” include plural references
unless the context clearly dictates ise. For example, the term “a Cas protein” or “at
least one Cas protein” can e a plurality of Cas proteins, including mixtures f.
1. Methods and itions to Make a Fertile Female XY Animal in an F0 Generation
Methods for making non-human animals from donor ES cells and host embryos
are known. Donor ES cells are selected for certain characteristics that enhance the ability of
the cells to populate a host embryo and thus contribute in part or in substantial part to an
animal formed by the donor ES cells and the host embryo. The animal formed may be male
or female, based in large part on the genotype of the ES cell (e. g., XY or XX).
The ty of ES cell lines for making transgenic animals have a male XY
genotype. e of the dominance of the Y chromosome in mammalian sex determination,
when XY ES cells are introduced into a blastocyst host embryo and gestated, they nearly
always produce in the first generation (F0) phenotypically male animals that are chimeras,
i.e., that contain cells derived from the male donor ES cell (XY) and cells derived from the
host embryo, which can be either male (XY) or female (XX). XY ES cells, when introduced
2015/038001
into an 8-cell host embryo by the Mouse method and gestated, can produce in the first
generation (F0) phenotypically male animals that are fully derived from the XY ES cells.
W0201 1/156723 es s and compositions which employ a culture
media for maintaining XY donor cells in culture such that after introduction of the XY donor
cells into a host embryo and gestation in a suitable host, fertile XY female animals are
produced in the F0 population. Such compositions find use in making Fl progeny that are
homozygous for the given targeted genetic modification.
The instant application provides s and compositions that employ a
combination of XY donor cells having a modification that decreases the level and/or activity
of the Sry protein in combination with a culture media that promotes the production of
anatomically normal, e and fecund, XY F0 females. Such s and compositions
allow for making a fertile female XY man animal in an F0 generation. The
combination of XY ES cells having a modification that decreases the level and/or activity of
the Sry protein in combination with the culture media described herein significantly increases
the percentage of fertile female XY y in the F0 generation. Methods for the
ent male to female sex conversion are valuable to the domestic animal industry. For
example, female calves are much more valuable to the dairy cattle industry than males. The
same is true for poultry. For breeding es, whether it be cattle or hogs or sheep, it is
preferred to breed many females to only a few bulls, boars, or rams. Thus, the various
methods provided herein find use in various commercially important breeding industries.
Methods and compositions are also provided for making a XY embryonic stem
(ES) cell line capable of producing a fertile XY female man mammal in an F0
generation without culturing in a feminizing media. In such methods, the XY ES cell line
having a modification that decreases the level and/or activity of an Sry protein can produce
an ES cell line capable of producing a fertile XY female non-human mammal in an F0
generation in the absence of a feminizing media provided elsewhere herein (e. g., by culturing
in a base medium, such as DMEM, described elsewhere herein).
A. Animal XY Cells Having a Modification that Decreases the Level and/or Activity
of an Sry Protein
Various itions and methods are provided herein which comprise various
XY pluripotent and/or totipotent cells from an animal. The term “pluripotent cell” as used
herein es an undifferentiated cell that possesses the ability to develop into more than
one differentiated cell types. Such pluripotent and/or totipotent XY cells can be, for example,
an embryonic stem (ES) cell or an induced pluripotent stem (iPS) cell. The term "embryonic
stem cell" or "ES cell" as used herein includes an -derived totipotent or pluripotent
cell that is capable of contributing to any tissue of the developing embryo upon introduction
into an embryo.
The term "animal," in reference to cells, pluripotent and/or totipotent cells, XY
cells, ES cells, iPS cells, donor cells and/or host embryos, includes mammals, fishes, and
birds. Mammals include, e. g., humans, non-human es, monkey, ape, cat dog, horse,
bull, deer, bison, sheep, rodents (e. g., mice, rats, hamsters, guinea pigs), livestock (e. g.,
bovine species, e. g., cows, steer, etc.; ovine species, e. g., sheep, goats, etc.; and porcine
species, e.g., pigs and boars). Birds e, e.g., chickens, turkeys, ostrich, geese, ducks, etc.
icated animals and agricultural animals are also included. The phrase "non-human
animal," in reference to cells, XY cells, ES cells, donor cells and/or host embryos, excludes
humans.
In specific embodiments, the pluripotent cell is a human XY ES cell, a human XY
iPS cell, a human adult XY ES cell, a developmentally cted human progenitor ES cell, a
non-human XY ES cell, a non-human XY iPS cell, a rodent XY ES cell, a rodent XY iPS
cell, a mouse XY ES cell, a mouse XY iPS cell, a rat XY ES cell, a rat XY iPS cell, a hamster
XY ES cell, a hamster XY iPS cell, a monkey XY ES cell, a monkey XY iPS cell, an
agricultural mammal XY ES cell, an agricultural XY iPS cell, a domesticated mammal XY
ES cell, or a domesticated XY iPS cell. Moreover, the XY ES cell or the XY iPS cell can be
from an inbred strain, a hybrid strain or an outbred strain. It is further recognized that the
pluripotent and/or totipotent XY cells can comprise an XYY karyotype or an XXY
ype.
Mouse pluripotent and/or totipotent cells (i.e., XY ES cells or XY iPS cells) can
be from a 129 strain, a C57BL/6 strain, a mix of 129 and C57BL/6, a BALB/c strain, or a
Swiss Webster strain. In a specific embodiment, the mouse is 50% 129 and 50% C57BL/6.
In one embodiment, the mouse is a 129 strain selected from the group consisting of a strain
that is 129P1
, 129P2, 129P3, 129X1 , 129S1 (e.g., 129S1/SV, 129S1/Svlm), 129S2, 129S4,
129S5, 129S9/SvaH, 129S6 (129/SvaTac), 129S7, 129S8, 129T1 , 129T2. See, for
example, Festing et al. (1999) Mammalian Genome 10:836). In one embodiment, the mouse
is a C57BL strain, and in a specific embodiment is from C57BL/A, C57BL/An, GrFa,
C57BL/Kal_wN, C57BL/6, 6J, C57BL/6ByJ, C57BL/6NJ, C57BL/6NTac,
C57BL/10, C57BL/10ScSn, C57BL/10Cr, or C57BL/Ola. In a ic embodiment, the
mouse is a mix of an entioned 129 strain and an aforementioned C57BL/6 strain. In
another specific embodiment, the mouse is a mix of aforementioned 129 s, or a mix of
aforementioned BL/6 strains. In a specific embodiment, the 129 strain of the mix is a 129S6
(129/SvaTac) strain. In some embodiments, the mouse XY ES cell comprises a Y
some derived from the 129 strain.
In yet another embodiment, the XY mouse ES cell is a VGF1 mouse ES cell.
VGF1 (also known as F1H4) mouse ES cells were derived from hybrid embryos produced by
ng a female C57BL/6NTac mouse to a male 129S6/SvaTac mouse. Therefore, VGF1
ES cells contain a Y chromosome from 129S6/SvaTac mouse. See, for example, Auerbach,
W. et a1. (2000) Establishment and chimera analysis of 129/Sva- and C57BL/6-derived
mouse embryonic stem cell lines. Biotechniques 29, 1024—1028, 1030, 1032, herein
incorporated by reference in its ty.
A rat pluripotent and/or totipotent cell (i.e., XY ES cell or XY iPS cell) can be
from any rat strain, including but not limited to, an ACI rat strain, a Dark Agouti (DA) rat
strain, a Wistar rat strain, a LEA rat strain, a Sprague Dawley (SD) rat strain, or a Fischer rat
strain such as Fisher F344 or Fisher F6. Rat pluripotent and/or totipotent cells (i.e., XY ES
cells or XY iPS cells) can also be obtained from a strain derived from a mix of two or more
strains recited above. In one embodiment, the rat pluripotent and/or totipotent cell (i.e., XY
ES cell or XY iPS cell) is derived from a strain selected from a DA strain and an ACI strain.
In a specific embodiment, the rat otent and/or totipotent cell (i.e., XY ES cell or XY
iPS cell) is derived from an ACI strain. The ACI rat strain is characterized as having black
agouti, with white belly and feet and an RT] haplotype. Such s are available from a
variety of sources including Harlan Laboratories. In other embodiments, the various rat
pluripotent and/or totipotent cell (i.e., XY ES cell or XY iPS cell) are from a Dark Agouti
(DA) rat strain, which is characterized as having an agouti coat and an RT] ype.
Such rats are available from a variety of sources including Charles River and Harlan
Laboratories. In a further embodiment, the rat pluripotent and/or tent cells (i.e., XY ES
cells or XY iPS cells) are from an inbred rat strain. In specific embodiments the rat ES cell
line is from an ACI rat and comprises the ACI.G1 rat ES cell. In another ment, the rat
ES cell line is from a DA rat and comprises the DA.2B rat ES cell line or the DA.2C rat ES
cell line. See, for example, US. Utility Application No. 14/185,703, filed on ry 20,
2014 and herein incorporated by nce in its entirety.
In s embodiments, the pluripotent and/or totipotent cell (i.e., XY ES cell or
XY iPS cell), the donor cell and/or the host embryo are not from one or more of the
following: Akodon spp., Myopus spp., Microtus spp., Talpa spp. In various embodiments, the
donor cell and/or the host embryo are not from any species of which a normal wild-type
characteristic is XY female fertility. In various embodiments, where a genetic modification
is present in the pluripotent and/or totipotent cell (i.e., XY ES cell or XY iPS cell), the donor
cell or the host embryo, the genetic modification is not an XYY or XXY, a Tdy-negative sex
reversal, Tdy-positive sex reversal, an X0 modification, an aneuploidy, an fgf9'/' genotype, or
a SOX9 modification.
The pluripotent and/or totipotent XY cells (i.e., an XY ES cell or an XY iPS cell)
employed in the methods and compositions have a genetic modification that results in a
decreased level and/or activity of the Sry protein. The “Sex Determining Region Y” n
or the “Sry” n is a transcription factor that is a member of the high mobility group
(HMG)-box family of DNA-binding proteins. Sry is the testis-determining factor that
initiates male sex determination. The sequence of the Sry protein from a variety of sms
is known, including from mouse (Accession No. Q05738); rat (GenBank: CAA61882.1)
human (Accession No. Q05066); cat (Accession No. Q67C50), and horse (Accession No.
P36389), each of which is herein incorporated by reference.
In general, the level and/or activity of the Sry protein is decreased if the protein
level and/or the activity level of the Sry protein is statistically lower than the protein level of
Sry in an appropriate l cell that has not been genetically ed or mutagenized to
inhibit the sion and/or activity of the Sry protein. In specific embodiments, the
concentration and/or activity of the Sry n is decreased by at least 1%, 5%, 10%, 20%,
%, 40%, 50%, 60%, 70%, 80%, or 90% relative to a control cell which has not been
modified to have the decreased level and/or activity of the Sry protein.
A ct cell” is one in which a genetic tion, such as a genetic
modification disclosed herein has been effected, or is a cell which is descended from a cell so
d and which comprises the alteration. A “control” or “control cell” provides a reference
point for measuring changes in phenotype of the subject cell. In one embodiment, a control
cell is as closely matched as possible with the cell with reduced Sry activity except it lacks
the genetic modification or mutation resulting in the d activity (for example, the
respective cells can originate from the same cell line). In other instances, the control cell may
comprise, for example: (a) a wild-type cell, i.e., of the same genotype as the ng material
for the genetic alteration which resulted in the subject cell; (b) a cell of the same genotype as
the starting material but which has been genetically modified with a null uct (i.e. with a
construct which has no known effect on the trait of st, such as a construct comprising a
marker gene); (c) a cell which is a non-genetically modified progeny of a subject cell (i.e.,
the control cell and the t cell originate from the same cell line); (d) a cell genetically
identical to the subject cell but which is not exposed to ions or stimuli that would
induce expression of the gene of interest; or (e) the subject cell itself, under conditions in
which the genetic cation does not result in an alteration in expression of the
polynucleotide of interest.
The expression level of the Sry polypeptide may be measured directly, for
e, by assaying for the level of the Sry polypeptide in the cell or organism, or
indirectly, for example, by measuring the activity of the Sry polypeptide. Various methods
for determining the activity of the Sry protein are known. See, Wang et a1. (2013) Cell
153:910-918, Mandalos et a1. (2012) PLOS ONE 7:e45768:1-9, and Wang et al. (2013) Nat
Biotechnol. 31:530-532, each of which is herein incorporated by reference.
In other instances, cells having the targeted genetic modification that reduces the
activity and/or level of the Sry polypeptide are selected using methods that include, but are
not limited to, Southern blot is, DNA sequencing, PCR is, or phenotypic
analysis. Such cells are then employed in the s methods, compositions and kits
described herein.
A targeted genetic modification can comprise a targeted alteration to a
cleotide of interest including, for example, a targeted alteration to a target genomic
locus on the Y chromosome, a ed alteration to the Sry gene, or a targeted alteration to
other desired polynucleotides. Such ed modifications include, but are not limited to,
additions of one or more tides, deletions of one or more nucleotides, substitutions of
one or more nucleotides, a knockout of the polynucleotide of interest or a portion thereof, a
knock-in of the polynucleotide of interest or a portion thereof, a replacement of an
endogenous nucleic acid sequence with a heterologous nucleic acid sequence, or a
combination thereof. In ic embodiments, at least 1, 2, 3, 4, 5, 7, 8, 9, 10 or more
nucleotides are changed to form the targeted genomic cation.
A decrease in the level and/or activity of the Sry protein can result from a genetic
modification in the Sry gene (i.e., a genetic modification in a regulatory region, the coding
region, and/or introns etc). Such genetic modifications include, but are not limited to,
additions, deletions, and substitutions of nucleotides into the genome. Such genetic
modifications can e an alteration of the Sry gene, including, for example, an insertion
of one or more nucleotides into the Sry gene, a deletion of one or more nucleotides from the
Sry gene, a substitution of one or more nucleotides in the Sry gene, a knockout of the Sry
gene or a portion thereof, a n of the Sry gene or a portion thereof, a replacement of an
endogenous nucleic acid sequence with a heterologous nucleic acid sequence, or a
combination thereof. Thus, in specific embodiments, the activity of an Sry polypeptide may
be reduced or eliminated by disrupting the gene encoding the Sry polypeptide. In specific
embodiments, at least 1, 2, 3, 4, 5, 7, 8, 9, 10 or more nucleotides are d in the Sry
gene. s methods can be used to generate the additional targeted genetic modification.
See, for example, Wang et a1. (2013) Cell 153:910-918, Mandalos et a1. (2012) PLOS ONE
7:e45768:1-9, and Wang et al. (2013) Nat Biotechnol. 31:530-532, each of which is herein
incorporated by reference. In on, the various s described herein to modify
genomic locus on the Y chromosome can be used to introduce targeted genetic modification
to the Sry gene.
In other embodiments, the activity and/or level of the Sry polypeptide is reduced
or eliminated by introducing into the cell a polynucleotide that ts the level or activity of
the Sry polypeptide. The polynucleotide may inhibit the expression of the Sry polypeptide
directly, by preventing translation of the Sry messenger RNA, or indirectly, by encoding a
polypeptide that inhibits the transcription or translation of the gene encoding an Sry protein.
In other embodiments, the activity of Sry polypeptide is reduced or eliminated by introducing
into the cell a ce encoding a polypeptide that inhibits the activity of the Sry
polypeptide.
In one embodiment, the XY otent and/or totipotent cells (i.e., XY ES cell or
XY iPS cell) comprise a conditional Sry allele that reduces the activity and/or level of the Sry
protein. A “conditional Sry allele” includes a modified Sry gene designed to have the
decreased level and/or activity of the Sry protein at a desired developmental time and/or
within a d tissue of st. Reduced level and/or activity can be compared with a
control cell lacking the modification giving rise to the conditional allele, or in the case of
d activity at a desired developmental time with ing and/or following times, or in
the case of a desired tissue, with a mean activity of all tissues. In one embodiment, the
conditional Sry allele comprises a ional null allele of Sry that can be switch off at a
d developmental time point and/or in specific tissues. Such a conditional allele can be
used to create fertile XY females derived from any gene-targeted clone. As described
elsewhere herein, such a method enables the creation of a desired homozygous genetic
modification in the F1 generation. Such methods provide a quick look at the phenotype
t having to breed to the F2 generation.
In a non-limiting embodiment, the conditional Sry allele is a multifunctional
, as described in US 2011/0104799, which is incorporated by reference in its entirety.
2015/038001
In specific embodiments, the conditional allele comprises: (a) an actuating sequence in sense
orientation with t to transcription of a target gene, and a drug selection cassette (DSC)
in sense or antisense orientation; (b) in antisense orientation a nucleotide sequence of st
(NSI) and a conditional by inversion module (COIN, which utilizes an exon-splitting intron
and an invertible genetrap-like module; see, for example, US 2011/0104799, which is
incorporated by reference in its entirety); and (c) recombinable units that recombine upon
exposure to a first recombinase to form a conditional allele that (i) lacks the actuating
ce and the DSC, and (ii) contains the NSI in sense orientation and the COIN in
antisense ation.
The conditional allele of the Sry gene can be ted in any cell type, and is not
limited to an XY pluripotent and/or totipotent cell. Such cells types along with non-limiting
methods to target a genomic locus on the Y chromosome are discussed in further detail
elsewhere herein.
As discussed elsewhere herein, the pluripotent and/or totipotent XY cell (i.e., an
XY ES cell or an XY iPS cell) having genetic modification that decreases the level and/or
activity of the Sry protein can further comprise at least one onal targeted genetic
modification to a polynucleotide of st. The at least one onal targeted genetic
cation can comprise a substitution of one or more nucleic acids, a replacement of an
endogenous nucleic acid sequence with a heterologous nucleic acid sequence, a knockout,
and a knock-in. The additional targeted genetic modification can be on the Y chromosome,
the X chromosome or on an autosome. Various s can be used to generate the
onal targeted genetic modification, including employing targeting plasmids and large
targeting vectors as discussed ere herein. See, also, [[820080092249,
WG/1999/005266A2, USEOOL’EQIWZ’éS’O, WO/2008/017234Al, and US Patent No. 7,612,250,
each of which is herein incorporated by reference, for methods related to nuclear transfer. In
addition, the various methods described herein to modify genomic locus on the Y
chromosome (i.e., the Sry gene) can also be used to introduce targeted genetic modifications
to polynucleotides of interest that are not d on the Y chromosome.
B. Mediafor Culturing the Pluripotent and/or Totipotent XY Cells Having a
Modification that Decreases the Level and/or Activity ofan Sry Protein
The culture media employed in the various s and compositions that
promote XY fertile female in the F0 generation is such that it maintains the pluripotent and/or
totipotent cells (i.e., ES cell, iPS cells, XY ES cells, XY iPS cells, etc.). The terms
WO 00805
“maintain77 ll
, ining" and "maintenance" refer to the stable preservation of at least one
or more of the characteristics or phenotypes of pluripotent and/or totipotent cells described
herein ding ES cells or iPS cells). Such ypes can e maintaining
pluripotency and/or totipotency, cell morphology, gene expression profiles and the other
functional characteristics of the cells. The terms “maintain”, "maintaining" and
"maintenance" can also encompass the propagation of cells, or an increase in the number of
cells being cultured. The terms further contemplate culture conditions that permit the cells to
remain pluripotent, while the cells may or may not continue to divide and increase in number.
In some embodiments, the XY cells having the genetic modification that reduces
the level and/or activity of the Sry protein are maintained by ing in any base medium
known in the art (e. g., DMEM) that is suitable for use (with added supplements) in growing
or maintaining the pluripotent and/or totipotent cells (i.e., ES cell, iPS cells, XY ES cells, XY
iPS cells, etc.) in culture. In such cases, the cultured XY ES cells have the potential to
develop into fertile female animals but still retain pluripotency and/or totipotency, such that
the cells can be implemented into a recipient embryo and give rise to a fertile female
progeny.
In other embodiments, XY cells having the c modification that s the
level and/or activity of the Sry n are maintained by culturing in a medium as further
defined below for sufficient time that some of the cells convert to KY cells with the potential
to develop into fertile female animals but still retain pluripotency and/or totipotency, such
that the cells can be implemented into a recipient embryo and give rise to a fertile female
progeny.
The medium employed to maintain the XY pluripotent and/or totipotent cells (i.e.,
XY ES cells, XY iPS cells, etc.) having the genetic modification that reduces the level and/or
activity of the Sry protein promotes the pment of XY F0 fertile females. Thus,
ing in such a medium increases the number of XY F0 e females that are obtained
when compared to culturing in an appropriate control medium (such as, for example, one
based on DMEM). Thus, an increased number of XY F0 fertile females can comprise at least
%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 81%, 82%, 83%, 84%, 85%,
86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of
the F0 non-human animals (following introduction of the non-human animal XY ES cells
into a host embryo and gestation of the host embryo) are XY females and which upon
attaining sexual maturity the F0 XY female non-human animal is fertile.
The phrase "base medium" or "base media" es, for example, a base medium
known in the art (e. g., DMEM) that is suitable for use (with added ments) in growing
or ining the pluripotent and/or totipotent cells (i.e., ES cell, iPS cells, XY ES cells, XY
iPS cells, etc.) in culture. Base media suitable for making a fertile XY female (i.e., "low-salt
DMEM" or “low-osmolality medium”) differs from base media typically used to maintain ES
cells in culture. For purposes of discussing base media in general, base media that are not
suitable for making fertile XY females are described in this section as "DMEM" and in Table
1 (e. g., typical DMEM media). For purposes of discussing base media suitable for making
fertile XY females, the phrase "low-salt DMEM" or “low-osmolality DMEM” is used.
Differences between base media typically used to maintain pluripotent and/or totipotent cells
in culture (e. g., DMEM) and base media suitable for making fertile XY females (e. g., "low-
salt DMEM") are articulated herein. The phrase "low-salt DMEM" is used for convenience;
suitable DMEM for making e XY females exhibits characteristics not limited to "low-
salt," but includes those bed herein. For example, the DMEM shown in Table 1 can be
made suitable for making fertile XY females by altering the sodium de and/or sodium
bicarbonate concentrations as provided for herein, which will also result in a different
osmolality and a different conductivity as compared with the DMEM shown in Table 1. An
example of base medium is Dulbeco's ed Eagle's Medium (DMEM), in various forms
(e. g., ogen DMEM, Cat. No. 1 1971 -025) (Table 1). A suitable low-salt DMEM is
available commercially as KO-DMEMTM (Invitrogen Cat. No. 10829-018). Base medium is
lly supplemented with a number of supplements known in the art when used to
maintain cells in e for use as donor cells. Such supplements are indicated as
"supplements" or "+ supplements" in this disclosure.
Table 1 : DMEM Base Media for Maintaining or Culturing Pluripotent and/or Totipotent
Cells
Component Mg/L mM
Glycine 30 0.4
L-Arginine0HCI 84 0.398
L-Cystine02HCI 63 0.201
L-Glutamine 584 4
L—Histidine-HCI-H2O 42 0.2
L—Isoleucine 105 0.802
L-Leucine 105 0.802
L-Lysine0HCI 146 0.798
L-Methionine 30 0.201
L-Phenylalanine 66 0.4
L-Serine 42 0.4
L—Threonine 95 0.798
tophan 16 0.0784
L-Tyrosine um salt dihydrate 104 0.398
ne 94 0.803
e chloride 4 0.0286
D-Calcium pantothenate 4 8.39 X 10'3
Folic Acid 4 9.07 x 10'3
Niacinamide 4 0.0328
Pyridoxine°HCI 4 0.0196
Riboflavin 0.4 1.06 x 10'3
Thiamine°HCI 4 0.0119
i-Inositol 7.2 0.04
Calcium Chloride (CaClz) (anhydrous) 200 1.8
Ferric Nitrate (Fe(N03)3.9HzO) 0.1 2.48 x 10-4
Magnesium Sulfate (MgSO4) (anhyd.) 97.67 0.814
Potassium Chloride (KCI) 400 5.33
D-Glucose (Dextrose) 4500 25
Phenol Red 15 0.0399
NaCL/NaHCO3 Content of DMEM
Sodium onate (NaHCOg) 3700 44.05
Sodium Chloride (NaCl) 6400 110.34
NaCl/NaHCO3 Content of Low-salt
DMEM
Sodium Bicarbonate (NaHCOg) <3700 <44.05
Sodium Chloride (NaCl) <6400 <110.34
The term "supplements" or the phrase "+ supplements," includes elements added
to base medium for growing or maintaining pluripotent and/or totipotent cells (i.e., XY ES
cell or XY iPS cells) in culture, e. g., for maintaining pluripotency or totipotency of donor
cells in culture. For example, media supplements suitable for growing or maintaining
otent and/or totipotent cells in culture e, but are not limited to, fetal bovine serum
(FBS), glutamine, antibiotic(s), penicillin and streptomycin (e. g., penstrep), pyruvate salts
(e. g., sodium pyruvate), nonessential amino acids (e. g., MEM NEAA), 2-mercaptoethanol,
and Leukemia Inhibitory Factor (LIF).
In one embodiment, the base medium comprises one or more supplements le
for maintaining pluripotent cells in culture, including for example, XY ES cells or XY iPS
cells having a reduced capacity to contribute to the male sex determination developmental
program after injection into an embryo and intrauterine er to a surrogate mother mouse.
In a specific embodiment, the one or more ments suitable for maintaining
the pluripotent cell in e are PBS (90 ml PBS/0.5L base medium), glutamine (2.4
mmoles/0.5 L base medium), sodium pyruvate (0.6 mmoles/0.5L base medium), nonessential
amino acids (< 0.1 mmol/0.5 L base medium), 2-mercaptoethanol, LIF, and one or more
antibiotics.
In other embodiments, the media for maintaining pluripotent cells in culture,
including for example, XY ES cells or XY iPS cells having a reduced capacity to contribute
to the male sex ination developmental program after injection into an embryo and
intrauterine transfer to a surrogate mother mouse, comprises about 500 ml of base medium in
which the ing supplements are added: about 90 ml FBS (e.g., Hylcone FBS Cat. No.
SH30070.03), about 2.4 millimoles of glutamine (e. g., about 12 ml of a 200 mM glutamine
solution, e. g., Invitrogen Cat. No. 081, penicillin:streptomycin (e. g., 60,000 units of
Penicillin G sodium and 60 mg of streptomycin sulfate, with about 51 mg of NaCl; e. g., about
6 ml. of Invitrogen pennstrep, Cat. No. 15140-122), about 0.6 millimoles of sodium pyruvate
(e. g., 6 ml. of 100 mM sodium pyruvate, Invitrogen Cat. No. 1 1360-070), about 0.06
oles of ential amino acids (e. g., about 6 ml. of MEM NEAA, e. g., MEM NEAA
from Invitrogen Cat. No. 1 1 140-050), about 1.2 ml. 2-mercaptoethanol, and about 1.2
micrograms of LIF (e.g., about 120 microliters of a 106 units/mL LIF preparation; e. g., about
120 microliters of Millipore ESGROTM-LIF, Cat. No. ESGl 107). When composing base
media for maintaining XY ES or XY iPS cells for making e XY females, lly the
same supplements in about the same amounts are employed, but the composition of the base
medium will differ (from DMEM, e.g., from the medium described in the table above) and
the difference(s) correspond to the difference(s) taught herein.
In some embodiments, ments include Wnt-conditioned media, e.g., Wnt-3a
conditioned media.
In one embodiment, the pluripotent cell, including for example, an XY ES cell or
an XY iPS cell having a reduced capacity to contribute to the male sex determination
developmental program after injection into an embryo and intrauterine transfer to a surrogate
mother mouse, is maintained in an in vitro culture in a medium sing base medium and
supplements, wherein the base medium exhibits one or more of the following characteristics:
(a) an osmolality from about 200 mOsm/kg to less than about 329 mOsm/kg; (b) a
conductivity of about 11 mS/cm to about 13 mS/cm; (C) a salt of an alkaline metal and a
halide in a concentration of about 50mM to about 110 mM; (d) a carbonic acid salt
concentration of about 17mM to about 30 mM; (e) a total ne metal halide salt and
carbonic acid salt concentration of about 85mM to about 130 mM; and/or (f) a combination
of any two or more thereof. In other embodiments, the XY pluripotent and/or totipotent cells
(i.e., XY ES cell or XY iPS cell) is maintained in an in vitro e in a media as described
in WO201 1/156723, herein incorporated by reference in its entirety.
In one embodiment, the base medium is a low-salt DMEM. In a specific
embodiment, the low-salt DMEM has a NaCl concentration of 85-130 mM. In one
embodiment, the base medium is a low lity DMEM. In a specific embodiment, the
low osmolality DMEM has an osmolality of 250-310 mOsm/kg. In one embodiment, the
base medium is a low conductivity DMEM. In a specific embodiment, the low conductivity
DMEM has a conductivity of 11 -13 mS/cm.
In other embodiments, the base medium exhibits an osmolality of no more than
about 320, 310, 300, 290, 280, 275, 270, 260, 250, or 240 mOsm/kg. In one embodiment, the
base medium or the medium comprising the base medium and the supplements ts an
osmolality of no more than about 240-320, 250-310, 275-295, or 260-300 mOsm/kg. In a
specific embodiment, the base medium or the medium comprising the base medium and the
supplements exhibits an osmolality of about 270 mOsm/kg.
In other embodiments, the base medium ts a conductivity of no more than
about 10.0, 10.5, 1 1.0, 1 1.5, 12.0, 12.5, 13.0, 13.5, or 14.0 mS/cm. In one embodiment, the
base medium exhibits a conductivity of no more than about 10-14 mS/cm or 1 1 -13 mS/cm.
In a specific embodiment, the base medium exhibits a conductivity of about 12-13 mS/cm.
In a specific embodiment, the base medium exhibits a conductivity of about 12-13
mS/cm and an osmolality of about 260-300 mOsm/kg. In a further specific embodiment, the
base medium comprises sodium chloride at a concentration of about 90 mM NaCl. In a
further specific embodiment, the concentration of sodium chloride is about 70-95 mM. In a
further ic embodiment, the base medium comprises sodium bicarbonate at a
tration of less than about 35 mM. In a further specific embodiment, the concentration
of sodium onate is about 20-30 mM.
In one embodiment, the base medium exhibits a concentration of a salt of an
alkaline metal and a halide of no more than about 100 mM. In one embodiment, the salt of
the alkaline metal and the halide is NaCl. In one embodiment, the concentration of the salt of
the alkaline metal and halide is no higher than 90, 80, 70, 60, or 50 mM. In one embodiment,
the concentration in the base medium of the salt of the alkaline metal and halide is about 60-
105, 70-95, or 80-90 mM. In a specific embodiment, the concentration is about 85 mM.
In one embodiment, the base medium exhibits a tration of a salt of ic
acid. In one embodiment, the salt of carbonic acid is a sodium salt. In one embodiment, the
sodium salt is sodium bicarbonate. In one embodiment, the concentration of carbonic acid
salt in the base medium is no higher than 40, 35, 30, 25, or 20 mM. In one embodiment the
concentration of carbonic acid salt in the base medium is about 10-40, in another ment
about 20-30 mM. In a specific embodiment, the concentration is about 25 or 26 mM. In still
other embodiments, the sodium bicarbonate concentration is about 26 mM, about 18 mM,
about 18 mM to about 26 mM or about 18 mM to about 44 mM.
In one embodiment, the sum of the concentration of the salt of the ne metal
and halide and the salt of carbonic acid in the base medium is no more than 140, 130, 120,
110, 100, 90, or 80 mM. In one embodiment, the sum of the concentration of the salt of the
alkaline metal and halide and the salt of carbonic acid in the base medium is about 80-140,
85-130, 90-120, 95-120, or 100-120 mM. In a specific embodiment, the sum of the
concentration of the salt of the alkaline metal and halide and the salt of carbonic acid in the
base medium is about 115 mM.
In one embodiment, the molar ratio of the salt of the alkaline metal and halide and
the salt of carbonic acid is higher than 2.5. In one embodiment, the ratio is about 2.6-4.0, 2.8-
3.8, 3-3.6, or 3.2-3.4. In one embodiment, the ratio is 3.3-3.5. In a specific embodiment, the
ratio is 3.4.
In one embodiment, the base medium exhibits an osmolality of about 250-310
mOsm/kg, and a concentration of a salt of an alkaline metal and a halide of about 60-105
mM. In a further embodiment, the base medium has a concentration of a salt of ic acid
of about 20-30 mM. In a further ment, the sum of the concentrations of the salt of an
ne metal and halide and the salt of carbonic acid is about 80-140 mM. In a further
embodiment, the conductivity of the base medium is about 12-13 mS/cm.
In one embodiment, the base medium comprises about 50 i 5 mM NaCl and about
26 i 5 mM carbonate, with an osmolality of about 218 i 22 mOsm/kg. In a specific
embodiment, the base medium comprises about 3 mg/mL NaCl and 2.2 mg/mL sodium
bicarbonate, with an osmolality of about 218 g.
In another ment, the base medium comprises about 87 i 5 mM NaCl and
about 18 i 5 mM, with an osmolality of about 261 i 26 mOsm/kg. In a specific embodiment,
the base medium comprises about 5.1 mg/mL NaCl and about 1.5 mg/mL sodium
bicarbonate, with an osmolality of about 261 mOsm/kg.
In another embodiment, the base medium comprises about 110 i 5 mM NaCl and
about 18 i 5 mM carbonate, with an osmolality of about 294 i 29 mOsm/kg. In a specific
embodiment, the base medium comprises about 6.4 mg/mL NaCl and about 1.5 mg/mL
sodium bicarbonate, with an osmolality of about 294 mOsm/kg.
In another embodiment, the base medium ts about 87 i 5 mM NaCl and
about 26 i 5 mM carbonate, with an lity of about 270 i 27 mOsm/kg. In a specific
embodiment, the base medium exhibits about 5.1 mg/mL NaCl and about 2.2 mg/mL sodium
bicarbonate, with an osmolality of about 270 mOsm/kg.
In another embodiment, the base medium comprises about 87 i 5 mM NaCl,
about 26 i 5 mM ate, and about 86 i 5 mM e, with an osmolality of about 322 i
32 mOsm/kg. In a specific embodiment, the base medium comprises about 5.1 mg/mL NaCl,
about 2.2 mg/mL sodium bicarbonate, and about 15.5 mg/mL glucose, with an osmolality of
about 322 mOsm/kg.
Additional base media that can be employed in the various methods and
compositions disclosed herein e, a base medium comprising 50 i 5 mM NaCl and 26 i
mM carbonate, with an osmolality of 218 i 22 mOsm/kg. In a particular embodiment, the
base medium comprises about 3 mg/mL NaCl and 2.2 mg/mL sodium bicarbonate, with an
osmolality of about 218 mOsm/kg.
In other ments, the base medium comprises 50 i 5 mM NaCl and 26 i 5
mM carbonate, with an osmolality of 218 i 22 mOsm/kg. In a specific embodiment, the base
medium comprises about 3 mg/mL NaCl and 2.2 mg/mL sodium bicarbonate, with an
osmolality of about 218 mOsm/kg.
In other embodiments, high glucose DMEM media (LifeTech) with NaHC03
concentrations as disclosed herein, including, about 44mM, 26mM or 18mM, were
supplemented with 0.1mM nonessential amino acids, 1mM sodium pyruvate, 0.1mM 2-
mercaptoethanol, 2mM L-glutamine, l each penicillin and streptomycin (LifeTech),
% FBS (Hyclone), and 2000U/ml LIF pore).
C. Methodfor Making Targeted Genetic Modifications
Various methods for making targeted c modifications that se the level
and/or the activity of the Sry protein can be used. For example, in one ce, the targeted
genetic modification employs a system that will generate a targeted genetic modification via
a homologous recombination event. In other ces, the animal cell can be modified using
nuclease agents that generate a single or double strand break at a targeted genomic location.
The single or double-strand break is then repaired by the non-homologous end g
pathway (NHEJ). Such systems find use, for example, in generating targeted loss of function
genetic modifications. Non-limiting methods for generating such targeted genetic
modification are discussed in detail elsewhere herein, including, for example, the use of
ing plasmids, small targeting vectors (smallTVECs) or large targeting vectors. See,
also, Wang et al. (2013) Cell 0-918, Mandalos et al. (2012) PLOS ONE 7:e45768:1-9,
and Wang et al. (2013) Nat Biotechnol. 31:530-532, each of which is herein incorporated by
reference.
It is recognized that in specific embodiments, the targeted c modification of
the Sry gene and/or the targeted genetic modification of any other polynucleotide of interest
can occur while the pluripotent cell (i.e., ES cell) is being maintained in the culture media
described herein (e.g. a medium that promotes the development of XY F0 fertile s).
Alternatively, the targeted genetic modification of the Sry gene and/or any other
polynucleotide of interest can occur while the pluripotent cell (i.e., ES cell) is being
maintained in different culture media, and subsequently transferred to the culture media
disclosed herein (e. g. a medium that promotes the pment of XY F0 fertile females).
D. Method of Culturing and Maintaining a Pluripotent and/0r T0tip0tent Cell In
A method for maintaining or ing an XY pluripotent and/or totipotent cell
(i.e., an XY ES cell or an XY iPS cell) in an in vitro culture is provided, wherein the cell
comprises a modification that decreases the level and/or activity of an Sry n and the cell
is maintained in an in vitro culture under conditions described herein. Such methods of
ining or culturing an XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an
XY iPS cell) in an in vitro culture is such as to e an increase in the number XY F0
fertile female animals upon the introduction of the non-human animal XY ES cells into a host
embryo and following gestation of the host embryos.
While any media disclosed herein can be employed for such ining or
culturing methods, one non-limiting example, includes culturing in a medium sing a
base medium and supplements suitable for maintaining or culturing the XY pluripotent and/or
totipotent cell (i.e., an XY ES cell or an XY iPS cell) in culture, wherein the base medium or
the medium sing the base medium and the supplements exhibits an osmolality from
about 200 g to less than about 329 mOsm/kg.
In some embodiments, the base medium or the medium comprising the base
medium and the supplements exhibits one or more of the following characteristic: a
conductivity of about 11 mS/cm to about 13 mS/cm; a salt of an alkaline metal and a halide in
a concentration of about 50mM to about 110 mM; a carbonic acid salt tration of about
l7mM to about 30 mM; a total alkaline metal halide salt and carbonic acid salt tration
of about 85mM to about 130 mM; and/or a combination of any two or more thereof.
In one embodiment, the method comprises maintaining or culturing the XY
pluripotent and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell) in a suitable culture
medium that comprises a base medium and supplements, wherein the base medium or the
medium comprising the base medium and the supplements comprises an osmolality of about
240-320 mOsm/kg, a conductivity of about 10-14 mS/cm, an alkaline metal halide salt
concentration of about 50-105 mM, a salt of carbonic acid concentration of 10-40 mM, and/or
a combined alkaline metal salt and carbonic acid salt tration of about 80-140 mM. In
one embodiment, the XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an XY iPS
cell) is maintained in the medium (with supplements for maintaining ES cells) for a period of
l, 2, 3, 4, 5, 6, 7, 8, 9, 10, ll, 12, or 13 days, or 2 weeks, 3 weeks, or 4 weeks prior to
introduction into a host embryo. In a specific embodiment, the XY pluripotent and/or
totipotent cell (i.e., an XY ES cell or an XY iPS cell) is maintained in the medium (low-salt
base medium with ments for ining ES cells) for about 2-4 weeks prior to
introduction into the host embryo.
In another embodiment, the XY pluripotent and/or totipotent cell (i.e., an XY ES
cell or an XY iPS cell) is maintained in a medium with a low-salt base medium for at least 1,
2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 days, 2 weeks, 3 weeks, or 4 weeks prior to introducing the
donor cell into a host embryo. In a ic embodiment, the XY pluripotent and/or totipotent
cell (i.e., an XY ES cell or an XY iPS cell) is maintained in a medium with a low-salt base
medium at least 2-4 weeks prior to introduction of the cell into the host embryo.
In another embodiment, the XY pluripotent and/or totipotent cell (i.e., an XY ES
cell or an XY iPS cell) is ined (e. g., frozen) in a medium that promotes XY fertile F0
females and the donor cell is thawed in and maintained in the medium that promotes XY
fertile F0 females for at least 1, 2, 3, or 4 or more days before introducing the XY otent
and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell) into the host embryo. In a
specific embodiment, the XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an KY
iPS cell) is passaged at least once in a medium that promotes XY fertile F0 females, the cell
is frozen in the medium that promotes XY fertile F0 females, and the cell is thawed in a
medium that promotes XY fertile F0 females and grown for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12,
or 13 days, 2 weeks, 3 weeks, 4 weeks, or more prior to introduction into the host .
In still another embodiment, the XY pluripotent and/or totipotent cell (i.e., an XY
ES cell or an XY iPS cell) is maintained in the medium that promotes XY fertile F0 females
for a period of one, two, three, or four days prior to introduction into a host embryo. In one
embodiment, the XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell)
is maintained in the medium that promotes XY fertile F0 females for a period of 3 days.
In one embodiment, the XY pluripotent and/or totipotent cell (i.e., an XY ES cell
or an XY iPS cell) is maintained the medium that promotes XY e F0 females before
introduction into the host embryo for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 days, 2
weeks, 3 weeks, or 4 weeks or more. In a specific embodiment, the donor cell is maintained
in the medium that promotes XY fertile F0 females for at least a week before introduction
into the host embryo. In a specific embodiment, the XY pluripotent and/or totipotent cell
(i.e., an XY ES cell or an XY iPS cell) is ined in the medium that promotes XY fertile
F0 females for 2-4 weeks before introduction into the host embryo.
Thus, a method for maintaining or culturing an XY pluripotent and/or totipotent
cell (i.e., an XY ES cell or an XY iPS cell) in culture is provided, wherein the cell is
maintained under conditions that promote or favor development of a female XY animal
following uction of the XY cell into a host embryo and following ion in a suitable
female host.
In one aspect, a method for ining or culturing a donor XY pluripotent
and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell) in culture is provided, under
conditions as described herein, n ing introduction of the donor XY ES cell into a
host embryo to form a F0 embryo and gestation of the F0 embryo in a suitable , the F0
embryo develops into an F0 animal that is at least 70%, 75%, 80%, 85%, 90%, 91%, 92%,
93%, 94%, 95%, 96%, 97%, 98%, 99% or greater KY and is a female which, upon attaining
sexual maturity, is fertile.
E. Generating F0 Embryos and F1 Progeny Having A Targeted Genetic Modification
The various methods and compositions employing the XY pluripotent and/or
totipotent cell (i.e., an XY ES cell or an XY iPS cell) having a decreased level and/or ty
of Sry protein provided herein can be used to generate a genetically ed animal.
s methods for introducing genetic modifications are discussed in detail elsewhere
herein.
i. Methodfor Making a Fertile Female XYNon-Human Animal in an F0
Generation
A method for making a e female XY non-human animal in an F0 generation
is provided. Such s se: (a) maintaining or culturing a donor non-human
animal XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell) having a
modification that decreases the level and/or activity of an Sry n in a medium that
promotes the development of XY fertile female ES cells; (b) ucing the donor XY non-
human animal XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell)
into a host embryo; (c) gestating the host embryo; and, (d) obtaining an F0 XY female non-
human animal, wherein upon attaining sexual maturity the F0 XY female non-human animal
is fertile. In specific embodiments, the donor non-human animal XY donor cell can se
at least one additional targeted genetic modification in a polynucleotide of interest. Such
modifications are discussed in detail elsewhere herein.
The XY ES cells having a modification that decreases the level and/or activity of
an Sry protein can be ined without a lt medium and can develop into an XY
fertile female.
In some embodiments, the medium that promotes the development of XY fertile
F0 female s can comprise a low-salt based medium which comprises a base medium
and supplements suitable for maintaining or culturing the non-human mammalian ES cell in
culture, wherein the low-salt base medium exhibits a characteristic comprising one or more of
the following: an osmolality from about 200 mOsm/kg to less than about 329 mOsm/kg; a
conductivity of about 11 mS/cm to about 13 mS/cm; a salt of an alkaline metal and a halide in
a concentration of about 50 mM to about 110 mM; a carbonic acid salt concentration of about
17 mM to about 30 mM; a total alkaline metal halide salt and carbonic acid salt concentration
of about 85 mM to about 130 mM; and/or a combination of any two or more thereof.
In other embodiments, such methods for making a fertile female XY non-human
animal in an F0 tion can be performed using the mediums disclosed herein including,
but not limited to, (a) a base medium comprising 50 i 5 mM NaCl, 26 i 5 mM carbonate,
and 218 i 22 mOsm/kg; (b) a base medium comprising about 3 mg/mL NaCl, 2.2 mg/mL
sodium bicarbonate, and 218 mOsm/kg; (c) a base medium comprising 87 i 5 mM NaCl, 18
i 5 mM carbonate, and 261 i 26 mOsm/kg; (d) a base medium comprising about 5.1 mg/mL
WO 00805
NaCl, 1.5 mg/mL sodium bicarbonate, and 261 mOsm/k; (e) a base medium comprises 110 i
mM NaCl, 18 i 5 mM carbonate, and 294 i 29 mOsm/kg; (f) a base medium comprises
about 6.4 mg/mL NaCl, 1.5 mg/mL sodium bicarbonate, and 294 mOsm/kg; (g) a base
medium ses 87 i 5 mM NaCl, 26 i 5 mM carbonate, and 270 i 27 mOsm/kg; (h) a
base medium comprises about 5.1 mg/mL NaCl, 2.2 mg/mL sodium bicarbonate, and 270
mOsm/kg; (i) a base medium comprises 87 i 5 mM NaCl, 26 i 5 mM carbonate, 86 i 5 mM
glucose, and 322 i 32 mOsm/kg; and/or (1') a base medium comprises about 5.1 mg/mL NaCl,
2.2 mg/mL sodium bicarbonate, 15.5 mg/mL glucose, and 322 mOsm/kg.
The genetically modified XY pluripotent and/or totipotent cell (i.e., an XY ES cell
or an XY iPS cell) having a modification that ses the level and/or activity of an Sry
protein and having been ed in the medium that promotes the development of XY F0
fertile females can be implanted into a host embryo. Cells that have been ted into a
host embryo are referred to herein as “donor cells.” In specific embodiments, the genetically
modified XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell) is from
the same strain as the host embryo or from a different strain as the host embryo. se,
the surrogate mother can be from the same strain as the genetically modified XY pluripotent
and/or tent cell (i.e., an XY ES cell or an XY iPS cell) and/or the host embryo, or the
surrogate mother can be from a different strain as the genetically modified XY pluripotent
and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell) and/or the host embryo. In one
embodiment, the XY donor cell is implanted into an XX host embryo.
A variety of host embryos can be employed in the methods and compositions
sed herein. In some embodiments, the XY pluripotent and/or totipotent cells (i.e., the
XY ES cell or the XY iPS cell) having the targeted genetic modification resulting in a
decreased level and/or activity of the Sry n are introduced into a pre-morula stage
embryo from a corresponding organism, e. g., an 8-cell stage embryo. See, e.g., US
259, US 7,659,442, US 7,294,754, and US 2008-0078000 A1, all of which are
incorporated by reference herein in their entireties. In other embodiments, the donor ES cells
may be implanted into a host embryo at the 2-cell stage, 4-cell stage, 8-cell stage, 16-cell
stage, 32-cell stage, or 64-cell stage host embryo. In another embodiment, the host embryo is
a blastocyst. In one embodiment, the host embryo is in a stage selected from a astocyst
embryo, a pre-morula stage, a morula stage, an uncompacted morula stage, and a compacted
morula stage. In one embodiment, when employing a mouse embryo, the host embryo stage
is selected from a Theiler Stage 1 (TSl), a TS2, a TS3, a TS4, a TS5, and a TS6,
, with
nce to the Theiler stages described in Theiler (1989) "The House Mouse: Atlas of
Mouse Development," er-Verlag, New York. In a specific embodiment, the Theiler
Stage is selected from TSl, TS2, TS3, and a TS4. In one embodiment, the host embryo
comprises a zona pellucida, and the donor cell is an XY ES cell that is introduced into the
host embryo h a hole in the zona pellucida, while in other embodiments, the host
embryo is a zona-less embryo. In yet other specific embodiments, the morula-stage host
embryo is aggregated.
Nuclear transfer techniques can also be used to generate the genetically modified
animals. Briefly, methods for nuclear transfer include the steps of: (l) enucleating an oocyte;
(2) isolating a donor cell or nucleus to be combined with the enucleated oocyte; (3) inserting
the cell or nucleus into the enucleated oocyte to form a reconstituted cell; (4) implanting the
reconstituted cell into the womb of an animal to form an embryo; and (5) allowing the
embryo to develop. In such methods oocytes are generally retrieved from deceased animals,
although they may be isolated also from either oviducts and/or ovaries of live animals.
Oocytes can be d in a variety of medium known to those of ordinary skill in the art
prior to enucleation. ation of the oocyte can be performed in a number of s
well known to those of ordinary skill in the art. Insertion of the donor cell or s into the
enucleated oocyte to form a reconstituted cell is usually by microinjection of a donor cell
under the zona pellucida prior to fusion. Fusion may be d by application of a DC
electrical pulse across the contact/fusion plane rofusion), by exposure of the cells to
fusion-promoting chemicals, such as polyethylene , or by way of an inactivated virus,
such as the Sendai virus. A reconstituted cell is typically activated by electrical and/or non-
electrical means , during, and/or after fusion of the nuclear donor and recipient .
Activation methods include electric pulses, chemically induced shock, penetration by sperm,
increasing levels of divalent cations in the oocyte, and reducing phosphorylation of cellular
proteins (as by way of kinase inhibitors) in the oocyte. The activated reconstituted cells, or
s, are typically cultured in medium well known to those of ordinary skill in the art and
then transferred to the womb of an animal. See, for example, US20080092249,
WO/l999/005266A2, US20040177390, WO/2008/017234Al, and US Patent No. 7,612,250,
each of which is herein incorporated by reference.
The host embryo comprising the cally modified XY pluripotent and/or
totipotent cell (i.e., an XY ES cell or an XY iPS cell) having the decreased level and/or
activity of the Sry protein is incubated until the cyst stage and then implanted into a
surrogate mother to produce an F0 animal. Animals bearing the genetically modified
genomic locus can be identified via modification of allele (MOA) assay as described herein.
In one embodiment, the host embryo comprising the genetically modified XY
pluripotent and/or totipotent cells (i.e., an XY ES cell or an XY iPS cell) having the
sed level and/or activity of the Sry n is maintained in a medium that promotes the
development of XY e female ES cells (i.e., a low-salt base medium) for one, two, three,
or four or more days prior to implantation in a suitable host. Such methods provide for
favoring the generation of an F0 fertile female animal.
] In one embodiment, the cultured host embryo is implanted into a surrogate
mother, and the cultured host embryo is gestated in the surrogate .
In specific embodiments, upon introduction of the non-human animal XY
pluripotent and/or totipotent cells (i.e., an XY ES cell or an XY iPS cell) into a host embryo
and following ion of the host embryo, at least 15%, 20%, 30%, 40%, 50%, 60%, 70%,
75%, 80% 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% of the F0
non-human animals are XY s which upon attaining sexual maturity the F0 XY female
non-human mammal is fertile.
] Further provided is an F0 embryo comprising an inner cell mass having at least
one heterologous stem cell comprising an XY ES cell or XY iPS cell having a targeted
genetic modification that decreases the level and/or activity of the Sry protein.
] The various methods described herein to generate a fertile female XY non-human
animal in an F0 generation can employ XY pluripotent and/or totipotent cells (i.e., an XY ES
cell or an XY iPS cell) having (1) the genetic modification to reduce the level and/or activity
of the Sry polypeptide; and, in specific embodiments, (2) one or more additional targeted
genetic modification in a polynucleotide of interest. As outlined elsewhere herein, at least 1,
2, 3, 4, 5, 6, 7, 8, 9, 10 or more additional targeted c modifications can be made in the
XY pluripotent and/or totipotent cell (i.e., an XY ES cell or an XY iPS cell). In such
instances, the F0 fertile female XY non-human animal can comprises one or more of these
additional ed genetic modifications.
In other embodiments, the F0 fertile female XY non-human animal produces l, 2,
3, 4, 5, 6, 7, 8, or 9 litters during its me. In one embodiment, the F0 fertile female XY
non-human animal produces at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 offspring per litter. In one
embodiment, the F0 fertile female XY non-human animal produces about 4-6 offspring per
litter. In one embodiment, the F0 e female XY non-human animal produces 2-6 litters,
wherein each litter has at least 2, 3, 4, 5, or 6 offspring. In one embodiment, at least about
%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 85%, 90%, 95% or
100% of the offspring are XY fertile female offspring.
A method for generating a rodent litter (i.e., a mouse or a rat ) is also
provided and comprises introducing an XY pluripotent and/or totipotent donor cell (i.e., an
XY donor ES cell or XY donor iPS cell) having the decreased level and/or activity of Sry
protein prepared according to the methods set forth herein into host embryos, gestating the
embryos in a suitable segregate , and obtaining F0 progeny that comprises at least one
XY female rodent that upon reaching sexual maturity is a fertile XY female rodent. In one
embodiment, the percentage of F0 XY female s born that upon reaching sexual
maturity are fertile is about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%,
75%, 80%, 85%, 95% or 100%.
In other embodiments, the F0 progeny ed from such methods are about 3%,
about 10% or more, or about 63% or more derived from the genetically modified donor XY
cell.
The methods and compositions provided herein allow for at least 1%, 3%, 5%,
%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75% or greater of
the F0 animals to have the targeted genetic modification (i.e., the se in Sry protein
level and/or activity and/or the targeted genetic modification to a cleotide of interest)
to transmit the genetic modification to the F1 progeny.
In one embodiment, the F0 generation female XY non-human animal and/or the
male XY non-human animal is at least 90%, 92%, 94%, 96%, 98%, 99%, or 99.8% derived
from the donor cell. In one embodiment, the F0 female XY non-human animal and/or the F0
male XY non-human animal has a coat color that is 100% derived from the donor cell.
In one embodiment, the non-human female XY animal in the F0 generation is a
rodent (i.e., a mouse or a rat) and has a coat color 100% derived from the donor cell. In one
embodiment, the non-human female XY non-human animal formed in the F0 tion is at
least 90%, 92%, 94%, 96%, 98%, or 99.8% derived from the XY donor cell. In one
embodiment, the non-human female XY animal in the F0 tion is about 100% derived
from the donor cell. In one embodiment, the contribution of a host embryo cell to the non-
human female XY animal in the F0 generation is ined by a quantitative assay that is
capable of detecting 1 cell in 2,000 (0.05%), and no tissue of the female XY animal is
positive for host embryo cell contribution.
ii. Various Methods ding the Female Fertile XY F0 Generation
In specific embodiments, the resulting female fertile XY F0 generation derived
from the XY pluripotent and/or totipotent cells (i.e., the XY ES cell or XY iPS cell) having
2015/038001
the genetic modification that decreases the level and/or activity of the Sry protein is crossed
to an animal to obtain F1 generation ing. In ic embodiments, the female fertile
XY F0 is crossed to a wild type animal. In one embodiment, the female XY F0 non-human
mammal is fertile when crossed to a wild type mouse. In specific embodiments, the wild type
mouse is C57BL/6. The F1 progeny can be genotyped using ic primers and/or probes
to determine if the targeted genetic modification comprising the decreased level and/or
activity of the Sry protein is present. Moreover, if additional targeted genetic modifications
were present in the F0 generation, the F1 progeny can be genotyped using specific primers
and/or probes that determine if such modifications are t. An riate Fl progeny
for a desired use can then be identified. In specific embodiments, Fl y g the
genetic modification that reduced the level and/or activity of the Sry protein are selected. In
other embodiments, Fl progeny lacking the genetic modification that reduced the level and/or
activity of the Sry protein and which comprise at least one additional targeted c
modification are selected.
In one non-limiting example, following genotyping with ic primers and/or
probes, Fl animals that are heterozygous for the targeted genetic modification to the
polynucleotide of interest and lacking the targeted modification that reduces the level and/or
activity of the Sry protein are crossed to one another. Such a cross produces an F2 progeny
that is homozygous for the genetically ed genomic locus of interest and does not
comprise the genetic modification to reduce Sry protein levels and/or activity.
Further provided is a method of producing a transgenic non-human animal
homozygous for a targeted genetic modification in the F1 generation. The method ses
(a) crossing an F0 XY fertile female non-human animal having a targeted genetic
modification that decreases the level and/or activity of the Sry protein with a F0 XY male
non-human animal, wherein the F0 XY fertile female non-human animal and the F0 XY male
non-human animal are each heterozygous for the same genetic modification of a
polynucleotide of interest, and (b) ing an F1 progeny that is homozygous for the
targeted genetic modification in the polynucleotide of interest. In a specific embodiment, the
F1 progeny selected are homozygous for the targeted genetic cation in the
polynucleotide of interest and lack the targeted genetic modification that decreases the
activity and/or level of the Sry protein. Such methods can be employed to develop breeding
pairs of non-human animals, each fully d from a donor ES cell or iPS cell, in the same
F0 generation.
Various methods can be employed to obtain the F0 animals described above. In
one non-limiting embodiment, an XY cell clone with a targeted modification in a
polynucleotide of interest on any chromosome is isolated. It is ized that various
methods can be used to generate the targeted modification in the polynucleotide of st.
In a second step, a targeted modification is introduced into the Sry gene such that the
modification decreases the level and/or activity of the Sry protein. Such methods will further
employ culturing the XY ES cell in a media that es the development of XY F0 fertile
females, as describe in detail elsewhere . Methods of targeted modification of the Sry
gene are disclosed in detail elsewhere herein and can comprise, for example, the use of a
targeting vector (including an LTVEC) either alone or in ation with a nuclease as
described elsewhere herein (i.e., a Talen or CRISPR- or ZFN- system). A subclone is
isolated that comprises both the first ed modification in the polynucleotide of interest
and the second targeted modification of the Sry gene that decreases the level and/or activity
of the Sry protein. Both the original XY clone with the targeted modification in the
polynucleotide of interest and the XY subclone comprising both the targeted modification to
the Sry gene and the polynucleotide of interest are introduced into separate non-human host
embryos, as discussed elsewhere herein. In specific embodiments, the non-human host
embryos comprise a pre-morula embryo (i.e., an 8 cell stage embryo). Each of the non-
human host embryos comprising the modified pluripotent cells is uced into a surrogate
mother for gestation. Each of the surrogate mothers es F0 progeny comprising the
targeted genome modification (i.e., an F0 XY male having the targeted modification in the
polynucleotide of interest and an F0 XY fertile female having the targeted modification in the
polynucleotide of interest and having the genetic modification that decreases the level and/or
activity of the Sry n). In specific embodiments, each of the targeted genomic
modifications is capable of being transmitted through the germline. Each of these F0 animals
are bred to one another, to generate an F1 animal sing a homozygous targeted
cation in the polynucleotide of interest. One-quarter of the F1 generation are expected
to be homozygous for the targeted modification in the polynucleotide of interest. Fl progeny
can be ed to retain the ed modification to the Sry gene or the F1 progeny can be
ed to not retain the targeted modification to the Sry gene.
In another embodiment, the uction of the targeted modification of the Sry
gene employing a targeting vector (and, in specific embodiments, nucleases such as Talen,
Crispr, or an) can occur simultaneously with the vector targeting for the genetic
modification of the polynucleotide of interest. Such methods allow for the generation of an
XY ES cell having both a genetic modification that decreases the level and/or activity of the
Sry protein and further comprises the targeted modification to the polynucleotide of interest.
In one ment, the F1 generation progeny comprises a genome completely
derived from the donor ES cell. In other ments, the frequency of s of F0
generation male and F0 generation female mice that give rise to fully ES cell-derived mice is
100%.
II. Methods and Compositionsfor Modifying a Challenging Target Genomic Locus or a
Target Genomic Locus on the Y Chromosome
Methods and itions are provided that allow for modifying a target genomic
locus on the Y some in a cell. Further provided are s that allow for modifying
a “challenging” genomic locus. The term “challenging locus” includes a somal
region that is difficult to target by conventional gene targeting strategies. Such loci can be
located on the Y chromosome, the X chromosome, or an autosome. In certain embodiments,
challenging loci are located within or in proximity to gene-poor, repeat-rich, and/or largely
heterochromatic chromosomal regions. See, e. g., Bernardini et al., Proc. Natl. Acad. Sci.
USA 111:7600-7605 (2014), herein incorporated by reference in its entirety for all purposes.
In certain embodiments, a challenging locus is located within or in proximity to chromosomal
regions in which accessibility of the chromosomal DNA is limited by chromatin structure. In
certain embodiments, a challenging locus is within or in proximity to chromosomal regions
characterized by a high tage of heterochromatin, such as at least about ~20%, at least
about ~30%, at least about 40%, at least about 50%, at least about 60%, or at least about 70%
heterochromatin. In certain embodiments, a challenging locus is located within or in
ity to chromosomal regions that have undergone duplications and rearrangements or
that are characterized by the presence of repeats or ed repeats. See, e. g., Gubbay et al.,
Proc. Natl. Acad. Sci. USA 3-7957 (1992), herein incorporated by reference in its
entirety for all purposes.
The term “chromatin” includes nucleoprotein complexes which t and
organize cellular genetic material to contain it within cells. The term “heterochromatin”
includes s in the genome that are in a highly condensed state and are generally
transcriptionally silent. Heterochromatin is generally more tightly coiled and generally has
more repetitive DNA sequences than euchromatin. The term “euchromatin” includes regions
in the genome characterized by more extended and less condensed chromatin domains that
are often transcriptionally active and accessible.
The term “exposing” includes using any method by which desired components are
brought into immediate proximity or direct contact.
Methods and compositions are provided that allow for modifying a challenging
target genomic locus or a target genomic locus on the Y chromosome in a cell. Perhaps due
to unique structural es of the Y chromosome, conventional gene targeting strategies in
mouse embryonic stem cells to generate mutations on the Y-linked genes has had limited
success. Therefore, often the understanding of the functions of murine Y-linked genes is
limited to insights gained from studies of mice that carry spontaneous deletions, random gene
trap insertions or autosomal transgenes. Methods provided herein allow for the targeting of a
genomic locus on the Y some by employing a targeting vector in the absence of or in
combination with a nuclease agent.
Some such s utilize a small ing vector or VEC. A
“smallTVEC” includes a targeting vector that comprises short homology arms. The length of
a homology arm on a smallTVEC can be from about 400-1000 bp. A homology arm of the
smallTVEC can be of any length that is sufficient to promote a gous recombination
event with a corresponding target site, including for example, from about 400 bp to about 500
bp, from about 500 bp to about 600 bp, from about 600 bp to about 700 bp, from about 700
bp to about 800 bp, from about 800 bp to about 900 bp, or from about 900 bp to about 1000
bp. A red length of a homology arm on a VEC is from about 700 bp to about 800
bp. In another embodiment, the sum total of 5’ and 3’ homology arms of the smallTVEC is
about 0.5 kb, 1 kb, 1.5 kb, 2 kb, 3 kb, 4 kb, 5kb, 6kb, 7kb, 8kb, 9kb, about 0.5 kb to about 1
kb, about 1 kb to about 1.5 kb, about 1.5 kb to about 2 kb, about 2 kb to about 3 kb, about 3
kb to about 4 kb, about 4 kb to about 5kb, about 5kb to about 6 kb, about 6 kb to about 7 kb,
about 8 kb to about 9 kb, or is at least 10 kb. In such methods, the short length of the
gy arms increases the targeting efficiency as compared to a targeting vector with
longer homology arms. Due to the nature of the Y chromosome which has highly repetitive
sequences, the short arms of the smallTVECs allow for highly specific targeting on the Y
some.
] Methods are provided for modifying a target genomic locus on the Y chromosome
in a cell comprising: (a) providing a cell comprising a target c locus on the Y
chromosome comprising a recognition site for a nuclease agent, (b) introducing into the cell a
first targeting vector comprising a first insert polynucleotide flanked by a first and a second
homology arm corresponding to a first and a second target site; and (c) identifying at least
one cell comprising in its genome the first insert polynucleotide integrated at the target
genomic locus on the Y chromosome. In specific embodiments, the sum total of the first
homology arm and the second homology arm of the targeting vector is about 0.5 kb, 1 kb, 1.5
kb, 2 kb, 3 kb, 4 kb, 5kb, 6kb, 7kb, 8kb, 9kb, about 0.5 kb to about 1 kb, about 1 kb to about
1.5 kb, about 1.5 kb to about 2 kb, about 2 kb to about 3 kb, about 3 kb to about 4 kb, about 4
kb to about 5kb, about 5kb to about 6 kb, about 6 kb to about 7 kb, about 8 kb to about 9 kb,
or is at least 10 kb or at least 10 kb and less than 150 kb. In some embodiments, a
smallTVEC is employed. In specific embodiments, an LTVEC is employed. Similar methods
can be med when targeting a challenging target genomic locus. In one non-limiting
embodiment, such methods are performed employing the culture media that promotes the
development of XY F0 fertile females sed herein and thereby generating XY F0 fertile
female animals. In other instance, the s described herein are ed to produce a
targeted genetic modification in the Sry gene, as discussed elsewhere herein.
Further provided are methods for modifying a target genomic locus on the Y
chromosome in a cell sing: (a) providing a cell comprising a target c locus on
the Y some comprising a recognition site for a nuclease agent, (b) introducing into the
cell (i) the nuclease agent, wherein the se agent s a nick or double-strand break
at the first recognition site; and, (ii) a first targeting vector comprising a first insert
polynucleotide flanked by a first and a second homology arm corresponding to a first and a
second target site located in ient proximity to the first recognition site; and (c)
identifying at least one cell comprising in its genome the first insert polynucleotide integrated
at the target genomic locus on the Y chromosome. In specific embodiments, the sum total of
the first homology arm and the second homology arm of the targeting vector is about 0.5 kb,
1 kb, 1.5 kb, 2 kb, 3 kb, 4 kb, 5kb, 6kb, 7kb, 8kb, 9kb, about 0.5 kb to about 1 kb, about 1 kb
to about 1.5 kb, about 1.5 kb to about 2 kb, about 2 kb to about 3 kb, about 3 kb to about 4
kb, about 4 kb to about 5kb, about 5kb to about 6 kb, about 6 kb to about 7 kb, about 8 kb to
about 9 kb, or is at least 10 kb or at least 10 kb and less than 150 kb. In some embodiments, a
smallTVEC is employed. In specific embodiments, an LTVEC is employed. Similar methods
can be performed when targeting a challenging target genomic locus. In one non-limiting
embodiment, such methods are performed employing the e media that promotes the
development of XY F0 fertile females sed herein and thereby generating XY F0 fertile
female animals. In other instance, the methods described herein are employed to produce a
targeted genetic modification in the Sry gene, as discussed elsewhere herein.
It is recognized that the various methods disclosed herein to te a targeted
cation in a genomic locus of the Y chromosome (or any challenging genomic locus)
2015/038001
employing a targeting vector, a smallTVEC, or an LTVEC can be performed in any cell type,
and is not limited to an XY pluripotent and/or totipotent cell. Such cell types include, but are
not limited to, a human cell, a non-human cell, a mammalian cell, non-human mammalian
cell, a rodent cell, a mouse cell, a rat cell, a hamster cell, a fibroblast cell or any other host
cell. Such cells include pluripotent cells, including, for example, induced pluripotent stem
(iPS) cells, mouse embryonic stem (ES) cells, rat nic stem (ES) cells, human
embryonic (ES) cells, or developmentally restricted human progenitor cells.
Methods are further disclosed to generate a large deletion on the Y chromosome
employing any of the various nuclease agents provided herein (e. g., CRISPR gRNAs in
ation with Cas9; ZFNs; or TALENs). Such a deletion on the Y chromosome can be a
on of an endogenous c acid sequence. The deletion can range from about 5 kb to
about 10 kb, from about 10 kb to about 20 kb, from about 20 kb to about 40 kb, from about
40 kb to about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to about 100 kb,
from about 100 kb to about 150 kb, from about 150 kb to about 200 kb, from about 200 kb to
about 300 kb, from about 300 kb to about 400 kb, from about 400 kb to about 500 kb, from
about 500 kb to about 600 kb, from about 600 kb to about 700 kb, from about 700 kb to about
800 kb, from about 800 kb to about 900 kb, from about 900 kb to about 1 Mb, from about
500 kb to about 1 Mb, from about 1 Mb to about 1.5 Mb, from about 1.5 Mb to about 2 Mb,
from about 2 Mb to about 2.5 Mb, or from about 2.5 Mb to about 3 Mb. In one embodiment,
the deletion is greater than 500 kb. In another embodiment, the deletion is from about 500 kb
to about 600 kb. In a specific embodiment, the deletion is about 500 kb. Such a deletion on
the Y some can be a deletion of any nucleic acid sequence. In one embodiment, the
deletion comprises a gene that is ated with ity/infertility. The deletion on the Y
chromosome can comprise a deletion of multiple genes. In such methods, 1, 2, 3, 4, 5, 6, 7, 8,
9, 10 or more genes can be deleted. In specific embodiments, the Kdm5d gene (Lysine (K)-
specific demethylase 5d; for example, Entrez Gene ID 20592 (mus musculus)) and/or the
Usp9y gene (ubiquitin specific peptidase 9, y-linked; for example, Entrez Gene ID
107868(mus musculus)) is targeted for deletion. In other embodiments, the Sry gene is
ed for deletion.
A. Nuclease Agents and Recognition Sites for Nuclease Agents
The term “recognition site for a nuclease agent” includes a DNA sequence at
which a nick or double-strand break is induced by a nuclease agent. The recognition site for
a nuclease agent can be endogenous (or native) to the cell or the ition site can be
exogenous to the cell. In specific ments, the recognition site is exogenous to the cell
and thereby is not naturally occurring in the genome of the cell. In still further embodiments,
the recognition site is exogenous to the cell and to the polynucleotides of st that one
desires to be positioned at the target locus. In further embodiments, the exogenous or
endogenous ition site is present only once in the genome of the host cell. In ic
embodiments, an endogenous or native site that occurs only once within the genome is
identified. Such a site can then be used to design nuclease agents that will produce a nick or
double-strand break at the endogenous recognition site.
The length of the recognition site can vary, and includes, for example, recognition
sites that are about 30-36 bp for a zinc finger nuclease (ZFN) pair (i.e., about 15-18 bp for
each ZFN), about 36 bp for a Transcription Activator-Like Effector Nuclease ), or
about 20bp for a CRISPR/Cas9 guide RNA.
In one embodiment, each monomer of the nuclease agent recognizes a recognition
site of at least 9 nucleotides. In other embodiments, the recognition site is from about 9 to
about 12 nucleotides in length, from about 12 to about 15 nucleotides in length, from about
to about 18 nucleotides in length, or from about 18 to about 21 nucleotides in , and
any combination of such subranges (e. g., 9-18 nucleotides). It is recognized that a given
nuclease agent can bind the recognition site and cleave that g site or alternatively, the
nuclease agent can bind to a sequence that is different from the recognition site. Moreover,
the term recognition site comprises both the nuclease agent g site and the nick/cleavage
site irrespective whether the nick/cleavage site is within or outside the se agent binding
site. In another variation, the cleavage by the se agent can occur at nucleotide
positions immediately opposite each other to produce a blunt end cut or, in other cases, the
incisions can be red to produce single-stranded overhangs, also called “sticky ends”,
which can be either 5' overhangs, or 3' ngs.
Any nuclease agent that induces a nick or double-strand break into a desired
recognition site can be used in the methods and compositions disclosed herein. A naturallyoccurring
or native nuclease agent can be employed so long as the nuclease agent induces a
nick or double-strand break in a desired recognition site. Alternatively, a modified or
engineered nuclease agent can be employed. An “engineered nuclease agent” includes a
nuclease that is engineered (modified or derived) from its native form to specifically
recognize and induce a nick or double-strand break in the desired recognition site. Thus, an
engineered nuclease agent can be d from a native, naturally-occurring nuclease agent or
it can be artificially created or synthesized. The modification of the nuclease agent can be as
little as one amino acid in a protein cleavage agent or one nucleotide in a nucleic acid
cleavage agent. In some embodiments, the engineered nuclease induces a nick or double-
strand break in a ition site, wherein the recognition site was not a sequence that would
have been recognized by a native (non-engineered or non-modified) nuclease agent.
Producing a nick or double-strand break in a recognition site or other DNA can be referred to
herein as “cutting” or “cleaving” the recognition site or other DNA.
Active variants and fragments of the exemplified recognition sites are also
provided. Such active variants can comprise at least 65%, 70%, 75%, 80%, 85%, 90%, 91%,
92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more ce identity to the given
recognition site, wherein the active variants retain biological activity and hence are capable of
being recognized and cleaved by a nuclease agent in a ce-specific manner. Assays to
measure the double-strand break of a recognition site by a nuclease agent are known in the art
(e. g., TaqMan® qPCR assay, Frendewey D. et (11., Methods in Enzymology, 2010, -
307, which is incorporated by reference herein in its entirety).
In specific ments, the recognition site is positioned within the
polynucleotide encoding the selection marker. Such a position can be located within the
coding region of the selection marker or within the tory regions, which influence the
expression of the selection marker. Thus, a recognition site of the nuclease agent can be
located in an intron of the selection marker, a promoter, an enhancer, a regulatory region, or
any non-protein-coding region of the polynucleotide ng the selection marker. In
specific embodiments, a nick or double-strand break at the recognition site disrupts the
activity of the selection . Methods to assay for the presence or absence of a onal
selection marker are known.
In one embodiment, the se agent is a ription Activator-Like Effector
Nuclease (TALEN). TAL effector nucleases are a class of sequence-specific nucleases that
can be used to make double-strand breaks at specific target sequences in the genome of a
prokaryotic or eukaryotic organism. TAL effector nucleases are created by fusing a native or
engineered transcription activator-like (TAL) effector, or onal part thereof, to the
catalytic domain of an clease, such as, for example, Fold. The unique, modular TAL
effector DNA binding domain allows for the design of proteins with potentially any given
DNA recognition specificity. Thus, the DNA binding domains of the TAL effector nucleases
can be engineered to recognize specific DNA target sites and thus, used to make double-
strand breaks at desired target sequences. See, ; Morbitzer et a1. (2010)
PNAS 10.1073/pnas.1013133107; Scholze & Boch (2010) nce 1:428-432; Christian et
a1. Genetics (2010) 186:757-761; Li et a1. (2010) Nuc. Acids Res. (2010)
2015/038001
doi:10.1093/nar/gkq704; and Miller et a1. (2011) Nature Biotechnology 29:143—148; all of
which are herein incorporated by reference.
es of suitable TAL ses, and methods for ing suitable TAL
nucleases, are disclosed, e. g., in US Patent Application No. 2011/0239315 A1, 2011/0269234
A1, 145940 A1, 2003/0232410 A1, 2005/0208489 A1, 2005/0026157 A1,
2005/0064474 A1, 2006/0188987 A1, and 2006/0063231 A1 (each hereby incorporated by
reference). In various ments, TAL effector ses are engineered that cut in or
near a target nucleic acid sequence in, e.g., a locus of interest or a genomic locus of interest,
wherein the target nucleic acid sequence is at or near a sequence to be modified by a targeting
vector. The TAL nucleases suitable for use with the various methods and compositions
provided herein include those that are specifically designed to bind at or near target nucleic
acid sequences to be modified by targeting vectors as bed herein.
In one embodiment, each monomer of the TALEN comprises 33-35 TAL repeats
that recognize a single base pair via two hypervariable residues. In one embodiment, the
nuclease agent is a chimeric protein comprising a TAL repeat-based DNA binding domain
operably linked to an independent nuclease. In one embodiment, the independent nuclease is
a FokI endonuclease. In one embodiment, the nuclease agent comprises a first TAL-repeat-
based DNA binding domain and a second TAL-repeat-based DNA binding domain, wherein
each of the first and the second TAL-repeat-based DNA binding domain is ly linked to
a FokI nuclease, wherein the first and the second peat-based DNA binding domain
ize two contiguous target DNA sequences in each strand of the target DNA sequence
ted by a spacer sequence of g length (12-20 bp), and wherein the FokI nuclease
subunits dimerize to create an active nuclease that makes a double strand break at a target
sequence.
The nuclease agent employed in the various methods and compositions sed
herein can further comprise a zinc-finger nuclease (ZFN). In one embodiment, each
monomer of the ZFN comprises 3 or more zinc finger-based DNA binding domains, wherein
each zinc -based DNA binding domain binds to a 3bp subsite. In other embodiments,
the ZFN is a chimeric protein comprising a zinc finger-based DNA binding domain operably
linked to an independent nuclease. In one embodiment, the independent endonuclease is a
FokI endonuclease. In one embodiment, the nuclease agent comprises a first ZFN and a
second ZFN, wherein each of the first ZFN and the second ZFN is operably linked to a FokI
nuclease subunit, wherein the first and the second ZFN recognize two contiguous target DNA
sequences in each strand of the target DNA sequence separated by about 5-7 bp spacer, and
n the Fokl nuclease subunits dimerize to create an active nuclease to make a double
strand break. See, for example, US20060246567; US20080182332; US20020081614;
US20030021776; WO/2002/057308A2; US20130123484; US20100291048;
WO/2011/017293A2; and Gaj et al. (2013) Trends in Biotechnology, 31(7):397-405 each of
which is herein incorporated by reference.
In still another embodiment, the nuclease agent is a meganuclease.
Meganucleases have been classified into four families based on ved sequence motifs,
the families are the ADG, GIY-YIG, H-N-H, and His-Cys box es. These
motifs participate in the coordination of metal ions and hydrolysis of phosphodiester bonds.
Meganucleases are notable for their long recognition sites, and for tolerating some sequence
polymorphisms in their DNA substrates. Meganuclease domains, structure and function are
known, see for e, Guhan and Muniyappa (2003) Crit Rev Biochem Mol Biol 38: 199-
248; Lucas et al., (2001) Nucleic Acids Res 29:960-9; Jurica and Stoddard, (1999) Cell Mol
Life Sci 55:1304-26; Stoddard, (2006) Q Rev Biophys 38:49-95; and Moure et al., (2002) Nat
Struct Biol 9:764. In some examples a naturally occurring variant, and/or engineered
derivative meganuclease is used. Methods for modifying the cs, cofactor interactions,
expression, optimal conditions, and/or recognition site icity, and screening for activity
are known, see for example, Epinat et al., (2003) Nucleic Acids Res 31:2952-62; ier et
al., (2002) Mol Cell 10:895-905; Gimble et al., (2003) Mol Biol 334:993-1008; Seligman et
al., (2002) Nucleic Acids Res 30:3870-9; Sussman et al., (2004) JMol Biol 342:31-41; Rosen
et al., (2006) Nucleic Acids Res 34:4791-800; Chames et al., (2005) c Acids Res
33:el78; Smith et al., (2006) Nucleic Acids Res 34:el49; Gruen et al., (2002) Nucleic Acids
Res 30:e29; Chen and Zhao, (2005) Nucleic Acids Res 33:el54; WO2005105989;
WO2003078619; WO2006097854; 097853; WO2006097784; and WO2004031346.
Any clease can be used herein, including, but not limited to, I-Scel, I-
SceII, I-SceIII, I—SceIV, I-SceV, I—SceVI, I-SceVII, I-Ceul, I-CeuAIIP, I—Crel, I-CrepsbIP, I-
CrepsbIIP, I—CrepsbIIIP, I-CrepsbIVP, , I-Ppol, PI-Pspl, F-SceI, I, , F-
TevI, F—TevII, I-Amal, I-Anil, I—Chul, I—Cmoel, I-Cpal, I-CpaII, I-Csml, I-Cvul, I-CvuAIP,
I-Ddil, I-DdiII, I—DirI, I—Dmol, , I—HmuII, I-HsNIP, I-LlaI, , I-NaaI, I—NanI, I-
NcIIP, I-NngP, I-NitI, I-NjaI, I—Nsp236IP, I-Pakl, I-PboIP, I-PcuIP, I-PcuAI, I-PcuVI, 1-
PngP, I-PobIP, I-Porl, I-PorIIP, I-PprP, I-SpBetaIP, I—Scal, I-SexIP, I-SneIP, I-SpomI, I-
, I-SpomIP, I-SpomlIP, I-SquIP, I-Ssp6803l, I-SthPhiJP, hiST3P, I-
SthPhiSTe3bP, I-TdeIP, I-TevI, I-Tevll, I—TevIII, I-UarAP, I—UarHGPAIP, I-UarHGPAl3P,
I-VinIP, I-ZbiIP, PI-Mtul, PI—MtuHIP PI—MtuHIIP, l, PI-PfuII, PI-Pkol, PI—Pkoll, PI-
Rma43812IP, PI—SpBetaIP, I, PI-TfuI, PI-TfuII, PI-ThyI, PI-TliI, PI—TliII, or any
active variants or fragments thereof.
In one embodiment, the meganuclease recognizes double-stranded DNA
sequences of 12 to 40 base pairs. In one embodiment, the clease izes one
perfectly matched target sequence in the genome. In one embodiment, the meganuclease is a
homing se. In one ment, the homing nuclease is a LAGLIDADG family of
homing nuclease. In one embodiment, the LAGLIDADG family of homing nuclease is
selected from I-SceI, I-CreI, and I-Dmol.
Nuclease agents can further comprise restriction endonucleases, which include
Type I, Type II, Type III, and Type IV endonucleases. Type I and Type III restriction
endonucleases recognize specific recognition sites, but typically cleave at a variable position
from the nuclease binding site, which can be hundreds of base pairs away from the cleavage
site (recognition site). In Type II systems the restriction activity is independent of any
methylase activity, and cleavage typically occurs at specific sites within or near to the
binding site. Most Type II enzymes cut palindromic sequences, however Type IIa enzymes
recognize non-palindromic ition sites and cleave outside of the recognition site, Type
IIb enzymes cut sequences twice with both sites outside of the recognition site, and Type IIs
enzymes recognize an asymmetric ition site and cleave on one side and at a d
distance of about 1-20 nucleotides from the recognition site. Type IV restriction enzymes
target methylated DNA. Restriction enzymes are further described and classified, for example
in the REBASE database ge at rebase.neb.com; Roberts et al., (2003) Nucleic Acids
Res 31 :418-20), s et al., (2003) Nucleic Acids Res 31 :1805-12, and Belfort et al.,
(2002) in Mobile DNA 11, pp. 761-783, Eds. Craigie et al., (ASM Press, gton, DC).
The nuclease agent employed in the various methods and compositions can
also comprise a CRISPR/Cas system. Such systems can employ a Cass? nuclease, which in
some instances, is cation—Optimized fer the desired cell type in which it is to he sed.
The system further s a fused chNA~trachNA construct that functions with the
coden—optimized Cas9. This single RNA is Often referred to as a guide RNA or gRNA,
Within a gRNA, the chNA perticn is identified as the ‘target sequence" for the given
recognition site and the trachNA is often referred to as the ‘scaffold’. This system has been
shown to functien in a variety of euliar‘yotic and prokaryetic cells, Briefly, a short DNA
fragment containing the target sequence is inserted into a guide RNA expression plasmid.
The gRNA expression plasmid ccniprises the target sequence (in some einbcdinients around
’20 nuclentides), a foiin of the trachNA ce (the scaffold) as well as a suitable
promoter that is active in the cell and ary elements for proper processing in enharyotic
cells. Many of the systems rely on custom, complementary oligos that are annealed to form a
double stranded DNA and then cloned into the gRNA expression plasmid. The gRNA
sion cassette and the Case”) expression cassette are then introduced into the cell. See,
for example, Mali P et al. (2013) Science 2013 Feb 15; 339 (6121):823-6; Jinek M et al.
e 2012 Aug 17;337(6096):816-21; Hwang WY et al. Nat Biotechnol 2013
Mar;31(3):227-9; Jiang W et al. Nat Biotechnol 2013 Mar;31(3):233-9; and, Cong L et al.
Science 2013 Feb 15;339(6121):819-23, each of which is herein incorporated by reference.
The methods and compositions disclosed herein can utilize Clustered
Regularly Interspersed Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas)
systems or components of such systems to modify a genome within a cell. CRISPR/Cas
systems include transcripts and other elements involved in the sion of, or directing the
activity of, Cas genes. A CRISPR/Cas system can be a type I, a type II, or a type 111 system.
The methods and compositions disclosed herein employ CRISPR/Cas systems by utilizing
CRISPR xes (comprising a guide RNA (gRNA) complexed with a Cas protein) for
site-directed cleavage of nucleic acids.
Some CRISPR/Cas systems used in the methods disclosed herein are non-
naturally occurring. A aturally occurring” system es anything indicating the
involvement of the hand of man, such as one or more ents of the system being altered
or mutated from their lly occurring state, being at least substantially free from at least
one other component with which they are naturally associated in nature, or being associated
with at least one other component with which they are not naturally associated. For example,
some CRISPR/Cas systems employ non-naturally occurring CRISPR complexes comprising
a gRNA and a Cas protein that do not naturally occur together.
(i) A. Cas RNA-Guided cleases
Cas proteins generally comprise at least one RNA recognition or binding
domain. Such domains can interact with guide RNAs (gRNAs, described in more detail
below). Cas proteins can also comprise nuclease domains (e.g., DNase or RNase s),
DNA binding domains, helicase domains, protein-protein interaction domains, dimerization
domains, and other domains. A nuclease domain possesses tic activity for nucleic acid
cleavage. Cleavage es the breakage of the covalent bonds of a nucleic acid le.
Cleavage can e blunt ends or staggered ends, and it can be single-stranded or double-
stranded.
Examples of Cas proteins include Casl, CaslB, Cas2, Cas3, Cas4, Cas5,
Cas5e (CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8al or
, Cas8a2, Cas8b, Cas8c, Cas9 (Csnl
Csx12), Cale, Caled, CasF, CasG, CasH, Csyl, Csy2, Csy3, Csel (CasA), Cse2 (CasB),
Cse3 (CasE), Cse4 , Cscl, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6,
Cmrl , Cmr3, Cmr4, Cmr5, Cmr6, Csbl, Csb2, Csb3, Csxl7, Csxl4, CsxlO, Csxl6, CsaX,
Csx3, Csxl, Csx15, Csfl, Csf2, Csf3, Csf4, and Cul966, and homologs or modified versions
thereof.
Cas proteins can be from a type II CRISPR/Cas system. For example, the Cas
protein can be a Cas9 protein or be derived from a Cas9 protein. Cas9 proteins typically
share four key motifs with a conserved ecture. Motifs l, 2, and 4 are Rqu-like ,
and motif 3 is an HNH motif. The Cas9 protein can be from, for example, Streptococcus
pyogenes, Streptococcus thermophilus, Streptococcus sp., Staphylococcus aureus,
Nocardiopsis dassonvillei, Streptomyces pristinaespiralis, omyces viridochromogenes,
Streptomyces chromogenes, Streptosporangium roseum, Streptosporangium roseum,
AlicyclobacHlus acidocaldarius, us pseudomycoides, Bacillus selenitireducens,
Exiguobacterium cum, Lactobacillus delbrueckii, Lactobacillus salivarius, Microscilla
, Burkholderiales bacterium, Polaromonas naphthalenivorans, monas sp.,
Crocosphaera watsonii, Cyanothece sp., Microcystis aeruginosa, Synechococcus sp.,
Acetohalobium arabaticum, Ammonifex degensii, Caldicelulosiruptor becscii, Candidatus
Desulforudis, Clostridium botulinum, Clostridium diflicile, ldia magna,
Natranaerobius thermophilus, Pelotomaculum thermopropionicum, Acidithiobacillus caldus,
Acidithiobacillusferrooxidans, Allochromatium vinosum, Marinobacter sp., Nitrosococcus
halophilus, Nitrosococcus watsoni, Pseudoalteromonas haloplanktis, Ktedonobacter
racemifer, Methanohalobium gatum, Anabaena variabilis, Nodularia spumigena,
Nostoc sp., Arthrospira maxima, Arthrospira platensis, spira sp., Lyngbya sp.,
Microcoleus chthonoplastes, Oscillatoria sp., Petrotoga s, Thermosipho africanus, or
Acaryochloris marina. Additional examples of the Cas9 family members are described in
WC 2014/131833, herein incorporated by reference in its ty. Cas9 protein from S.
pyogenes or d therefrom is a preferred enzyme. Cas9 protein from S. es is
assigned SwissProt accession number .
Cas proteins can be wild type proteins (i.e., those that occur in nature),
modified Cas proteins (i.e., Cas protein variants), or fragments of wild type or modified Cas
proteins. Cas proteins can also be active variants or fragments of wild type or modified Cas
proteins. Active variants or fragments can comprise at least 80%, 85%, 90%, 91%, 92%,
93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to the wild type or
modified Cas protein or a portion thereof, wherein the active variants retain the y to cut
at a desired cleavage site and hence retain nick-inducing or -strand-break-inducing
activity. Assays for nick-inducing or double-strand-break-inducing activity are known and
generally measure the overall ty and specificity of the Cas protein on DNA substrates
containing the cleavage site.
Cas proteins can be modified to increase or decrease nucleic acid binding
affinity, nucleic acid binding specificity, and/or enzymatic activity. Cas proteins can also be
modified to change any other activity or property of the protein, such as stability. For
example, one or more nuclease domains of the Cas protein can be modified, deleted, or
vated, or a Cas protein can be truncated to remove domains that are not essential for the
function of the protein or to optimize (e. g., enhance or reduce) the activity of the Cas protein.
Some Cas proteins se at least two nuclease domains, such as DNase
domains. For example, a Cas9 n can comprise a Rqu-like se domain and an
HNH-like nuclease domain. The Rqu and HNH domains can each cut a different strand of
double-stranded DNA to make a double-stranded break in the DNA. See, e. g., Jinek et al.
(2012) Science 337:816-821, hereby incorporated by reference in its entirety.
One or both of the nuclease s can be deleted or d so that they are
no longer functional or have reduced nuclease activity. If one of the se domains is
deleted or mutated, the resulting Cas protein (e. g., Cas9) can be referred to as a nickase and
can generate a single-strand break at a CRISPR RNA recognition sequence within a double-
ed DNA but not a double-strand break (i.e., it can cleave the complementary strand or
the non-complementary strand, but not both). If both of the se domains are deleted or
mutated, the resulting Cas n (e. g., Cas9) will have a reduced ability to cleave both
s of a double-stranded DNA. An example of a mutation that converts Cas9 into a
nickase is a DlOA (aspartate to alanine at position 10 of Cas9) mutation in the Rqu domain
of Cas9 from S. pyogenes. Likewise, H939A (histidine to alanine at amino acid position 839)
or H840A (histidine to alanine at amino acid position 840) in the HNH domain of Cas9 from
S. pyogenes can convert the Cas9 into a nickase. Other examples of mutations that convert
Cas9 into a nickase include the corresponding mutations to Cas9 from S. thermophilus. See,
e. g., Sapranauskas et a1. (2011) Nucleic Acids Research 39:9275-9282 and WC 2013/141680,
each of which is herein orated by reference in its entirety. Such ons can be
generated using methods such as irected mutagenesis, PCR-mediated mutagenesis, or
total gene synthesis. Examples of other mutations creating nickases can be found, for
example, in WO/2013/176772Al and WO/2013/142578Al, each of which is herein
incorporated by reference.
Cas proteins can also be fusion proteins. For example, a Cas protein can be
fused to a cleavage domain, an epigenetic modification domain, a transcriptional activation
domain, or a riptional repressor . See WO 89290, incorporated herein by
reference in its entirety. Cas proteins can also be fused to a heterologous polypeptide
providing sed or decreased stability. The fused domain or heterologous polypeptide
can be located at the N-terminus, the C-terminus, or internally within the Cas protein.
A Cas protein can be fused to a heterologous ptide that provides for
subcellular localization. Such heterologous peptides e, for example, a nuclear
localization signal (NLS) such as the SV40 NLS for targeting to the s, a mitochondrial
localization signal for targeting to the mitochondria, an ER retention signal, and the like.
See, e.g., Lange et a1. (2007) J. Biol. Chem. 282:5101-5105. Such lular localization
s can be located at the inus, the C-terminus, or anywhere within the Cas protein.
An NLS can comprise a stretch of basic amino acids, and can be a monopartite sequence or a
bipartite sequence.
Cas proteins can also be linked to a cell-penetrating domain. For e, the
cell-penetrating domain can be derived from the HIV-1 TAT protein, the TLM cell-
penetrating motif from human hepatitis B virus, MPG, Pep-l, VP22, a cell penetrating
peptide from Herpes simplex virus, or a polyarginine peptide sequence. See, for example,
, herein incorporated by reference in its entirety. The cell-penetrating
domain can be located at the N-terminus, the C-terminus, or anywhere within the Cas protein.
Cas proteins can also se a heterologous polypeptide for ease of tracking
or purification, such as a fluorescent protein, a purification tag, or an epitope tag. es
of fluorescent proteins include green fluorescent proteins (e. g., GFP, GFP-2, tagGFP,
turboGFP, eGFP, Emerald, Azami Green, Monomeric Azami Green, CopGFP, AceGFP,
ZsGreenl), yellow fluorescent proteins (e. g., YFP, eYFP, Citrine, Venus, YPet, PhiYFP,
ZsYellowl), blue cent proteins (e.g. eBFP, eBFPZ, Azurite, mKalamal, GFPuv,
Sapphire, T-sapphire), cyan fluorescent proteins (e.g. eCFP, Cerulean, CyPet, AmCyanl,
Midoriishi-Cyan), red fluorescent proteins (mKate, mKate2, mPlum, DsRed monomer,
mCherry, mRFPl, DsRed-Express, DsRed2, DsRed-Monomer, HcRed-Tandem, HcRedl,
AsRed2, eqFP6ll, mRaspberry, mStrawberry, Jred), orange fluorescent proteins (mOrange,
mKO, Kusabira-Orange, ric Kusabira-Orange, mTangerine, o), and any
other suitable fluorescent protein. Examples of tags include glutathione-S-transferase (GST),
chitin binding protein (CBP), e binding protein, thioredoxin (TRX), p01y(NANP),
tandem affinity purification (TAP) tag, myc, ACV5, AU1
, AU5, E, ECS, E2, FLAG,
hemagglutinin (HA), nus, Softag 1, Softag 3, Strep, SBP, Glu-Glu, HSV, KT3, S, S1 , T7,
V5, VSV-G, histidine (His), biotin carboxyl carrier protein (BCCP), and calmodulin.
Cas proteins can be provided in any form. For example, a Cas n can be
provided in the form of a protein, such as a Cas protein complexed with a gRNA.
Alternatively, a Cas protein can be provided in the form of a nucleic acid encoding the Cas
protein, such as an RNA (e. g., messenger RNA (mRNA)) or DNA. Optionally, the nucleic
acid encoding the Cas protein can be codon optimized for efficient translation into protein in
a ular cell or organism.
Nucleic acids encoding Cas proteins can be stably integrated in the genome of
the cell and operably linked to a promoter active in the cell. Alternatively, c acids
encoding Cas ns can be operably linked to a promoter in an expression construct.
Expression constructs e any nucleic acid constructs e of directing expression of a
gene or other c acid sequence of interest (e.g., a Cas gene) and which can transfer such
a nucleic acid sequence of interest to a target cell. Promoters that can be used in an
expression construct include, for example, promoters active in a pluripotent rat, eukaryotic,
mammalian, non-human mammalian, human, rodent, mouse, or hamster cell. Examples of
other promoters are described elsewhere herein.
(ii) B. Guide RNAs (gRNAs)
A “guide RNA” or “gRNA” includes an RNA molecule that binds to a Cas
protein and targets the Cas protein to a specific location within a target DNA. Guide RNAs
can comprise two segments: a “DNA-targeting segment” and a “protein-binding segment.”
“Segment” includes a segment, section, or region of a molecule, such as a contiguous stretch
of nucleotides in an RNA. Some gRNAs comprise two separate RNA molecules: an
“activator-RNA” and a “targeter-RNA.” Other gRNAs are a single RNA molecule (single
RNA polynucleotide), which can also be called a “single-molecule gRNA,” a “single-guide
RNA,” or an “ngNA.” See, e.g., WO/2013/176772A1, WO/2014/065596A1,
WO/2014/089290A1, WO/2014/093622A2, WO/2014/099750A2, WO/2013142578A1, and
WC 31833A1, each of which is herein orated by reference. The terms “guide
RNA” and “gRNA” include both double-molecule gRNAs and single-molecule gRNAs.
An exemplary lecule gRNA comprises a chNA-like (“CRISPR RNA”
or ter-RNA” or “chNA” or “chNA ”) molecule and a corresponding A-
like (“trans-acting CRISPR RNA” or “activator-RNA” or “trachNA” or “scaffold”)
le. A chNA comprises both the DNA-targeting segment (single-stranded) of the
gRNA and a stretch of nucleotides that forms one half of the dsRNA duplex of the protein-
binding segment of the gRNA.
A corresponding trachNA (activator-RNA) comprises a stretch of nucleotides
that forms the other half of the dsRNA duplex of the protein-binding segment of the gRNA.
A stretch of nucleotides of a chNA are mentary to and hybridize with a stretch of
tides of a trachNA to form the dsRNA duplex of the protein-binding domain of the
gRNA. As such, each chNA can be said to have a corresponding trachNA.
The chNA and the corresponding trachNA hybridize to form a gRNA. The
chNA additionally es the single-stranded rgeting segment that hybridizes to a
CRISPR RNA recognition sequence. If used for modification within a cell, the exact
sequence of a given chNA or trachNA molecule can be designed to be ic to the
species in which the RNA molecules will be used. See, for example, Mali et al. (2013)
Science 339:823-826; Jinek et al. (2012) Science 337:816-821; Hwang et al. (2013) Nat.
Biotechnol. 31:227-229; Jiang et al. (2013) Nat. Biotechnol. 31 :233-239; and Cong et al.
(2013) Science 339:819-823, each of which is herein incorporated by reference.
The DNA-targeting segment (chNA) of a given gRNA comprises a
nucleotide sequence that is complementary to a sequence in a target DNA. The DNA-
targeting segment of a gRNA cts with a target DNA in a sequence-specific manner via
hybridization (i.e., base pairing). As such, the nucleotide sequence of the DNA-targeting
segment may vary and determines the location within the target DNA with which the gRNA
and the target DNA will interact. The DNA-targeting segment of a t gRNA can be
modified to hybridize to any desired sequence within a target DNA. Naturally occurring
chNAs differ depending on the Cas9 system and sm but often contain a targeting
segment of n 21 to 72 nucleotides length, flanked by two direct repeats (DR) of a
length of between 21 to 46 nucleotides (see, e. g., WO2014/131833). In the case of S.
pyogenes, the DRs are 36 nucleotides long and the targeting segment is 30 tides long.
The 3’ located DR is complementary to and izes with the corresponding trachNA,
which in turn binds to the Cas9 protein.
The DNA-targeting segment can have a length of from about 12 nucleotides to
about 100 nucleotides. For example, the DNA-targeting segment can have a length of from
about 12 nucleotides (nt) to about 80 nt, from about 12 nt to about 50 nt, from about 12 nt to
about 40 nt, from about 12 nt to about 30 nt, from about 12 nt to about 25 nt, from about 12
nt to about 20 nt, or from about 12 nt to about 19 nt. Alternatively, the DNA-targeting
WO 00805
segment can have a length of from about 19 nt to about 20 nt, from about 19 nt to about 25 nt,
from about 19 nt to about 30 nt, from about 19 nt to about 35 nt, from about 19 nt to about 40
nt, from about 19 nt to about 45 nt, from about 19 nt to about 50 nt, from about 19 nt to about
60 nt, from about 19 nt to about 70 nt, from about 19 nt to about 80 nt, from about 19 nt to
about 90 nt, from about 19 nt to about 100 nt, from about 20 nt to about 25 nt, from about 20
nt to about 30 nt, from about 20 nt to about 35 nt, from about 20 nt to about 40 nt, from about
nt to about 45 nt, from about 20 nt to about 50 nt, from about 20 nt to about 60 nt, from
about 20 nt to about 70 nt, from about 20 nt to about 80 nt, from about 20 nt to about 90 nt, or
from about 20 nt to about 100 nt.
The nucleotide sequence of the DNA-targeting segment that is complementary
to a nucleotide sequence R RNA recognition sequence) of the target DNA can have a
length at least about 12 nt. For example, the rgeting sequence (i.e., the sequence
within the DNA-targeting segment that is complementary to a CRISPR RNA ition
sequence within the target DNA) can have a length at least about 12 nt, at least about 15 nt, at
least about 18 nt, at least about 19 nt, at least about 20 nt, at least about 25 nt, at least about
nt, at least about 35 nt, or at least about 40 nt. Alternatively, the DNA-targeting sequence
can have a length of from about 12 nucleotides (nt) to about 80 nt, from about 12 nt to about
50 nt, from about 12 nt to about 45 nt, from about 12 nt to about 40 nt, from about 12 nt to
about 35 nt, from about 12 nt to about 30 nt, from about 12 nt to about 25 nt, from about 12
nt to about 20 nt, from about 12 nt to about 19 nt, from about 19 nt to about 20 nt, from about
19 nt to about 25 nt, from about 19 nt to about 30 nt, from about 19 nt to about 35 nt, from
about 19 nt to about 40 nt, from about 19 nt to about 45 nt, from about 19 nt to about 50 nt,
from about 19 nt to about 60 nt, from about 20 nt to about 25 nt, from about 20 nt to about 30
nt, from about 20 nt to about 35 nt, from about 20 nt to about 40 nt, from about 20 nt to about
45 nt, from about 20 nt to about 50 nt, or from about 20 nt to about 60 nt. In some cases, the
DNA-targeting sequence can have a length of at about 20 nt.
TrachNAs can be in any form (e.g., full-length trachNAs or active partial
trachNAs) and of varying lengths. They can include primary transcripts or processed forms.
For example, trachNAs (as part of a single-guide RNA or as a separate molecule as part of a
lecule gRNA) may comprise or consist of all or a portion of a wild-type trachNA
sequence (e.g., about or more than about 20, 26, 32, 45, 48, 54, 63, 67, 85, or more
nucleotides of a wild-type trachNA sequence). Examples of wild-type trachNA sequences
from S. pyogenes e 171-nucleotide, 89-nucleotide, 75-nucleotide, and 65-nucleotide
versions. See, for example, Deltcheva et a1. (2011) Nature 471 :602-607; ,
each of which is incorporated herein by nce in their entirety. es of trachNAs
within -guide RNAs (ngNAs) e the A segments found within +48, +54,
+67, and +85 versions of ngNAs, where “+n” indicates that up to the +n nucleotide of wild-
type trachNA is included in the ngNA. See US 8,697,359, incorporated herein by
reference in its entirety.
The percent complementarity between the DNA-targeting sequence and the
CRISPR RNA recognition sequence within the target DNA can be at least 60% (e.g., at least
65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least
97%, at least 98%, at least 99%, or 100%). The t complementarity between the DNA-
targeting ce and the CRISPR RNA recognition ce within the target DNA can be
at least 60% over about 20 contiguous tides. As an example, the percent
mentarity between the DNA-targeting sequence and the CRISPR RNA recognition
sequence within the target DNA is 100% over the 14 contiguous nucleotides at the 5’ end of
the CRISPR RNA recognition sequence within the mentary strand of the target DNA
and as low as 0% over the remainder. In such a case, the DNA-targeting sequence can be
considered to be 14 nucleotides in length. As another example, the percent complementarity
between the DNA-targeting sequence and the CRISPR RNA recognition sequence within the
target DNA is 100% over the seven contiguous nucleotides at the 5’ end of the CRISPR RNA
recognition sequence within the complementary strand of the target DNA and as low as 0%
over the remainder. In such a case, the DNA-targeting sequence can be considered to be 7
nucleotides in length.
The protein-binding segment of a gRNA can comprise two stretches of
nucleotides that are complementary to one another. The complementary nucleotides of the
protein-binding t hybridize to form a double-stranded RNA duplex (dsRNA). The
protein-binding segment of a subject gRNA interacts with a Cas protein, and the gRNA
directs the bound Cas protein to a ic nucleotide sequence within target DNA via the
DNA-targeting segment.
Guide RNAs can include modifications or sequences that provide for
additional desirable features (e. g., modified or regulated stability; subcellular targeting;
tracking with a cent label; a binding site for a protein or protein complex; and the
like). Examples of such modifications include, for example, a 5' cap (e. g., a 7-
methylguanylate cap (m7G)); a 3' polyadenylated tail (i.e., a 3' poly(A) tail); a riboswitch
sequence (e. g., to allow for regulated stability and/or regulated accessibility by proteins
and/or protein complexes); a stability control sequence; a sequence that forms a dsRNA
duplex (i.e., a hairpin)); a modification or sequence that targets the RNA to a subcellular
location (e. g., nucleus, mitochondria, chloroplasts, and the like); a modification or sequence
that provides for ng (e. g., direct conjugation to a fluorescent molecule, conjugation to a
moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection,
and so forth); a modification or sequence that provides a binding site for proteins (e. g.,
proteins that act on DNA, including transcriptional activators, transcriptional repressors,
DNA transferases, DNA demethylases, histone acetyltransferases, histone
deacetylases, and the like); and combinations thereof.
Guide RNAs can be provided in any form. For example, the gRNA can be
ed in the form of RNA, either as two molecules (separate chNA and trachNA) or as
one molecule (ngNA), and optionally in the form of a complex with a Cas protein. The
gRNA can also be provided in the form of DNA encoding the RNA. The DNA encoding the
gRNA can encode a single RNA molecule (ngNA) or separate RNA molecules (e.g.,
separate chNA and trachNA). In the latter case, the DNA encoding the gRNA can be
provided as separate DNA les encoding the chNA and trachNA, respectively.
DNAs encoding gRNAs can be stably integrated in the genome of the cell and
operably linked to a promoter active in the cell. Alternatively, DNAs encoding gRNAs can be
operably linked to a promoter in an expression construct. Such ers can be active, for
example, in a pluripotent rat, eukaryotic, mammalian, non-human mammalian, human,
rodent, mouse, or hamster cell. In some instances, the promoter is an RNA polymerase III
promoter, such as a human U6 er, a rat U6 polymerase III er, or a mouse U6
polymerase III promoter. Examples of other promoters are described elsewhere .
Alternatively, gRNAs can be prepared by various other methods. For
e, gRNAs can be prepared by in vitro transcription using, for example, T7 RNA
polymerase (see, for example, and ). Guide RNAs can
also be a synthetically produced molecule prepared by chemical synthesis.
(iii) C. CRISPR RNA ition Sequences
The term "CRISPR RNA recognition sequence" includes nucleic acid
sequences t in a target DNA to which a rgeting segment of a gRNA will bind,
provided sufficient conditions for binding exist. For example, CRISPR RNA ition
ces include sequences to which a guide RNA is designed to have complementarity,
where hybridization between a CRISPR RNA recognition ce and a DNA targeting
sequence promotes the formation of a CRISPR complex. Full complementarity is not
necessarily required, provided there is ient complementarity to cause hybridization and
promote formation of a CRISPR complex. CRISPR RNA recognition sequences also include
cleavage sites for Cas proteins, described in more detail below. A CRISPR RNA recognition
sequence can comprise any polynucleotide, which can be d, for example, in the nucleus
or cytoplasm of a cell or within an organelle of a cell, such as a mitochondrion or chloroplast.
The CRISPR RNA ition sequence within a target DNA can be targeted
by (i.e., be bound by, or hybridize with, or be complementary to) a Cas protein or a gRNA.
le DNA/RNA g conditions include physiological conditions normally present in
a cell. Other suitable DNA/RNA binding conditions (e.g., conditions in a cell-free system)
are known in the art (see, e. g., Molecular Cloning: A tory Manual, 3rd Ed. (Sambrook
et al., Harbor Laboratory Press 2001)). The strand of the target DNA that is complementary
to and hybridizes with the Cas protein or gRNA can be called the "complementary strand,"
and the strand of the target DNA that is complementary to the "complementary strand" (and
is therefore not complementary to the Cas protein or gRNA) can be called
"noncomplementary strand" or ate strand.”
The Cas n can cleave the nucleic acid at a site within or outside of the
nucleic acid ce present in the target DNA to which the rgeting segment of a
gRNA will bind. The “cleavage site” includes the position of a nucleic acid at which a Cas
protein produces a single-strand break or a -strand break. For e, formation of a
CRISPR x (comprising a gRNA hybridized to a CRISPR RNA recognition sequence
and xed with a Cas protein) can result in cleavage of one or both strands in or near
(e.g., within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, or more base pairs from) the nucleic acid
sequence present in a target DNA to which a DNA-targeting segment of a gRNA will bind.
If the cleavage site is outside of the nucleic acid sequence to which the DNA-targeting
segment of the gRNA will bind, the ge site is still considered to be within the “CRISPR
RNA recognition sequence.” The cleavage site can be on only one strand or on both strands
of a nucleic acid. Cleavage sites can be at the same position on both strands of the nucleic
acid (producing blunt ends) or can be at ent sites on each strand (producing staggered
ends). Staggered ends can be produced, for example, by using two Cas proteins, each of
which produces a single-strand break at a different cleavage site on each strand, thereby
producing a double-strand break. For example, a first nickase can create a single-strand
break on the first strand of double-stranded DNA (dsDNA), and a second nickase can create a
single-strand break on the second strand of dsDNA such that overhanging sequences are
created. In some cases, the CRISPR RNA recognition sequence of the nickase on the first
strand is separated from the CRISPR RNA recognition sequence of the nickase on the second
strand by at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 40, 50, 75, 100, 250, 500, or 1,000
base pairs.
Site-specific cleavage of target DNA by Cas9 can occur at locations
determined by both (i) base-pairing complementarity between the gRNA and the target DNA
and (ii) a short motif, called the pacer adjacent motif (PAM), in the target DNA. The
PAM can flank the CRISPR RNA recognition sequence. Optionally, the CRISPR RNA
ition sequence can be flanked by the PAM. For example, the cleavage site of Cas9 can
be about 1 to about 10 or about 2 to about 5 base pairs (e. g., 3 base pairs) upstream or
downstream of the PAM sequence. In some cases (e. g., when Cas9 from S. pyogenes or a
y related Cas9 is used), the PAM sequence of the non-complementary strand can be 5'-
N1GG-3', where N1is any DNA nucleotide and is immediately 3' of the CRISPR RNA
recognition sequence of the non-complementary strand of the target DNA. As such, the PAM
sequence of the complementary strand would be 5'-CC N2-3', where N2 is any DNA
nucleotide and is immediately 5' of the CRISPR RNA recognition sequence of the
complementary strand of the target DNA. In some such cases, N1 and N2 can be
complementary and the N1- N2 base pair can be any base pair (e. g., N1=C and N2=G; N1=G
and N2=C; N1=A and N2=T, N1=T, and N2=A).
Examples of CRISPR RNA recognition sequences include a DNA sequence
mentary to the DNA-targeting segment of a gRNA, or such a DNA sequence in
addition to a PAM sequence. For example, the target motif can be a 20-nucleotide DNA
sequence immediately preceding an NGG motif recognized by a Cas protein (see, for
example, WC 2014/165825). The guanine at the 5’ end can facilitate transcription by RNA
rase in cells. Other examples of CRISPR RNA recognition sequences can include two
guanine tides at the 5’ end (e. g., GGNZONGG; SEQ ID NO: 9) to facilitate efficient
ription by T7 polymerase in vitro. See, for e, WO 65596.
The CRISPR RNA recognition sequence can be any nucleic acid ce
endogenous or exogenous to a cell. The CRISPR RNA ition sequence can be a
sequence coding a gene product (e. g., a protein) or a non-coding sequence (e. g., a regulatory
sequence) or can include both.
In one embodiment, the target sequence is immediately flanked by a Protospacer
Adjacent Motif (PAM) sequence. In one embodiment, the locus of interest comprises the
nucleotide sequence of SEQ ID NO: 1. In one embodiment, the gRNA comprises a third
nucleic acid sequence encoding a Clustered rly Interspaced Short Palindromic Repeats
(CRISPR) RNA (chNA) and a trans-activating CRISPR RNA (trachNA). In another
WO 00805
embodiment, the genome of the pluripotent rat cell comprises a target DNA region
complementary to the target sequence. In some such methods, the Cas protein is Cas9. In
some embodiments, the gRNA ses (a) the chimeric RNA of the nucleic acid sequence
of SEQ ID NO: 2; or (b) the ic RNA of the c acid sequence of SEQ ID NO: 3.
In some such methods, the chNA comprises the sequence set forth in SEQ ID NO: 4, SEQ
ID NO: 5, or SEQ ID NO: 6. In some such methods, the trachNA comprises the sequence
set forth in SEQ ID NO: 7 or SEQ ID NO: 8.
Active variants and fragments of nuclease agents (i.e. an ered nuclease
agent) are also provided. Such active variants can comprise at least 65%, 70%, 75%, 80%,
85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to
the native nuclease agent, wherein the active variants retain the ability to cut at a d
recognition site and hence retain nick or double-strand-break-inducing activity. For example,
any of the nuclease agents described herein can be modified from a native endonuclease
sequence and designed to recognize and induce a nick or -strand break at a recognition
site that was not recognized by the native nuclease agent. Thus, in some embodiments, the
engineered nuclease has a specificity to induce a nick or double-strand break at a recognition
site that is different from the corresponding native nuclease agent recognition site. Assays for
nick or double-strand-break-inducing activity are known and generally measure the overall
activity and specificity of the endonuclease on DNA substrates containing the recognition
site.
The nuclease agent may be introduced into the cell by any means known in the art.
The polypeptide encoding the nuclease agent may be directly introduced into the cell.
Alternatively, a polynucleotide encoding the nuclease agent can be introduced into the cell.
When a cleotide encoding the nuclease agent is introduced into the cell, the nuclease
agent can be transiently, conditionally or constitutive expressed within the cell. Thus, the
cleotide encoding the nuclease agent can be contained in an expression cassette and be
operably linked to a conditional promoter, an inducible promoter, a constitutive promoter, or
a tissue-specific promoter. Such promoters of interest are discussed in further detail
elsewhere . Alternatively, the nuclease agent is introduced into the cell as an mRNA
encoding a nuclease agent.
In specific ments, the cleotide encoding the nuclease agent is stably
integrated in the genome of the cell and ly linked to a promoter active in the cell. In
other embodiments, the polynucleotide encoding the nuclease agent is in the same targeting
vector comprising the insert cleotide, while in other instances the polynucleotide
encoding the nuclease agent is in a vector or a plasmid that is separate from the targeting
vector comprising the insert polynucleotide.
When the nuclease agent is ed to the cell through the introduction of a
polynucleotide encoding the nuclease agent, such a polynucleotide encoding a nuclease agent
can be modified to substitute codons having a higher frequency of usage in the cell of
st, as compared to the naturally ing polynucleotide ce encoding the
nuclease agent. For example the polynucleotide encoding the nuclease agent can be modified
to substitute codons having a higher frequency of usage in a given prokaryotic or eukaryotic
cell of interest, including a bacterial cell, a yeast cell, a human cell, a non-human cell, a
mammalian cell, a rodent cell, a mouse cell, a rat cell or any other host cell of interest, as
compared to the naturally occurring polynucleotide sequence.
B. Employing the CRISPR/Cas System in Combination with a Large Targeting
Vector (LTVEC) or a Small Targeting Vector (SmallTVEC) to Modify a Challenging
Genomic Loci or a Y Chromosome Locus
] Non-limiting methods for modifying a nging genomic locus or a locus of the
Y chromosome comprise exposing the chromosome (i.e., the Y chromosome) to a Cas protein
and a CRISPR RNA in the ce of a large targeting vector (LTVEC) comprising a
c acid sequence of at least 10 kb, wherein following exposure to the Cas protein, the
CRISPR RNA, and the LTVEC, the chromosome (i.e., the Y chromosome) is modified to
contain at least 10 kb nucleic acid sequence.
The method can employ any of the LTVECs or VECs described herein. In
non-limiting embodiments, the LTVEC or smallTVEC comprises a nucleic acid sequence of
at least 20 kb, at least 30 kb, at least 40 kb, at least 50 kb, at least 60 kb, at least 70 kb, at
least 80 kb, at least 90 kb, at least 100 kb, at least 150 kb, or at least 200 kb. In other
embodiments, the sum total of 5’ and 3’ homology arms of the LTVEC is from about 10 kb
to about 150 kb, about 10 kb to about 20 kb, from about 20 kb to about 40 kb, from about 40
kb to about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to about 100 kb, from
about 100 kb to about 120 kb, or from about 120 kb to 150 kb. In another embodiment, the
sum total of 5’ and 3’ homology arms of the smallTVEC is about 0.5 kb, 1 kb, 1.5 kb, 2 kb, 3
kb, 4 kb, 5kb, 6kb, 7kb, 8kb, 9kb, about 0.5 kb to about 1 kb, about 1 kb to about 1.5 kb,
about 1.5 kb to about 2 kb, about 2 kb to about 3 kb, about 3 kb to about 4 kb, about 4 kb to
about 5kb, about 5kb to about 6 kb, about 6 kb to about 7 kb, about 8 kb to about 9 kb, or is at
least 10 kb.
Further provided is a method for modifying a challenging target locus or a target
genomic locus on the Y chromosome, sing: (a) providing a mammalian cell
comprising the challenging target locus or a target genomic locus on the Y some,
wherein the target genomic locus comprises a guide RNA (gRNA) target sequence; (b)
introducing into the mammalian cell: (i) a large targeting vector (LTVEC) comprising a first
nucleic acid flanked with ing arms homologous to the target genomic locus, wherein the
LTVEC is at least 10 kb; (ii) a first expression construct comprising a first promoter operably
linked to a second nucleic acid encoding a Cas protein, and (iii) a second sion construct
sing a second promoter operably linked to a third nucleic acid encoding a guide RNA
(gRNA) comprising a nucleotide sequence that hybridizes to the gRNA target ce and a
trans-activating CRISPR RNA (trachNA), wherein the first and the second promoters are
active in the mammalian cell; and, (c) identifying a modified mammalian cell comprising a
targeted genetic cation at the challenging target genomic locus or at the target genomic
locus on the Y some. In specific embodiments, the first and the second expression
constructs are on a single nucleic acid molecule. In other embodiments, the target genomic
locus of the Y chromosomes is the Sry locus.
As outlined above, in one embodiment, the Cas protein can comprise a Cas9
protein. In another embodiment, the gRNA target sequence is immediately flanked by a
Protospacer Adjacent Motif (PAM) sequence.
The method can employ any of the LTVECs or smallTVECs described herein. In
miting embodiments, the LTVEC or smallTVEC is at least 0.5 kb, at least 1 kb, at least
kb, at least 10 kb, at least 15 kb, at least 20 kb, at least 30kb, at least 40 kb, at least 50 kb, at
least 60 kb, at least 70 kb, at least 80 kb, at least 90 kb, at least 100 kb, at least 150 kb, or at
least 200 kb. In other embodiments, the sum total of 5’ and 3’ homology arms of the LTVEC
is from about 10 kb to about 150 kb, about 10 kb to about 20 kb, from about 20 kb to about
40 kb, from about 40 kb to about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to
about 100 kb, from about 100 kb to about 120 kb, or from about 120 kb to 150 kb.
The various methods employing the CRISPR/Cas system (or any method
disclosed herein) can be performed on, for example, mammalian cells, non-human
mammalian cells, fibroblast cells, rodent cells, rat cells, mouse cells, or hamster cells. The
cell can be a pluripotent cell, an d pluripotent stem (iPS) cell, a mouse embryonic stem
(ES) cell, a rat embryonic stem (ES) cell, a human embryonic stem (ES) cell or a
developmentally restricted human progenitor cell.
As discussed in detail below, following the modification a challenging genomic
locus or a genomic locus of interest on the Y chromosome (i.e., the Sry locus) of a non-
human pluripotent cell ing, for example, using the CRISPR/CAS system outline
above, the genetically modified non-human otent cell that is produced can be
introduced into a non-human host embryo; and the non-human host embryo comprising the
modified pluripotent cell in a surrogate mother is gestated. The surrogate mother es
F0 progeny comprising the targeted genetic modification. In specific embodiments, the
targeted genetic modification is capable of being itted through the germline.
C. Selection Markers
Various selection markers can be used in the methods and itions disclosed
herein which provide for modifying a target genomic locus on the Y some or a
challenging target genomic locus. Such markers are disclosed elsewhere herein and include,
but are not limited to, selection markers that impart resistance to an antibiotic such as G418,
hygromycin, blastocidin, neomycin, or puromycin. The polynucleotide encoding the
selection markers are operably linked to a promoter active in the cell. Such expression
cassettes and their various regulatory components are discussed in further detailed elsewhere
herein.
D. Target c Locus
Various methods and compositions are provided which allow for the integration of
at least one insert polynucleotide at a target genomic locus on the Y chromosome or a
challenging target genomic locus. As used herein, a “target c locus on the Y
some” comprises any t or region of DNA on the Y chromosome that one
desires to integrate an insert polynucleotide.
The genomic locus on the Y chromosome or a challenging target genomic locus
being targeted can be native to the cell, or alternatively can comprise a heterologous or
exogenous t of DNA that was integrated into the chromosome of the cell. Such
heterologous or ous ts of DNA can include transgenes, expression cassettes,
polynucleotide encoding ion makers, or heterologous or exogenous regions of genomic
DNA. The target genomic locus on the Y chromosome or the challenging target genomic
locus can comprise any of the targeted genomic ation system including, for example,
the recognition site, the selection marker, previously integrated insert polynucleotides,
polynucleotides encoding nuclease agents, ers, etc. Alternatively, the target genomic
locus on the Y chromosome or the challenging target genomic locus can be located within a
yeast artificial chromosome (YAC), bacterial artificial chromosome (BAC), a human
artificial chromosome, or any other engineered genomic region contained in an appropriate
host cell. Thus, in specific embodiments, the targeted genomic locus on the Y chromosome
or the challenging target genomic locus can comprise native, heterologous or exogenous
genomic nucleic acid sequence from a non-human mammal, a non-human cell, a rodent, a
human, a rat, a mouse, a hamster, a rabbit, a pig, a , a deer, a sheep, a goat, a chicken, a
cat, a dog, a ferret, a primate (e. g., marmoset, rhesus monkey), domesticated mammal or an
agricultural mammal or any other organism of interest or a combination thereof.
Non-limiting examples of the target genomic locus on the Y chromosome include,
the Sry gene, the Uty gene, the Eif2s3y gene, the Ddx3y gene, the gene, the Ubely gene, the
Tspy gene, the Usp9y gene, the ny1 gene, and the ny2 gene and the region on the Y
chromosome encompassing the Kdm5d, Eif2s3y, Tspy, Uty, Ddx3y, and Usp9y genes. Such
a locus on the Y chromosome can be from a non-human mammal, a mammal, a rodent, a
human, a rat, a mouse, a hamster, a , a pig, a , a deer, a sheep, a goat, a chicken, a
cat, a dog, a ferret, a primate (e. g., marmoset, rhesus monkey), domesticated mammal or an
ltural mammal or any other organism of interest or a combination thereof. Such cells
include pluripotent cells, including, for example, induced pluripotent stem (iPS) cells, mouse
embryonic stem (ES) cells, rat embryonic stem (ES) cells, human embryonic stem (ES) cell,
or developmentally restricted human progenitor cells.
As described elsewhere , s methods and compositions are provided
which comprise XY pluripotent and/or totipotent cells (such as XY ES cells or iPS cells)
having a decreased activity or level of the Sry protein. The various methods described herein
to modify genomic locus on the Y chromosome can also be used to uce ed genetic
modifications to polynucleotides of interest that are not located on the Y chromosome.
E. Targeting Vectors and Insert cleotides
As ed above, methods and itions provided herein employ ing
vectors alone or in combination with a nuclease agent. “Homologous recombination” is used
conventionally to refer to the exchange of DNA fragments between two DNA molecules at
over sites within the regions of homology.
1'. Insert Polynucleotide
As used herein, the “insert polynucleotide” comprises a t of DNA that one
desires to integrate at the target genomic locus. In specific embodiments, the target genomic
locus is on the Y chromosome. In other embodiments, the target genomic locus is a
challenging genomic locus. In one embodiment, the insert polynucleotide comprises one or
more polynucleotides of interest. In other embodiments, the insert polynucleotide can
comprise one or more expression cassettes. A given expression te can comprise a
polynucleotide of st, a polynucleotide encoding a selection marker and/or a reporter
gene along with the various regulatory ents that influence expression. miting
examples of polynucleotides of interest, selection markers, and reporter genes that can be
included within the insert polynucleotide are discussed in detail elsewhere herein.
In specific embodiments, the insert polynucleotide can comprise a genomic
nucleic acid. In one embodiment, the genomic c acid is d from an animal, a
mouse, a human, a non-human, a rodent, a man, a rat, a hamster, a rabbit, a pig, a
bovine, a deer, a sheep, a goat, a chicken, a cat, a dog, a ferret, a primate (e.g., marmoset,
rhesus monkey), icated mammal or an agricultural mammal, an avian, or any other
organism of interest or a combination thereof.
In further embodiments, the insert polynucleotide comprises a conditional allele.
In one embodiment, the conditional allele is a multifunctional allele, as described in US
2011/0104799, which is incorporated by reference in its entirety. In specific embodiments,
the conditional allele comprises: (a) an ing sequence in sense orientation with respect to
transcription of a target gene, and a drug ion cassette in sense or antisense orientation;
(b) in antisense orientation a nucleotide sequence of interest (NSI) and a conditional by
inversion module (COIN, which utilizes an exon-splitting intron and an invertible genetrap-
like module; see, for example, US 2011/0104799, which is incorporated by nce in its
entirety); and (c) recombinable units that recombine upon re to a first recombinase to
form a conditional allele that (i) lacks the actuating sequence and the DSC, and (ii) contains
the NSI in sense orientation and the COIN in antisense orientation.
The insert polynucleotide can be from about 5kb to about 200kb, from about 5kb
to about 10kb, from about 10kb to about 20kb, from about 20kb to about 30kb, from about
30kb to about 40kb, from about 40kb to about 50kb, from about 60kb to about 70kb, from
about 80kb to about 90kb, from about 90kb to about 100kb, from about 100kb to about
110kb, from about 120kb to about 130kb, from about 130kb to about 140kb, from about
140kb to about 150kb, from about 150kb to about 160kb, from about 160kb to about 170kb,
from about 170kb to about 180kb, from about 180kb to about 190kb, from about 190kb to
about 200kb, from about 200kb to about 250kb, from about 250kb to about 300kb, from
about 300kb to about 350kb, or from about 350kb to about 400kb.
In ic embodiments, the insert polynucleotide comprises a nucleic acid
flanked with pecific recombination target sequences. It is recognized that while the
entire insert polynucleotide can be flanked by such site-specific recombination target
sequence, any region or individual polynucleotide of interest within the insert polynucleotide
can also be flanked by such sites. The term "recombination site" as used herein includes a
nucleotide sequence that is ized by a site-specific recombinase and that can serve as a
substrate for a recombination event. The term "site-specific inase" as used herein
includes a group of enzymes that can facilitate recombination between recombination sites
where the two recombination sites are physically separated within a single nucleic acid
molecule or on separate nucleic acid molecules. es of site-specific inases
include, but are not limited to, Cre, Flp, and Dre inases. The site-specific
recombinase can be introduced into the cell by any means, including by introducing the
recombinase polypeptide into the cell or by ucing a polynucleotide encoding the site-
specific recombinase into the host cell. The polynucleotide encoding the site-specific
recombinase can be located within the insert polynucleotide or within a separate
polynucleotide. The site-specific recombinase can be operably linked to a er active in
the cell including, for example, an inducible promoter, a promoter that is nous to the
cell, a promoter that is heterologous to the cell, a cell-specific promoter, a tissue-specific
promoter, or a developmental stage-specific promoter. Site-specific recombination target
sequences which can flank the insert polynucleotide or any polynucleotide of interest in the
insert polynucleotide can include, but are not limited to, loxP, lox511, lox2272, lox66, lox71,
loxM2, lox5171, FRT, FRTl 1, FRT71, attp, att, FRT, rox, and a combination thereof.
In other embodiments, the site-specific recombination sites flank a polynucleotide
encoding a selection marker and/or a reporter gene contained within the insert
polynucleotide. In such instances following integration of the insert cleotide at the
ed genomic locus the sequences between the site-specific recombination sites can be
removed.
In one embodiment, the insert polynucleotide comprises a polynucleotide
encoding a selection marker. Such selection markers include, but are not limited, to neomycin
phosphotransferase (neor), hygromycin B otransferase (hygr), puromycin-N-
acetyltransferase (puror), cidin S deaminase (bsrr), ne/guanine phosphoribosyl
erase (gpt), or herpes x virus thymidine kinase (HSV-k), or a combination
thereof. In one embodiment, the polynucleotide encoding the selection marker is operably
linked to a promoter active in the cell. When serially tiling polynucleotides of interest into a
targeted genomic locus, the selection marker can comprise a ition site for a nuclease
agent, as outlined above. In one embodiment, the polynucleotide encoding the selection
marker is flanked with a pecific recombination target sequences.
The insert polynucleotide can further comprise a reporter gene operably linked to
a promoter, wherein the reporter gene encodes a reporter n selected from the group
consisting of LacZ, mPlum, mCherry, thomato, mStrawberry, J-Red, DsRed, mOrange,
mKO, ne, Venus, YPet, enhanced yellow fluorescent protein (EYFP), Emerald,
enhanced green cent protein (EGFP), CyPet, cyan fluorescent n (CFP), Cerulean,
T-Sapphire, luciferase, alkaline phosphatase, and a combination thereof. Such reporter genes
can be operably linked to a promoter active in the cell. Such promoters can be an inducible
promoter, a promoter that is nous to the reporter gene or the cell, a er that is
heterologous to the reporter gene or to the cell, a cell-specific promoter, a tissue-specific
promoter manner or a developmental stage-specific promoter.
ii. Targeting Vectors
Targeting vectors are employed to introduce the insert polynucleotide into the
targeted genomic locus on the Y chromosome or into a challenging target locus or on another
chromosome of interest. The targeting vector comprises the insert polynucleotide and r
comprises an upstream and a ream gy arm that flank the insert polynucleotide.
The homology arms that flank the insert polynucleotide correspond to genomic regions
within the targeted genomic locus. For ease of reference, the ponding genomic regions
within the targeted c locus are referred to herein as “target sites”. Thus, in one
example, a targeting vector can comprise a first insert polynucleotide flanked by a first and a
second homology arm corresponding to a first and a second target site located in sufficient
proximity to the first recognition site within the polynucleotide encoding the selection
marker. As such, the targeting vector thereby aids in the integration of the insert
polynucleotide into the targeted genomic locus through a homologous recombination event
that occurs between the homology arms and the corresponding target sites within the genome
of the cell.
A gy arm of the targeting vector can be of any length that is sufficient to
promote a homologous recombination event with a corresponding target site, including for
2015/038001
example, from about 400 bp to about 500 bp, from about 500 bp to about 600 bp, from about
600 bp to about 700 bp, from about 700 bp to about 800 bp, from about 800 bp to about 900
bp, or from about 900 bp to about 1000 bp; or at least 5-10, 5-15, 5-20, 5-25, 5-30, 5-35, 5-
40, 5-45, 5- 50, 5-55, 5-60, 5-65, 5- 70, 5-75, 5-80, 5-85, 5-90, 5-95, 5-100, 100-200, or 200-
300 kilobases in length or greater. In specific embodiments, the sum total of the targeting
arms is at least 0.5 kb, 1 kb, 1.5 kb, 2 kb, 3 kb, 4kb, 5kb, 6kb, 7kb, 8kb, 9kb or at least 10kb.
In other ments, the sum total of the homology arms is between about 0.5 kb to about 1
kb, about 1 kb to about 1.5 kb, about 1.5 kb to about 2 kb, about 2 kb to about 3 kb, about 3
kb to about 4 kb, about 4kb to about 5kb, about 5kb to about 6kb, about 6kb to about 7kb,
about 7kb to about 8kb, about 8kb to about 9kb, or about 10kb to about 150kb. As outlined
in further detail below, large targeting vectors can employ targeting arms of greater length.
The target sites within the targeted genomic locus that correspond to the am
and downstream homology arms of the targeting vector are located in “sufficient proximity to
the recognition site” located in the polynucleotide encoding the selection marker. As used
herein, the upstream and downstream homology arms of a targeting vector are “located in
ient proximity” to a recognition site when the distance is such as to promote the
occurrence of a homologous recombination event between the target sites and the homology
arms upon a nick or double-strand break at the recognition site. Thus, in specific
embodiments, the target sites corresponding to the upstream and/or downstream homology
arm of the targeting vector are within at least 10 nucleotide to about 14 kb of a given
recognition site. In specific embodiments, the recognition site is immediately adjacent to at
least one or both of the target sites.
The spatial relationship of the target sites that correspond to the homology arms of
the targeting vector to the recognition site within the cleotide encoding the selection
marker can vary. For example, both target sites can be d 5’ to the recognition site, both
target sites can be d 3’ to the recognition site, or the target sites can flank the
recognition site.
In specific embodiments, the target genomic locus comprises (i) a 5’ target
sequence that is homologous to a 5’ gy arm; and (ii) a 3’ target sequence that is
homologous to a 3’ homology arm. In specific embodiments, the 5’ target ce and the
3’ target sequence is separated by at least 5 kb but less than 3 Mb, at least 5 kb but less than
kb, at least 10 kb but less than 20 kb, at least 20 kb but less than 40 kb, at least 40 kb but
less than 60 kb, at least 60 kb but less than 80 kb, at least about 80 kb but less than 100 kb, at
least 100 kb but less than 150 kb, or at least 150 kb but less than 200 kb, at least about 200 kb
2015/038001
but less than about 300 kb, at least about 300 kb but less than about 400 kb, at least about 400
kb but less than about 500 kb, at least about 500 kb but less than about 1Mb, at least about 1
Mb but less than about 1.5 Mb, at least about 1.5 Mb but less than about 2 Mb, at least about
2 Mb but less than about 2.5 Mb, or at least about 2.5 Mb but less than about 3 Mb.
As used herein, a homology arm and a target site “correspond” or are
“corresponding” to one another when the two regions share a sufficient level of sequence
identity to one another to act as substrates for a homologous recombination reaction. By
“homology” is meant DNA sequences that are either identical or share sequence identity to a
corresponding sequence. The ce identity between a given target site and the
corresponding homology arm found on the targeting vector can be any degree of ce
identity that allows for homologous recombination to occur. For example, the amount of
sequence ty shared by the homology arm of the targeting vector (or a fragment thereof)
and the target site (or a fragment thereof) can be at least 50%, 55%, 60%, 65%, 70%, 75%,
80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%,
96%, 97%, 98%, 99% or 100% sequence identity, such that the ces undergo
homologous recombination. Moreover, a corresponding region of homology between the
homology arm and the corresponding target site can be of any length that is sufficient to
promote homologous recombination at the cleaved recognition site. For e, a given
homology arm and/or corresponding target site can comprise corresponding regions of
homology that are from about 400 bp to about 500 bp, from about 500 bp to about 600 bp,
from about 600 bp to about 700 bp, from about 700 bp to about 800 bp, from about 800 bp to
about 900 bp, or from about 900 bp to about 1000 bp (such as described for the smallTVEC
vectors described elsewhere herein); or at least about 5-10, 5-15, 5-20, 5-25, 5-30, 5-35, 5-40,
-45, 5- 50, 5-55, 5-60, 5-65, 5- 70, 5-75, 5-80, 5-85, 5-90, 5-95, 5-100, 100-200, or 200-300
kilobases in length or more (such as described in the LTVEC vectors described elsewhere
) such that the homology arm has sufficient homology to undergo homologous
recombination with the corresponding target sites within the genome of the cell.
For ease of reference the homology arms are referred to herein an upstream and a
downstream homology arm. This terminology relates to the relative on of the homology
arms to the insert polynucleotide within the targeting vector.
The homology arms of the targeting vector are therefore designed to pond to
a target site with the targeted genomic locus on the Y chromosome or within a nging
target locus. Thus, the homology arms can correspond to a c locus that is native to the
cell, or alternatively they can correspond to a region of a heterologous or exogenous segment
of DNA that was integrated into the Y chromosome, including, but not limited to, transgenes,
expression cassettes, or heterologous or ous regions of genomic DNA. Alternatively,
the homology arms of the targeting vector can correspond to a region of a yeast artificial
chromosome (YAC), a bacterial artificial chromosome (BAC), a human artificial
chromosome, or any other engineered c region ned in an appropriate host cell.
Still further the homology arms of the targeting vector can correspond to or be derived from a
region of a BAC y, a cosmid y, or a P1 phage library. Thus, in specific
embodiments, the gy arms of the targeting vector pond to a genomic locus on
the Y chromosome or to a challenging target locus that is native, heterologous or exogenous
to a non-human mammal, a rodent, a human, a rat, a mouse, a hamster a rabbit, a pig, a
bovine, a deer, a sheep, a goat, a chicken, a cat, a dog, a ferret, a e (e.g., marmoset,
rhesus monkey), domesticated mammal or an agricultural mammal, an avian, or any other
sm of interest. In further embodiments, the homology arms correspond to a genomic
locus of the cell that is not targetable using a conventional method or can be targeted only
ectly or only with significantly low efficiency, in the absence of a nick or double-strand
break induced by a nuclease agent. In one embodiment, the homology arms are derived from
a synthetic DNA.
In still other embodiments, the upstream and downstream gy arms
correspond to the same genome as the targeted genome. In one embodiment, the homology
arms are from a related , e. g., the targeted genome is a mouse genome of a first strain,
and the targeting arms are from a mouse genome of a second strain, wherein the first strain
and the second strain are different. In other embodiments, the homology arms are from the
genome of the same animal or are from the genome of the same strain, e.g., the targeted
genome is a mouse genome of a first strain, and the targeting arms are from a mouse genome
from the same mouse or from the same strain.
The targeting vector (such as a large targeting vector) can also comprise a
selection cassette or a reporter gene as discussed elsewhere herein. The ion cassette can
se a nucleic acid sequence encoding a selection , wherein the nucleic acid
sequence is operably linked to a promoter. Such promoters can be an inducible promoter, a
promoter that is endogenous to the report gene or the cell, a promoter that is heterologous to
the reporter gene or to the cell, a cell-specific promoter, a tissue-specific promoter manner or
a developmental stage-specific promoter. In one embodiment, the selection marker is
selected from neomycin phosphotransferase (neor), hygromycin B phosphotransferase (hygr),
puromycin-N-acetyltransferase (puror), blasticidin S deaminase (bsrr), xanthine/guanine
phosphoribosyl transferase (gpt), and herpes simplex virus thymidine kinase (HSV-k), and a
combination thereof. The ion marker of the targeting vector can be flanked by the
upstream and downstream homology arms or found either 5’ or 3’ to the homology arms.
In one embodiment, the targeting vector (such as a large targeting vector)
comprises a reporter gene operably linked to a promoter, wherein the reporter gene encodes a
reporter protein ed from the group consisting of LacZ, mPlum, mCherry, thomato,
mStrawberry, J-Red, DsRed, e, mKO, mCitrine, Venus, YPet, enhanced yellow
fluorescent protein , Emerald, enhanced green fluorescent protein (EGFP), CyPet,
cyan fluorescent n (CFP), Cerulean, T-Sapphire, luciferase, alkaline phosphatase, and a
combination thereof. Such reporter genes can be ly linked to a er active in the
cell. Such promoters can be an inducible promoter, a promoter that is nous to the
report gene or the cell, a promoter that is logous to the reporter gene or to the cell, a
cell-specific promoter, a tissue-specific promoter manner or a pmental stage-specific
promoter.
In one non-limiting ment, the combined use of the targeting vector
(including, for example, a large targeting vector) with the nuclease agent results in an
increased targeting efficiency compared to the use of the targeting vector alone. In one
embodiment, when the targeting vector is used in conjunction with the nuclease agent,
targeting efficiency of the targeting vector is increased at least by two-fold, at least three-fold,
at least 4-fold, or at least 10-fold when compared to when the ing vector is used alone.
iii. Large Targeting Vectors
The term “large targeting vector” or “LTVEC” as used herein includes large
targeting vectors that comprise homology arms that correspond to and are derived from
nucleic acid sequences larger than those typically used by other approaches intended to
perform homologous targeting in cells and/or comprising insert polynucleotides comprising
nucleic acid sequences larger than those typically used by other approaches intended to
perform homologous recombination ing in cells. In specific embodiments, the
homology arms and/or the insert polynucleotide of the LTVEC comprises a genomic
sequence of a otic cell. The size of the LTVEC is too large to enable screening of
targeting events by conventional assays, e. g., southern blotting and long-range (e. g., lkb-5kb)
PCR. Examples of the LTVEC, include, but are not limited to, vectors derived from a
bacterial artificial chromosome (BAC), a human artificial chromosome or a yeast artificial
chromosome (YAC). Non-limiting examples of LTVECs and methods for making them are
described, e.g., in US Pat. No. 6,586,251, 6,596,541, 7,105,348, and
(PCT/USOl/45375), each of which is herein incorporated by reference.
The LTVEC can be of any length, including, but not limited to, at least about
10kb, about 15kb, about 20kb, about 30kb, about 40kb, about 50kb, about 60kb, about 70kb,
about 80kb, about 90kb, about 100kb, about 150kb, about 200kb, from about 10kb to about
15kb, about 15 kb to about 20kb, about 20kb to about 30kb, from about 30kb to about 50kb,
from about 50kb to about 300kb, from about 50kb to about 75kb, from about 75kb to about
100kb, from about 100kb to 125kb, from about 125kb to about 150kb, from about 150kb to
about 175kb, about 175kb to about 200kb, from about 200kb to about 225kb, from about
225kb to about 250kb, from about 250kb to about 275kb or from about 275kb to about
300kb.
In one embodiment, the LTVEC comprises an insert polynucleotide g from
about 5kb to about 200kb, from about 5kb to about 10kb, from about 10kb to about 20kb,
from about 20kb to about 30kb, from about 30kb to about 40kb, from about 40kb to about
50kb, from about 60kb to about 70kb, from about 80kb to about 90kb, from about 90kb to
about 100kb, from about 100kb to about 110kb, from about 120kb to about 130kb, from
about 130kb to about 140kb, from about 140kb to about 150kb, from about 150kb to about
160kb, from about 160kb to about 170kb, from about 170kb to about 180kb, from about
180kb to about 190kb, or from about 190kb to about 200kb, from about 200kb to about
250kb, from about 250kb to about 300kb, from about 300kb to about 350kb, or from about
350kb to about 400kb.
In other instances, the LTVEC design can be such as to allow for the replacement
of a given sequence that is from about 5kb to about 200kb or from about 5kb to about 3.0Mb
as described herein. In one ment, the replacement is from about 5kb to about 10kb,
from about 10kb to about 20kb, from about 20kb to about 30kb, from about 30kb to about
40kb, from about 40kb to about 50kb, from about 50kb to about 60kb, from about 60kb to
about 70kb, from about 80kb to about 90kb, from about 90kb to about 100kb, from about
100kb to about 110kb, from about 110kb to about 120kb, from about 120kb to about 130kb,
from about 130kb to about 140kb, from about 140kb to about 150kb, from about 150kb to
about 160kb, from about 160kb to about 170kb, from about 170kb to about 180kb, from
about 180kb to about 190kb, from about 190kb to about 200kb, from about 5kb to about
10kb, from about 10kb to about 20kb, from about 20kb to about 40kb, from about 40kb to
about 60kb, from about 60kb to about 80kb, from about 80kb to about 100kb, from about
100kb to about 150kb, or from about 150kb to about 200kb, from about 200kb to about
2015/038001
300kb, from about 300kb to about 400kb, from about 400kb to about 500kb, from about
500kb to about 1Mb, from about 1Mb to about 1.5Mb, from about 1.5Mb to about 2Mb, from
about 2Mb to about 2.5Mb, or from about 2.5Mb to about 3Mb.
In one embodiment, the homology arms of the LTVEC are derived from a BAC
y, a cosmid library, or a P1 phage library. In other embodiments, the gy arms
are derived from the targeted genomic locus of the cell and in some instances the target
genomic locus that the LTVEC is designed to target is not targetable using a conventional
method. In still other embodiments, the homology arms are derived from a synthetic DNA.
In one embodiment, a sum total of the upstream homology arm and the
downstream homology arm in the LTVEC is at least 10kb. In other embodiments, the
upstream homology arm ranges from about 5kb to about 100kb. In one embodiment, the
downstream homology arm ranges from about 5kb to about 100kb. In other embodiments,
the sum total of the upstream and downstream homology arms are from about 5kb to about
10kb, from about 10kb to about 20kb, from about 20kb to about 30kb, from about 30kb to
about 40kb, from about 40kb to about 50kb, from about 50kb to about 60kb, from about 60kb
to about 70kb, from about 70kb to about 80kb, from about 80kb to about 90kb, from about
90kb to about 100kb, from about 100kb to about 110kb, from about 110kb to about 120kb,
from about 120kb to about 130kb, from about 130kb to about 140kb, from about 140kb to
about 150kb, from about 150kb to about 160kb, from about 160kb to about 170kb, from
about 170kb to about 180kb, from about 180kb to about 190kb, or from about 190kb to about
200kb. In one embodiment, the size of the deletion is the same or similar to the size of the
sum total of the 5' and 3' homology arms of the LTVEC.
In one embodiment, the LTVEC comprises a selection cassette or a er gene
as discussed elsewhere herein.
iv. Methods ofIntegrating an Insert Polynucleotide Near the Recognition Site
on the Y Chromosome by Homologous ination
Methods are provided for modifying a target genomic locus on the Y chromosome
in a cell comprising: (a) providing a cell comprising a target c locus on the Y
chromosome, (b) introducing into the cell a first targeting vector comprising a first insert
polynucleotide flanked by a first and a second homology arm corresponding to a first and a
second target site; and (c) identifying at least one cell comprising in its genome the first insert
polynucleotide integrated at the target genomic locus on the Y chromosome. Similar
s can be performed to target a challenging chromosomal locus. As discussed in detail
elsewhere herein, in specific ments, the sum total of the first homology arm and the
second homology arm of the targeting vector is about 0.5 kb, 1 kb, 1.5 kb, 2 kb, 3 kb, 4 kb,
5kb, 6kb, 7kb, 8kb, 9kb, about 0.5 kb to about 1 kb, about 1 kb to about 1.5 kb, about 1.5 kb
to about 2 kb, about 2 kb to about 3 kb, about 3 kb to about 4 kb, about 4 kb to about 5kb,
about 5kb to about 6 kb, about 6 kb to about 7 kb, about 8 kb to about 9 kb, or is at least 10
kb or at least 10 kb and less than 150 kb. In specific embodiments, an LTVEC is employed.
In other specific embodiments, a smallTVEC is employed. In one non-limiting embodiment,
such methods are performed employing the culture media that promotes the development of
XY F0 fertile females disclosed herein and thereby generating XY F0 fertile female s.
In other ce, the methods bed herein are employed to produce a ed genetic
modification in the Sry gene, as discussed elsewhere herein.
Further provided are methods for modifying a target genomic locus on the Y
chromosome in a cell comprising: (a) providing a cell sing a target genomic locus on
the Y chromosome comprising a recognition site for a nuclease agent, (b) ucing into the
cell (i) the nuclease agent, wherein the nuclease agent induces a nick or double-strand break
at the first recognition site; and, (ii) a first targeting vector comprising a first insert
polynucleotide flanked by a first and a second homology arm corresponding to a first and a
second target site located in sufficient proximity to the first recognition site; and (c)
identifying at least one cell comprising in its genome the first insert polynucleotide integrated
at the target genomic locus on the Y chromosome. Similar s can be performed to
target a challenging target locus. As discussed in detail elsewhere herein, in specific
ments, the sum total of the first homology arm and the second homology arm of the
targeting vector is about 0.5 kb, 1 kb, 1.5 kb, 2 kb, 3 kb, 4 kb, 5kb, 6kb, 7kb, 8kb, 9kb, about
0.5 kb to about 1 kb, about 1 kb to about 1.5 kb, about 1.5 kb to about 2 kb, about 2 kb to
about 3 kb, about 3 kb to about 4 kb, about 4 kb to about 5kb, about 5kb to about 6 kb, about
6 kb to about 7 kb, about 8 kb to about 9 kb, or is at least 10 kb or at least 10 kb and less than
150 kb. In specific embodiments, an LTVEC is employed. In other specific embodiments, a
smallTVEC is ed. In one non-limiting embodiment, such s are med
ing the culture media that promotes the development of XY F0 fertile females
disclosed herein and thereby generating XY F0 fertile female animals. In other instance, the
methods described herein are employed to produce a targeted genetic modification in the Sry
gene, as discussed elsewhere herein.
Various methods can also be employed to identify cells having the insert
polynucleotide integrated at the genomic target locus. Insertion of the insert polynucleotide
at the genomic target locus results in a “modification of ”. The term "modification of
allele” or “MOA” includes the modification of the exact DNA sequence of one allele of a
gene(s) or chromosomal locus (loci) in a genome. Examples of “modification of allele
(MOA)” include, but are not limited to, ons, substitutions, or insertions of as little as a
single nucleotide or deletions of many kilobases spanning a gene(s) or chromosomal locus
(loci) of interest, as well as any and all possible modifications n these two extremes.
In various embodiments, to facilitate identification of the targeted modification, a
high-throughput quantitative assay, namely, modification of allele (MOA) assay, is
employed. The MOA assay described herein allows a large-scale screening of a modified
allele(s) in a parental chromosome following a genetic cation. The MOA assay can be
carried out via various analytical techniques, including, but not limited to, a quantitative
PCR, e.g., a real-time PCR (qPCR). For example, the real-time PCR comprises a first
primer-probe set that recognizes the target locus and a second primer-probe set that
recognizes a rgeted reference locus. In addition, the primer-probe set comprises a
fluorescent probe that recognizes the amplified sequence. The quantitative assay can also be
carried out via a variety of ical techniques, including, but not limited to, fluorescence-
ed in situ hybridization (FISH), comparative genomic hybridization, isothermic DNA
amplification, quantitative hybridization to an immobilized probe(s), Invader Probes®, MMP
assays®, TaqMan® Molecular Beacon, and EclipseTM probe technology. (See, for example,
U82005/0144655, incorporated by reference herein in its entirety).
In various embodiments, in the presence of the nick or double strand bread,
targeting ency of a targeting vector (such as a LTVEC or a smallTVEC) at the target
genomic locus is at least about 2-fold higher, at least about 3-fold higher, at least about 4-fold
higher than in the absence of the nick or double-strand break (using, e. g., the same targeting
vector and the same homology arms and corresponding target sites at the c locus of
interest but in the absence of an added nuclease agent that makes the nick or double strand
break).
The various methods set forth above can be tially repeated to allow for the
ed integration of any number of insert polynucleotides into a given targeted genomic
locus on the Y chromosome or into a challenging target locus. Thus, the s methods
provide for the insertion of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,11, 12,13, 14,15, 16,17, 18,
19, 20 or more insert polynucleotides into the target genomic locus on the Y chromosome or
into a nging target locus. In particular embodiments, such sequential tiling methods
allow for the reconstruction of large genomic regions from an animal cell or from a
WO 00805
mammalian cell (i.e., a human, a man, a rodent, a mouse, a monkey, a rat, a hamster, a
icated mammal or an agricultural ) into a targeted genomic locus on a Y
chromosome. In such instances, the transfer and reconstruction of genomic regions that
include both coding and non-coding regions allow for the complexity of a given region to be
preserved by retaining, at least in part, the coding regions, the non-coding s and the
copy number variations found within the native c region. Thus, the s methods
provide, for example, methods to generate “heterologous” or “exogenous” genomic regions
within any mammalian cell or animal of interest. In one non-limiting e, a
“humanized” genomic region within a non-human animal is generated.
It is further recognized that along with ing the target genomic locus on the
Y chromosome, the s methods and compositions disclosed herein can be employed to
also generate at targeted genetic modification on another chromosome.
v. Polynucleotides ofInterest
Any polynucleotide of interest may be contained in the various insert
polynucleotides and thereby integrated at the target genomic locus on the Y chromosome or
into a challenging target locus. The methods disclosed herein, e for at least 1, 2, 3, 4,
, 6 or more polynucleotides of interest to be integrated into the targeted genomic locus.
The polynucleotide of interest within the insert polynucleotide when integrated at
the target genomic locus on the Y chromosome or at a challenging target locus can introduce
one or more genetic modifications into the cell. The genetic modification can comprise a
deletion of an endogenous nucleic acid sequence and/or the addition of an exogenous or
heterologous or orthologous polynucleotide into the target genomic locus. In one
embodiment, the genetic modification comprises a replacement of an endogenous nucleic
acid sequence with an exogenous polynucleotide of interest at the target genomic locus.
Thus, s provided herein allow for the generation of a genetic modification comprising
a knockout, a deletion, an insertion, a replacement (“knock-in”), a point mutation, a domain
swap, an exon swap, an intron swap, a regulatory sequence swap, a gene swap, or a
combination f in a target genomic locus on the Y chromosome. Such modifications
may occur upon integration of the first, second, third, fourth, fifth, six, seventh, or any
subsequent insert polynucleotides into the target genomic locus.
The polynucleotide of st within the insert polynucleotide and/or integrated at
the target genomic locus can comprise a sequence that is native or homologous to the cell it is
introduced into; the polynucleotide of interest can be heterologous to the cell it is introduced
to; the polynucleotide of interest can be exogenous to the cell it is introduced into; the
polynucleotide of interest can be orthologous to the cell it is introduced into; or the
polynucleotide of interest can be from a different species than the cell it is introduced into.
As used herein “homologous” in reference to a sequence is a sequence that is native to the
cell. As used herein, “heterologous” in reference to a sequence is a sequence that ates
from a foreign s, or, if from the same species, is substantially modified from its native
form in ition and/or genomic locus by deliberate human intervention. As used herein,
“exogenous” in nce to a sequence is a sequence that originates from a n species.
As used herein, “orthologous” is a polynucleotide from one species that is functionally
equivalent to a known reference sequence in another species (i.e., a s variant). The
polynucleotide of interest can be from any organism of interest including, but not limited to,
non-human, a rodent, a r, a mouse, a rat, a human, a monkey, an avian, an agricultural
mammal or a non-agricultural mammal. The cleotide of interest can r comprise
a coding region, a non-coding , a tory region, or a genomic DNA. Thus, the 1“,
2nd, 3rd, 4th, 5th, 6th, 7m, and/or any of the subsequent insert polynucleotides can se such
sequences.
In one embodiment, the polynucleotide of interest within the insert polynucleotide
and/or integrated at the target genomic locus on the Y chromosome is homologous to a
mouse nucleic acid sequence, a human nucleic acid, a non-human nucleic acid, a rodent
nucleic acid, a rat nucleic acid, a hamster nucleic acid, a monkey nucleic acid, an agricultural
mammal nucleic acid, or a non-agricultural mammal nucleic acid. In still further
embodiments, the polynucleotide of interest integrated at the target locus is a fragment of a
genomic nucleic acid. In one embodiment, the genomic nucleic acid is a mouse genomic
nucleic acid, a human genomic nucleic acid, a non-human nucleic acid, a rodent nucleic acid,
a rat nucleic acid, a hamster nucleic acid, a monkey c acid, an agricultural mammal
nucleic acid or a non-agricultural mammal nucleic acid or a combination f.
In one embodiment, the polynucleotide of interest can range from about 500
nucleotides to about 200kb as described above. The polynucleotide of interest can be from
about 500 nucleotides to about 5kb, from about 5kb to about 200kb, from about 5kb to about
10kb, from about 10kb to about 20kb, from about 20kb to about 30kb, from about 30kb to
about 40kb, from about 40kb to about 50kb, from about 60kb to about 70kb, from about 80kb
to about 90kb, from about 90kb to about 100kb, from about 100kb to about 110kb, from
about 120kb to about 130kb, from about 130kb to about 140kb, from about 140kb to about
150kb, from about 150kb to about 160kb, from about 160kb to about 170kb, from about
l70kb to about 180kb, from about 180kb to about 190kb, or from about l90kb to about
200kb.
] The polynucleotide of interest within the insert polynucleotide and/or inserted at
the target genomic locus on the Y chromosome or into a challenging target locus can encode
a polypeptide, can encode an miRNA, can encode a long non-coding RNA, or it can comprise
any regulatory regions or non-coding regions of interest including, for example, a regulatory
sequence, a er sequence, an enhancer sequence, a riptional repressor-binding
sequence, or a deletion of a non-protein-coding sequence, but does not comprise a deletion of
a n-coding sequence. In addition, the cleotide of interest within the insert
polynucleotide and/or inserted at the target genomic locus on the Y chromosome or at a
challenging target locus can encode a protein expressed in the nervous system, the skeletal
system, the digestive , the circulatory system, the muscular system, the respiratory
system, the cardiovascular , the lymphatic system, the endocrine , the urinary
system, the uctive system, or a combination thereof.
The polynucleotide of st within the insert polynucleotide and/or integrated at
the target genomic locus on the Y chromosome or at a challenging target locus can comprises
a genetic modification in a coding sequence. Such genetic modifications include, but are not
limited to, a deletion mutation of a coding sequence or the fusion of two coding sequences.
The polynucleotide of interest within the insert polynucleotide and/or integrated at
the target genomic locus on the Y chromosome or at a challenging target locus can comprise
a polynucleotide encoding a mutant protein. In one embodiment, the mutant protein is
characterized by an altered binding characteristic, d localization, altered expression,
and/or altered expression pattern. In one embodiment, the polynucleotide of interest within
the insert polynucleotide and/or integrated at the genomic target locus on the Y chromosome
or at a challenging target locus ses at least one disease allele. In such instances, the
e allele can be a dominant allele or the disease allele is a ive allele. er,
the disease allele can comprise a single nucleotide polymorphism (SNP) allele. The
polynucleotide of interest encoding the mutant protein can be from any organism, ing,
but not limited to, a mammal, a non-human mammal, rodent, mouse, rat, a human, a monkey,
an agricultural mammal or a domestic mammal polynucleotide encoding a mutant protein.
The polynucleotide of interest within the insert polynucleotide and/or integrated at
the target genomic locus on the Y chromosome or at a challenging target locus can also
comprise a regulatory sequence, including for example, a promoter sequence, an enhancer
sequence, a transcriptional repressor-binding sequence, or a transcriptional terminator
sequence. In specific embodiments, the polynucleotide of interest within the insert
polynucleotide and/or integrated at the target genomic locus on the Y chromosome or at a
challenging target locus comprises a polynucleotide having a deletion of a non-protein-
coding sequence, but does not comprise a deletion of a protein-coding sequence. In one
embodiment, the deletion of the non-protein-coding ce comprises a deletion of a
regulatory sequence. In another embodiment, the deletion of the regulatory element
comprises a deletion of a promoter sequence. In one embodiment, the deletion of the
regulatory element comprises a on of an enhancer sequence. Such a polynucleotide of
interest can be from any organism, including, but not limited to, a mammal, a non-human
mammal, rodent, mouse, rat, a human, a , an agricultural mammal or a domestic
mammal polynucleotide encoding a mutant protein.
] The various methods disclosed herein can be employed to te a variety of
cations in a challenging genomic locus or in the Y chromosome locus (such as Sry).
Such modifications include, for example, a replacement of an nous nucleic acid
sequence with a gous or an orthologous nucleic acid sequence; a deletion of an
endogenous nucleic acid sequence; a deletion of an endogenous nucleic acid sequence,
wherein the deletion ranges from about 5 kb to about 10 kb, from about 10 kb to about 20 kb,
from about 20 kb to about 40 kb, from about 40 kb to about 60 kb, from about 60 kb to about
80 kb, from about 80 kb to about 100 kb, from about 100 kb to about 150 kb, from about 150
kb to about 200 kb, from about 200 kb to about 300 kb, from about 300 kb to about 400 kb,
from about 400 kb to about 500 kb, from about 500 kb to about 600 kb, from about 600 kb to
about 700 kb, from about 700 kb to about 800 kb, from about 800 kb to about 900 kb, from
about 900 kb to about 1 Mb, from about 500 kb to about 1 Mb, from about 1 Mb to about 1.5
Mb, from about 1.5 Mb to about 2 Mb, from about 2 Mb to about 2.5 Mb, or from about 2.5
Mb to about 3 Mb; an ion of an exogenous nucleic acid ce; an insertion of an
exogenous c acid sequence ranging from about 5kb to about 10kb, from about 10 kb to
about 20 kb, from about 20 kb to about 40 kb, from about 40 kb to about 60 kb, from about
60 kb to about 80 kb, from about 80 kb to about 100 kb, from about 100 kb to about 150 kb,
from about 150 kb to about 200 kb, from about 200 kb to about 250 kb, from about 250 kb to
about 300 kb, from about 300 kb to about 350 kb, or from about 350 kb to about 400 kb; an
insertion of an exogenous nucleic acid sequence comprising a homologous or an orthologous
nucleic acid sequence; an insertion of a ic nucleic acid sequence comprising a human
and a non-human nucleic acid sequence; an insertion of a conditional allele flanked with site-
specific recombinase target sequences; an insertion of a selectable marker or a reporter gene
operably linked to a third promoter active in the mammalian cell; or a combination thereof.
III. Methods ofIntroducing Sequences and Generation of Transgenic Animals
As outlined above, methods and compositions are provided herein to allow for the
targeted genetic modification of one or more polynucleotides of interest located on the Y
chromosome, at a challenging target locus, or a decrease in the level and/or activity of the Sry
protein. It is r recognized that in addition to a targeted genetic modification to a
sequence on the Y chromosome or on a challenging target chromosomal locus, additional
targeted genetic modification can be made on other chromosomes. Such systems that allow
for these targeted genetic cations can employ a variety of components and for ease of
reference, herein the term “targeted genomic integration system” generically includes all the
components required for an integration event (i.e. the various nuclease agents, recognition
sites, insert DNA polynucleotides, targeting vectors, target genomic locus, and
polynucleotides of interest).
The methods provided herein comprise introducing into a cell one or more
polynucleotides or polypeptide constructs comprising the various components of the targeted
genomic ation . "Introducing" means presenting to the cell the ce
eptide or polynucleotide) in such a manner that the sequence gains access to the
interior of the cell. The s provided herein do not depend on a ular method for
introducing any component of the targeted c integration system into the cell, only that
the polynucleotide gains access to the interior of a least one cell. Methods for introducing
polynucleotides into various cell types are known in the art and include, but are not limited
to, stable ection methods, transient transfection methods, and virus-mediated s.
In some embodiments, the cells employed in the methods and compositions have a
DNA construct stably orated into their genome. "Stably incorporated" or "stably
introduced" means the introduction of a polynucleotide into the cell such that the nucleotide
sequence integrates into the genome of the cell and is capable of being inherited by y
thereof. Any protocol may be used for the stable incorporation of the DNA constructs or the
various components of the targeted genomic ation system.
Transfection protocols as well as protocols for introducing ptides or
polynucleotide sequences into cells may vary. miting transfection methods include
chemical-based transfection methods include the use of liposomes; nanoparticles; calcium
ate (Graham et al. (1973). Virology 52 (2): 456—67, Bacchetti et al. (1977) Proc Natl
Acad Sci USA 74 (4): 1590—4 and, Kriegler, M (1991). Transfer and Expression: A
Laboratory Manual. New York: W. H. Freeman and Company. pp. 96—97); dendrimers; or
cationic polymers such as extran or polyethylenimine. Non chemical methods
include electroporation; Sono-poration; and optical transfection . Particle-based transfection
include the use of a gene gun, magnet assisted transfection ( m, J. (2006) Current
Pharmaceutical hnology 7, 277—28). Viral methods can also be used for transfection.
In one embodiment, the nuclease agent is introduced into the cell simultaneously
with the targeting vector, the smallTVEC, or the large targeting vector (LTVEC).
Alternatively, the nuclease agent is introduced separately from the targeting vector,
smallTVEC, or the LTVEC over a period of time. In one embodiment, the nuclease agent is
introduced prior to the uction of the targeting , smallTVEC, or the LTVEC, while
in other embodiments, the nuclease agent is introduced following introduction of the
targeting vector, smallTVEC, or the LTVEC.
Non-human animals can be generated employing the various methods disclosed
herein. Such methods comprises (1) integrating one or more polynucleotide of interest at the
target c locus of the Y chromosome of a pluripotent cell of the non-human animal to
generate a genetically modified pluripotent cell comprising the insert polynucleotide in the
targeted genomic locus of the Y chromosome employing the methods disclosed herein; (2)
selecting the genetically modified pluripotent cell having the one or more polynucleotides of
interest at the target genomic locus of the Y chromosome; (3) introducing the genetically
modified pluripotent cell into a host embryo of the non-human animal at a pre-morula stage;
and (4) implanting the host embryo comprising the genetically modified pluripotent cell into
a surrogate mother to te an F0 generation derived from the genetically modified
pluripotent cell. Similar methods can be employed to target a challenging target
chromosomal locus. The non-human animal can be a man , a , a
mouse, a rat, a hamster, a monkey, an ltural mammal or a domestic mammal, or a fish
or a bird.
The pluripotent cell can be a human ES cell, a non-human ES cell, a rodent ES
cell, a mouse ES cell, a rat ES cell, a hamster ES cell, a monkey ES cell, an agricultural
mammal ES cell or a icated mammal ES cell. In other embodiments, the pluripotent
cell is a mammalian cell, human cell, a non-human mammalian cell, a human pluripotent cell,
a human ES cell, a human adult stem cell, a developmentally-restricted human progenitor
cell, a human iPS cell, a human cell, a rodent cell, a rat cell, a mouse cell, a hamster cell. In
one embodiment, the targeted genetic cation decreases the level and/or activity of the
2015/038001
Sry protein. In such instances, the pluripotent cell can comprise an XY ES cell or an XY iPS
cell. Methods of culturing such cells to e the development of F0 fertile XY female
animals are described in detail elsewhere herein.
Nuclear transfer ques can also be used to generate the man
mammalian animals. Briefly, methods for nuclear er include the steps of: (l)
enucleating an oocyte; (2) isolating a donor cell or nucleus to be combined with the
enucleated oocyte; (3) inserting the cell or nucleus into the ated oocyte to form a
reconstituted cell; (4) implanting the reconstituted cell into the womb of an animal to form an
embryo; and (5) allowing the embryo to develop. In such methods oocytes are generally
retrieved from deceased animals, although they may be isolated also from either oviducts
and/or ovaries of live animals. s can be d in a variety of medium known to
those of ry skill in the art prior to ation. Enucleation of the oocyte can be
performed in a number of manners well known to those of ordinary skill in the art. Insertion
of the donor cell or nucleus into the enucleated oocyte to form a reconstituted cell is usually
by microinjection of a donor cell under the zona pellucida prior to fusion. Fusion may be
induced by application of a DC electrical pulse across the contact/fusion plane
rofusion), by exposure of the cells to fusion-promoting chemicals, such as polyethylene
glycol, or by way of an inactivated virus, such as the Sendai virus. A reconstituted cell is
typically activated by electrical and/or non-electrical means before, during, and/or after
fusion of the nuclear donor and recipient . Activation s include electric pulses,
chemically induced shock, penetration by sperm, increasing levels of divalent cations in the
oocyte, and reducing phosphorylation of cellular proteins (as by way of kinase inhibitors) in
the oocyte. The activated reconstituted cells, or embryos, are typically cultured in medium
well known to those of ordinary skill in the art and then transferred to the womb of an animal.
See, for example, US20080092249, WO/l999/005266A2, US20040177390,
WO/2008/017234Al, and US Patent No. 7,612,250, each of which is herein incorporated by
reference.
Other methods for making a non-human animal comprising in its ne one or
more genetic modifications as described herein is provided, comprising: (a) modifying a
targeted genomic locus on the Y chromosome of a non-human animal in a prokaryotic cell
employing the various methods described herein; (b) selecting a modified prokaryotic cell
comprising the genetic modification at the targeted c locus; (c) isolating the
genetically modified targeting vector from the genome of the modified prokaryotic cell; (d)
introducing the cally modified targeting vector into a pluripotent cell of the non-human
WO 00805
animal to generate a genetically modified pluripotent cell comprising the insert nucleic acid
at the targeted genomic locus of the Y chromosome; (e) ing the genetically modified
pluripotent cell; (f) introducing the genetically modified pluripotent cell into a host embryo of
the non-human animal at a pre-morula stage; and (g) implanting the host embryo sing
the genetically modified pluripotent cell into a surrogate mother to generate an F0 generation
derived from the genetically modified pluripotent cell. In such s the targeting vector
can comprise a large targeting vector or a smallTVEC. Similar s can be employed to
target a challenging target locus. The non-human animal can be a non-human mammal, a
rodent, a mouse, a rat, a hamster, a monkey, an agricultural mammal or a domestic .
The pluripotent cell can be a human ES cell, a non-human ES cell, a rodent ES cell, a mouse
ES cell, a rat ES cell, a hamster ES cell, a monkey ES cell, an agricultural mammal ES cell or
a domestic mammal ES cell. In other embodiments, the pluripotent cell is a ian cell,
human cell, a non-human mammalian cell, a human pluripotent cell, a human ES cell, a
human adult stem cell, a developmentally-restricted human progenitor cell, a human iPS cell,
a human cell, a rodent cell, a rat cell, a mouse cell, a hamster cell. In one embodiment, the
ed genetic modification ses the level and/or activity of the Sry protein. In such
instances, the pluripotent cell can comprise an XY ES cell or an XY iPS cell. Methods of
culturing such cells to promote the development of F0 fertile XY female animals are
described in detail ere herein.
In further methods, the isolating step (c) r comprises (cl) linearizing the
genetically modified targeting vector (i.e., the genetically ed LTVEC). In still r
embodiments, the introducing step (d) further comprises (dl) introducing a nuclease agent as
described herein into the pluripotent cell. In one embodiment, selecting steps (b) and/or (e)
are carried out by applying a selectable agent as described herein to the prokaryotic cell or the
pluripotent cell. In one embodiment, selecting steps (b) and/or (e) are carried out via a
modification of allele (MOA) assay as described herein.
Further methods for modifying a target genomic locus of an animal cell via
bacterial gous recombination (BHR) in a prokaryotic cell are provided and comprise:
(a) providing a prokaryotic cell comprising a target genomic locus of the Y chromosome; (b)
introducing into the prokaryotic cell a targeting vector (as described above) comprising an
insert cleotide flanked with a first upstream homology arm and a first downstream
homology arm, wherein the insert cleotide comprises a mammalian genomic region,
and introducing into the prokaryotic cell a nuclease agent that makes a nick or double-strand
break at or near the first recognition site, and (c) selecting a targeted prokaryotic cell
comprising the insert polynucleotide at the target genomic locus of the chromosome, wherein
the prokaryotic cell is capable of sing a recombinase that mediates the BHR. Similar
methods can be employed to target a challenging target locus. Steps (a)-(c) can be serially
repeated as sed herein to allow the introduction of multiple insert polynucleotides at the
targeted genomic locus in the prokaryotic cell. Once the targeted genomic locus is “built”
with the prokaryotic cell, a targeting vector comprising the modified target genomic locus of
the Y chromosome can be isolated from the yotic cell and introduced into a target
genomic locus of the Y chromosome within a mammalian cell. Mammalian cells comprising
the modified genomic locus of the Y chromosome can then be made into non-human
transgenic animals.
Further methods for modifying a target genomic locus of an animal cell via
bacterial homologous recombination (BHR) in a prokaryotic cell are provided and comprise:
(a) providing a prokaryotic cell comprising a target c locus of the Y chromosome; (b)
introducing into the prokaryotic cell a targeting vector (as described above) comprising an
insert polynucleotide flanked with a first am homology arm and a first downstream
homology arm, wherein the insert polynucleotide comprises a ian genomic region,
and (c) selecting a targeted prokaryotic cell comprising the insert polynucleotide at the target
genomic locus of the chromosome, wherein the prokaryotic cell is e of expressing a
recombinase that mediates the BHR. Similar s can be ed to target a
challenging target locus. Steps (a)-(c) can be serially repeated as disclosed herein to allow
the introduction of multiple insert polynucleotides at the targeted genomic locus in the
prokaryotic cell. Once the targeted genomic locus is “built” with the prokaryotic cell, a
targeting vector comprising the ed target genomic locus of the Y some can be
isolated from the prokaryotic cell and introduced into a target c locus of the Y
chromosome within a mammalian cell. Mammalian cells comprising the modified genomic
locus of the Y chromosome can then be made into non-human transgenic animals
In some embodiments, various genetic modifications of the target genomic loci
described herein can be carried out by a series of homologous recombination reactions (BHR)
in bacterial cells using an LTVEC d from Bacterial Artificial some (BAC)
DNA using VELOCIGENE® c engineering technology (see, e.g., US Pat. No.
6,586,251 and Valenzuela, D. M. et a1. (2003), High-throughput engineering of the mouse
genome coupled with high-resolution expression analysis, Nature hnology 21(6): 652-
659, which is incorporated herein by reference in their entireties).
] In some embodiments, targeted XY pluripotent and/or totipotent cells (i.e., X YES
cells or XY iPS cells) comprising various genetic cations as described herein are used
as insert donor cells and introduced into a pre-morula stage embryo from a corresponding
organism, e. g., an 8-cell stage mouse embryo, via the VELOCIMOUSE® method (see, e. g.,
US 7,576,259, US 7,659,442, US 7,294,754, and US 2008-0078000 Al, all of which are
incorporated by reference herein in their entireties). The non-human animal embryo
comprising the genetically modified XY pluripotent and/or totipotent cells (i.e., XY ES cells
or XY iPS cells) is ted until the blastocyst stage and then implanted into a surrogate
mother to e an F0 generation. In some embodiments, targeted mammalian ES cells
comprising s c cations as described herein are introduced into a blastocyst
stage embryo. Non-human animals bearing the cally modified genomic locus of the Y
chromosome can be identified via modification of allele (MOA) assay as described herein.
The resulting F0 generation non-human animal derived from the cally modified XY
pluripotent and/or totipotent cells (i.e., X YES cells or XY iPS cells) is crossed to a wild-type
non-human animal to obtain F1 generation offspring. Following genotyping with specific
primers and/or probes, Fl non-human animals that are heterozygous for the genetically
modified c locus are crossed to each other to produce F2 generation non-human
animal offspring that are homozygous for the genetically modified c locus of the Y
chromosome or for the genetically ed challenging target locus.
IV. Cells and Expression Cassettes
The various methods described herein employ a genomic locus targeting system
for the Y chromosome or for a challenging target locus in a cell. Such cells include
prokaryotic cells such as bacterial cells including E. coli, or eukaryotic cells such as yeast,
insect, amphibian, plant, or mammalian cells, including, but not limited to a mouse cell, a rat
cell, a rabbit cell, a pig cell, a bovine cell, a deer cell, a sheep cell, a goat cell, a chicken cell,
a cat cell, a dog cell, a ferret cell, a primate (e. g., et, rhesus monkey) cell, and the like
and cells from domesticated mammals or cells from agricultural mammals. Some cells are
non-human, particularly non-human mammalian cells. In some embodiments, for those
mammals for which suitable genetically modifiable pluripotent cells are not readily available,
other methods are employed to reprogram somatic cells into pluripotent cells, e. g., via
uction into somatic cells of a combination of pluripotency-inducing factors, including,
but not limited to, Oct3/4, Sox2, KLF4, Myc, Nanog, LIN28, and Glisl. In such methods,
the cell can also be a mammalian cell, human cell, a non-human mammalian cell, a non-
human cell, a cell from a rodent, a rat, a mouse, a hamster, a fibroblast cell or any other host
cell. In other ments, the cell is a pluripotent cell, an induced pluripotent stem (iPS)
cell, a non-human embryonic stem (ES) cell. Such cells include pluripotent cells, including,
for example, induced pluripotent stem (iPS) cells, mouse embryonic stem (ES) cells, rat
embryonic stem (ES) cells, human nic (ES) cells, or pmentally restricted
human progenitor cells, a rodent embryonic stem (ES) cell, a mouse embryonic stem (ES)
cell or a rat embryonic stem (ES) cell.
The terms “polynucleotide,” “polynucleotide sequence,77 (Cnucleic acid sequence,”
and “nucleic acid fragment” are used interchangeably herein. These terms encompass
nucleotide sequences and the like. A polynucleotide may be a polymer of RNA or DNA that
is single- or double-stranded, that optionally contains synthetic, non-natural or altered
nucleotide bases. A polynucleotide in the form of a polymer of DNA may be comprised of
one or more segments of cDNA, genomic DNA, synthetic DNA, or mixtures thereof.
Polynucleotides can comprise ibonucleotides and ribonucleotides include both
naturally occurring molecules and synthetic analogues, and any combination these. The
polynucleotides provided herein also encompass all forms of sequences including, but not
limited to, single-stranded forms, double-stranded forms, hairpins, nd-loop ures,
and the like.
] Further provided are inant polynucleotides. The terms “recombinant
polynucleotide” and “recombinant DNA construct” are used interchangeably herein. A
recombinant construct comprises an artificial or heterologous combination of c acid
sequences, e. g., regulatory and coding sequences that are not found together in nature. In
other embodiments, a recombinant construct may comprise regulatory sequences and coding
sequences that are derived from different s, or regulatory sequences and coding
sequences derived from the same source, but arranged in a manner different than that found
in nature. Such a construct may be used by itself or may be used in conjunction with a
vector. If a vector is used, then the choice of vector is ent upon the method that is
used to transform the host cells as is well known to those skilled in the art. For example, a
plasmid vector can be used. Screening may be accomplished by Southern is of DNA,
Northern analysis of mRNA expression, blotting analysis of n expression, or
phenotypic analysis, among .
In specific embodiments, one or more of the components described herein can be
provided in an expression cassette for expression in the pluripotent and/or totipotent cell.
The cassette can include 5' and 3' regulatory sequences operably linked to a polynucleotide
provided . “Operably linked” means a functional linkage between two or more
elements. For example, an operable linkage between a polynucleotide of interest and a
tory sequence (i.e., a promoter) is a functional link that allows for expression of the
polynucleotide of interest. Operably linked elements may be uous or non-contiguous.
When used to refer to the joining of two protein coding regions, operably linked means that
the coding regions are in the same reading frame. In another instance, a nucleic acid sequence
encoding a protein may be operably linked to regulatory sequences (e. g., promoter, enhancer,
silencer sequence, etc.) so as to retain proper transcriptional regulation. The cassette may
additionally contain at least one additional polynucleotide of interest to be co-introduced into
the ES cell. Alternatively, the additional polynucleotide of interest can be provided on
multiple expression cassettes. Such an expression cassette is provided with a plurality of
restriction sites and/or recombination sites for insertion of a recombinant polynucleotide to be
under the transcriptional regulation of the regulatory regions. The expression cassette may
additionally contain selection marker genes.
The expression cassette can e in the 5'-3' direction of ription, a
transcriptional and ational initiation region (i.e., a er), a recombinant
polynucleotide provided herein, and a transcriptional and translational termination region
(i.e., termination region) functional in mammalian cell or a host cell of interest. The
regulatory regions (i.e., promoters, transcriptional regulatory regions, and transcriptional and
translational termination regions) and/or a polynucleotide ed herein may be
native/analogous to the host cell or to each other. Alternatively, the regulatory regions and/or
a polynucleotide provided herein may be heterologous to the host cell or to each other. For
example, a er operably linked to a heterologous polynucleotide is from a species
different from the species from which the polynucleotide was d, or, if from the
same/analogous species, one or both are substantially modified from their original form
and/or genomic locus, or the promoter is not the native promoter for the operably linked
cleotide. Alternatively, the regulatory s and/or a recombinant polynucleotide
provided herein may be entirely synthetic.
The ation region may be native with the transcriptional initiation region,
may be native with the operably linked recombinant polynucleotide, may be native with the
host cell, or may be derived from r source (i.e., foreign or heterologous) to the
er, the recombinant polynucleotide, the host cell, or any combination thereof.
In preparing the expression cassette, the various DNA fragments may be
manipulated, so as to provide for the DNA ces in the proper orientation. Toward this
end, adapters or linkers may be employed to join the DNA fragments or other manipulations
may be ed to e for convenient restriction sites, removal of superfluous DNA,
removal of restriction sites, or the like. For this purpose, in vitro mutagenesis, primer repair,
restriction, annealing, resubstitutions, e. g., transitions and transversions, may be involved.
A number of promoters can be used in the expression cassettes provided herein.
The promoters can be selected based on the desired outcome. It is recognized that different
applications can be enhanced by the use of different promoters in the expression cassettes to
modulate the , location and/or level of expression of the polynucleotide of interest.
Such expression constructs may also contain, if desired, a promoter regulatory region (e. g.,
one conferring inducible, constitutive, environmentally- or developmentally-regulated, or
cell- or tissue-specific/selective expression), a transcription initiation start site, a me
binding site, an RNA sing signal, a transcription ation site, and/or a
polyadenylation signal.
Non-limiting embodiments include:
1. An in vitro culture comprising
(a) a non-human mammalian XY embryonic stem (ES) cell having a modification
that ses the level and/or activity of an Sry protein; and,
(b) a medium sing a base medium and ments suitable for
maintaining the non-human mammalian ES cell in culture, wherein the medium ts one
or more of the following teristic: an osmolality from about 200 mOsm/kg to less than
about 329 mOsm/kg; a conductivity of about 11 mS/cm to about 13 mS/cm; a salt of an
alkaline metal and a halide in a concentration of about 50 mM to about 110 mM; a carbonic
acid salt concentration of about l7mM to about 30 mM; a total alkaline metal halide salt and
carbonic acid salt concentration of about 85mM to about 130 mM; and/or a combination of
any two or more thereof.
2. The in vitro culture of claim 1, wherein the non-human mammalian XY ES
cell is from a rodent.
3. The in vitro e of claim 2, wherein the rodent is a mouse.
4. The in vitro culture of embodiment 3, wherein the mouse XY ES cell is a
VGFl mouse ES cell.
. The in vitro culture of embodiment 2, wherein the rodent is a rat or a hamster.
6. The in vitro e of any one of embodiments 1-5, wherein the decreased
level and/or activity of the Sry protein is from a genetic modification in the Sry gene.
7. The in vitro culture of embodiment 6, wherein the genetic modification in the
Sry gene comprises an insertion of one or more nucleotides, a on of one or more
nucleotides, a substitution of one or more nucleotides, a knockout, a knockin, a replacement
of an endogenous nucleic acid sequence with a heterologous nucleic acid sequence or a
combination thereof.
8. The in vitro e of any one of embodiments 1-7, wherein the non-human
mammalian ES cell comprises one, two, three or more targeted genetic modifications.
9. The in vitro culture of embodiment 8, wherein the targeted genetic
modification comprises an insertion, a deletion, a knockout, a knockin, a point mutation, or a
combination thereof.
. The in vitro culture of embodiment 8, wherein the targeted genetic
modification comprises at least one insertion of a heterologous polynucleotide into the
genome of the XY ES cell.
11. The in vitro culture of any one of ments 8-10, wherein the targeted
genetic modification is on an me.
12. The in vitro culture of any one of embodiments 1-11, wherein the base
medium exhibits 50 i 5 mM NaCl, 26 i 5 mM carbonate, and 218 i 22 mOsm/kg.
13. The in vitro culture of any one of embodiments 1-11, wherein the base
medium exhibits about 3 mg/mL NaCl, 2.2 mg/mL sodium bicarbonate, and 218 mOsm/kg.
14. The in vitro culture of any one of embodiments 1-11, wherein the base
medium ts 87 i 5 mM NaCl, 18 i 5 mM carbonate, and 261 i 26 mOsm/kg.
. The in vitro e of any one of ments 1-11, wherein the base
medium exhibits about 5.1 mg/mL NaCl, 1.5 mg/mL sodium bicarbonate, and 261 g.
16. The in vitro culture of any one of embodiments 1-11, wherein the base
medium exhibits 110 i 5 mM NaCl, 18 i 5 mM carbonate, and 294 i 29 g.
17. The in vitro culture of any one of embodiments 1-11, wherein the base
medium exhibits about 6.4 mg/mL NaCl, 1.5 mg/mL sodium bicarbonate, and 294 mOsm/kg.
18. The in vitro culture of any one of ments 1-11, wherein the base
medium exhibits 87 i 5 mM NaCl, 26 i 5 mM carbonate, and 270 i 27 mOsm/kg.
19. The in vitro culture of any one of embodiments 1-11, n the base
medium exhibits about 5.1 mg/mL NaCl, 2.2 mg/mL sodium bicarbonate, and 270 mOsm/kg.
. The in vitro culture of any one of embodiments 1-11, wherein the base
medium exhibits 87 i 5 mM NaCl, 26 i 5 mM carbonate, 86 i 5 mM glucose, and 322 i 32
mOsm/kg.
21. The in vitro culture of any one of embodiments 1-11, wherein the base
medium exhibits about 5.1 mg/mL NaCl, 2.2 mg/mL sodium onate, 15.5 mg/mL
glucose, and 322 mOsm/kg.
22. The in vitro culture of any one of ments 1-21, wherein upon
introduction of the non-human mammalian XY ES cells into a host embryo and following
gestation of the host embryo, at least 80% of the F0 non-human mammals are XY females
which upon attaining sexual maturity the F0 XY female non-human mammal is fertile.
23. A method for making a fertile female XY non-human mammal in an F0
generation, sing:
(a) ing a donor non-human mammalian XY embryonic stem (ES) cell
having a modification that decreases the level and/or activity of an Sry protein in a medium
comprising a base medium and supplements suitable for maintaining the non-human
mammalian ES cell in culture, wherein the medium exhibits a characteristic comprising one
or more of the following: an osmolality from about 200 mOsm/kg to less than about 329
mOsm/kg; a conductivity of about 11 mS/cm to about 13 mS/cm; a salt of an alkaline metal
and a halide in a concentration of about 50mM to about 110 mM; a carbonic acid salt
concentration of about 17 mM to about 30 mM; a total ne metal halide salt and carbonic
acid salt concentration of about 85 mM to about 130 mM; and/or a combination of any two or
more thereof;
(b) introducing the donor XY non-human mammalian ES cell into a host ;
(c) gestating the host embryo; and,
(d) obtaining an F0 XY female man mammal, wherein upon attaining
sexual maturity the F0 XY female non-human mammal is fertile.
24. The method of embodiment 23, wherein the non-human mammalian XY ES
cell is from a rodent.
. The method of embodiment 24, wherein the rodent is a mouse.
26. The method of embodiment 25, wherein the mouse XY ES cell is a VGFl
mouse ES cell.
27. The method of embodiment 24, wherein the rodent is a rat or a hamster.
28. The method of any one of embodiments 23-27, wherein the sed level
and/or activity of the Sry protein is from a genetic modification in the Sry gene.
29. The method of embodiment 28, wherein the genetic modification in the Sry
gene comprises an insertion of one or more nucleotides, a deletion of one or more
tides, a substitution of one or more nucleotides, a knockout, a knockin, a replacement
of an endogenous nucleic acid sequence with a heterologous nucleic acid sequence or a
ation thereof.
. The method of any one of embodiments 23-29, wherein the non-human
mammalian ES cell comprises one, two, three or more targeted genetic modifications.
31. The method of embodiment 30, wherein the targeted genetic modification
comprises an insertion, a deletion, a ut, a knockin, a point mutation, or a combination
thereof.
32. The method of embodiment 30, n the targeted genetic modification
comprises at least one insertion of a heterologous polynucleotide into a genome of the XY ES
cell.
33. The method of any one of embodiments 30-32, wherein the targeted genetic
modification is on an autosome.
34. The method of any one of embodiments 23-33, wherein the base medium
ts 50 i 5 mM NaCl, 26 i 5 mM carbonate, and 218 i 22 mOsm/kg.
. The method of any one of embodiments 23-33, wherein the base medium
exhibits about 3 mg/mL NaCl, 2.2 mg/mL sodium onate, and 218 mOsm/kg.
36. The method of any one of embodiments 23-33, wherein the base medium
exhibits 87 i 5 mM NaCl, 18 i 5 mM carbonate, and 261 i 26 mOsm/kg.
37. The method of any one of embodiments 23-33, wherein the base medium
exhibits about 5.1 mg/mL NaCl, 1.5 mg/mL sodium bicarbonate, and 261 mOsm/kg.
38. The method of any one of embodiments 23-33, wherein the base medium
exhibits 110 i 5 mM NaCl, 18 i 5 mM carbonate, and 294 i 29 mOsm/kg.
39. The method of any one of ments 23-33, wherein the base medium
exhibits about 6.4 mg/mL NaCl, 1.5 mg/mL sodium bicarbonate, and 294 mOsm/kg.
40. The method of any one of embodiments 23-33, wherein the base medium
ts 87 i 5 mM NaCl, 26 i 5 mM carbonate, and 270 i 27 mOsm/kg.
41. The method of any one of embodiments 23-33, wherein the base medium
exhibits about 5.1 mg/mL NaCl, 2.2 mg/mL sodium bicarbonate, and 270 mOsm/kg.
42. The method of any one of ments 23-33, wherein the base medium
exhibits 87 i 5 mM NaCl, 26 i 5 mM carbonate, 86 i 5 mM glucose, and 322 i 32
mOsm/kg.
43. The method of any one of embodiments 23-33, wherein the base medium
exhibits about 5.1 mg/mL NaCl, 2.2 mg/mL sodium bicarbonate, 15.5 mg/mL glucose, and
322 mOsm/kg.
44. A method of producing a transgenic non-human mammal homozygous for a
targeted genetic mutation in the F1 generation comprising: (a) crossing an F0 XY fertile
female having a decreased level and/or ty of the Sry protein with a cohort clonal sibling,
derived from the same ES cell clone, F0 XY male non-human mammal, wherein the F0 XY
fertile female man mammal and the F0 XY male non-human mammal each is
heterozygous for the genetic mutation; and, (b) obtaining an F1 progeny mouse that is
homozygous for the c modification.
45. A method for modifying a target genomic locus on the Y chromosome in a cell
comprising: (a) providing a cell comprising a target genomic locus on the Y chromosome
comprising a recognition site for a nuclease agent, (b) introducing into the cell (i) the
nuclease agent, wherein the nuclease agent induces a nick or -strand break at the first
recognition site; and, (ii) a first targeting vector comprising a first insert polynucleotide
flanked by a first and a second homology arm corresponding to a first and a second target site
located in sufficient proximity to the first recognition site, n a sum total of the first
homology arm and the second homology arm is at least 4kb but less than 150kb; and, (c)
identifying at least one cell comprising in its genome the first insert polynucleotide integrated
at the target genomic locus.
46. A method for modifying a target genomic locus on the Y chromosome in a cell
comprising:
(a) providing a cell sing a target genomic locus on the Y chromosome
comprising a recognition site for a nuclease agent,
(b) introducing into the cell a first targeting vector comprising a first insert
polynucleotide flanked by a first and a second homology arm ponding to a first and a
second target site, wherein a sum total of the first homology arm and the second homology
arm is at least 4kb but less than 150kb; and,
(c) identifying at least one cell comprising in its genome the first insert
polynucleotide integrated at the target c locus.
47. The method of embodiment 45 or 46, wherein the cell is a mammalian cell.
48. The method of embodiment 47, wherein the ian cell is a non-human
cell.
49. The method of embodiment 47, wherein the mammalian cell is from a rodent.
50. The method of embodiment 49, wherein the rodent is a rat, a mouse or a
hamster.
51. The method of any one of ments 45-50, wherein the cell is a
pluripotent cell.
52. The method of any one of embodiments 45-50, wherein the mammalian cell is
an induced pluripotent stem (iPS) cell.
53. The method of ment 51, wherein the pluripotent cell is a non-human
embryonic stem (ES) cell.
54. The method of embodiment 51, wherein the pluripotent cell is a rodent
embryonic stem (ES) cell, a mouse embryonic stem (ES) cell or a rat embryonic stem (ES)
cell.
55. The method of any one of embodiments 45 and 47-54, wherein the nuclease
agent is an mRNA encoding a nuclease.
56. The method of any one of ments 45 and 47-54, wherein the nuclease
agent is a zinc finger nuclease (ZFN).
57. The method of any one of embodiments 45 and 47-54, wherein the nuclease
agent is a Transcription Activator-Like Effector se ).
58. The method of any one of embodiments 45 and 47-54, wherein the nuclease
agent is a meganuclease.
59. The method any one of ments 45 and 47-54, wherein the nuclease
agent is a CRISPR RNA guided Cas9 endonuclease.
60. A method for modifying the Y chromosome comprising exposing the Y
chromosome to a Cas n and a CRISPR RNA in the presence of a large targeting vector
(LTVEC) comprising a nucleic acid sequence of at least 10 kb, wherein following exposure
to the Cas protein, the CRISPR RNA, and the LTVEC, the Y chromosome is modified to
contain at least 10 kb nucleic acid sequence.
61. The method of embodiment 60, wherein the LTVEC comprises a nucleic acid
sequence of at least 20 kb, at least 30 kb, at least 40 kb, at least 50 kb, at least 60 kb, at least
70 kb, at least 80 kb, or at least 90 kb.
62. The method of embodiment 60, wherein the LTVEC ses a nucleic acid
sequence of at least 100 kb, at least 150 kb, or at least 200 kb.
63. A method for modifying a target genomic locus on the Y chromosome,
comprising: (a) ing a mammalian cell comprising the target genomic locus on the Y
chromosome, wherein the target genomic locus comprises a guide RNA (gRNA) target
sequence; (b) introducing into the ian cell: (i) a large targeting vector (LTVEC)
comprising a first nucleic acid flanked with targeting arms homologous to the target genomic
locus, wherein the LTVEC is at least 10 kb; (ii) a first expression construct comprising a first
promoter operably linked to a second nucleic acid encoding a Cas protein, and (iii) a second
sion uct comprising a second promoter operably linked to a third nucleic acid
encoding a guide RNA (gRNA) comprising a nucleotide ce that hybridizes to the
gRNA target sequence and a trans-activating CRISPR RNA (trachNA), wherein the first and
the second promoters are active in the mammalian cell; and (c) identifying a modified
mammalian cell comprising a targeted genetic modification at the target genomic locus on the
Y some.
64. The method of embodiment 63, wherein the LTVEC is at least 15 kb, at least
kb, at least 30kb, at least 40 kb, at least 50 kb, at least 60 kb, at least 70 kb, at least 80 kb,
or at least 90 kb.
65. The method of embodiment 63, wherein the LTVEC is at least 100 kb, at least
150 kb, or at least 200 kb.
66. The method of ment 63, wherein the mammalian cell is a non-human
mammalian cell.
67. The method of embodiment 63, n the mammalian cell is a fibroblast
cell.
68. The method of embodiment 63, wherein the mammalian cell is from a rodent.
69. The method of embodiment 68, n the rodent is a rat, a mouse, or a
hamster.
70. The method of embodiment 63, wherein the mammalian cell is a pluripotent
cell.
71. The method of embodiment 70, wherein the pluripotent cell is an induced
pluripotent stem (iPS) cell.
72. The method of embodiment 70, wherein the pluripotent cell is a mouse
embryonic stem (ES) cell or a rat embryonic stem (ES) cell.
73. The method of embodiment 70, wherein the pluripotent cell is a
developmentally restricted human itor cell.
74. The method of embodiment 63, wherein the Cas protein is a Cas9 protein.
75. The method of embodiment 74, wherein the gRNA target sequence is
immediately flanked by a Protospacer Adjacent Motif (PAM) sequence.
76. The method of ment 63, wherein the sum total of 5’ and 3’ homology
arms of the LTVEC is from about 10 kb to about 150 kb.
77. The method of embodiment 76, wherein the sum total of the 5’ and the 3’
homology arms of the LTVEC is from about 10 kb to about 20 kb, from about 20 kb to about
40 kb, from about 40 kb to about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to
about 100 kb, from about 100 kb to about 120 kb, or from about 120 kb to 150 kb.
78. The method of embodiment 63, wherein the targeted genetic modification
comprises: (a) a replacement of an endogenous nucleic acid sequence with a homologous or
an ogous nucleic acid ce; (b) a deletion of an endogenous nucleic acid
sequence; (c) a deletion of an endogenous nucleic acid sequence, wherein the deletion ranges
from about 5 kb to about 10 kb, from about 10 kb to about 20 kb, from about 20 kb to about
40 kb, from about 40 kb to about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to
about 100 kb, from about 100 kb to about 150 kb, or from about 150 kb to about 200 kb, from
about 200 kb to about 300 kb, from about 300 kb to about 400 kb, from about 400 kb to about
500 kb, from about 500 kb to about 1 Mb, from about 1 Mb to about 1.5 Mb, from about 1.5
Mb to about 2 Mb, from about 2 Mb to about 2.5 Mb, or from about 2.5 Mb to about 3 Mb;
(d) insertion of an exogenous nucleic acid sequence; (e) insertion of an exogenous
nucleic acid sequence ranging from about 5kb to about 10kb, from about 10 kb to about 20
kb, from about 20 kb to about 40 kb, from about 40 kb to about 60 kb, from about 60 kb to
about 80 kb, from about 80 kb to about 100 kb, from about 100 kb to about 150 kb, from
about 150 kb to about 200 kb, from about 200 kb to about 250 kb, from about 250 kb to about
300 kb, from about 300 kb to about 350 kb, or from about 350 kb to about 400 kb; (f)
ion of an ous nucleic acid sequence comprising a gous or an orthologous
nucleic acid sequence; (g) insertion of a ic nucleic acid sequence comprising a
human and a man nucleic acid sequence; (h) insertion of a conditional allele flanked
with site-specific recombinase target sequences; (i) insertion of a selectable marker or a
reporter gene operably linked to a third promoter active in the mammalian cell; or (1') a
combination f.
79. The method of embodiment 63, wherein the target genomic locus comprises
(i) a 5’ target sequence that is homologous to a 5’ homology arm; and (ii) a 3’ target sequence
that is homologous to a 3’ homology arm.
80. The method of embodiment 79, wherein the 5’ target sequence and the 3’
target sequence is separated by at least 5 kb but less than 3 Mb.
81. The method of embodiment 79, wherein the 5’ target sequence and the 3’
target sequence is separated by at least 5 kb but less than 10 kb, at least 10 kb but less than 20
kb, at least 20 kb but less than 40 kb, at least 40 kb but less than 60 kb, at least 60 kb but less
than 80 kb, at least about 80 kb but less than 100 kb, at least 100 kb but less than 150 kb, or at
least 150 kb but less than 200 kb, at least about 200 kb but less than about 300 kb, at least
about 300 kb but less than about 400 kb, at least about 400 kb but less than about 500 kb, at
least about 500 kb but less than about le, at least about 1 Mb but less than about 1.5 Mb, at
least about 1.5 Mb but less than about 2 Mb, at least about 2 Mb but less than about 2.5 Mb,
or at least about 2.5 Mb but less than about 3 Mb.
82. The method of embodiment 63, wherein the first and the second expression
constructs are on a single nucleic acid molecule.
83. The method of embodiment 63, wherein the target genomic locus comprises
the Sry locus.
84. A method for targeted genetic modification on the Y chromosome of a non-
human animal, comprising: (a) modifying a genomic locus of interest on the Y chromosome
of a non-human pluripotent cell according to the method of embodiment 4, thereby producing
a genetically ed non-human pluripotent cell comprising a targeted genetic modification
on the Y chromosome; (b) introducing the modified non-human otent cell of (a) into a
non-human host embryo; and (c) gestating the non-human host embryo comprising the
modified pluripotent cell in a surrogate , wherein the surrogate mother produces F0
progeny comprising the targeted genetic cation, wherein the targeted genetic
modification is capable of being itted h the germline.
85. The method of embodiment 84, wherein the genomic locus of interest
comprises the Sry locus.
86. A method for modifying a target genomic locus on the Y chromosome in a cell
comprising: (a) providing a cell comprising a target genomic locus on the Y chromosome
comprising a recognition site for a nuclease agent, (b) introducing into the cell (i) the
se agent, wherein the nuclease agent induces a nick or -strand break at the first
recognition site; and, (ii) a first targeting vector comprising a first insert polynucleotide
flanked by a first and a second homology arm corresponding to a first and a second target site
located in sufficient proximity to the first recognition site, wherein the length of the first
gy arm and/or the second homology arm is at least 400 bp but less than 1000 bp; and,
(c) identifying at least one cell comprising in its genome the first insert polynucleotide
integrated at the target genomic locus.
87. The method of embodiment 86, wherein the length of the first homology arm
and/or the second homology arm is from about 700 bp to about 800 bp.
88. The method of embodiment 86, wherein the modification comprises a deletion
of an nous nucleic acid sequence.
89. The method of embodiment 88, n the deletion ranges from about 5 kb to
about 10 kb, from about 10 kb to about 20 kb, from about 20 kb to about 40 kb, from about
40 kb to about 60 kb, from about 60 kb to about 80 kb, from about 80 kb to about 100 kb,
from about 100 kb to about 150 kb, or from about 150 kb to about 200 kb, from about 200 kb
to about 300 kb, from about 300 kb to about 400 kb, from about 400 kb to about 500 kb, from
about 500 kb to about 1 Mb, from about 1 Mb to about 1.5 Mb, from about 1.5 Mb to about 2
Mb, from about 2 Mb to about 2.5 Mb, or from about 2.5 Mb to about 3 Mb.
90. The method of embodiment 88, wherein the deletion is at least 500 kb.
The t methods and compositions may be embodied in many different forms
and should not be construed as limited to the embodiments set forth herein; rather, these
embodiments are provided so that this disclosure will satisfy able legal requirements. Like
numbers refer to like elements hout.
Many modifications and other embodiments of the methods and compositions set
forth herein will come to mind to one skilled in the art to which this methods and compositions
pertains having the benefit of the teachings presented in the foregoing descriptions and the
associated drawings. Therefore, it is to be understood that the s and compositions are not
to be limited to the specific embodiments disclosed and that modifications and other
embodiments are included within the scope of the appended claims. gh specific terms are
employed herein, they are used in a generic and descriptive sense only and not for purposes of
limitation.
The following examples are offered by way of illustration and not by way of
limitation.
EXAMPLES
Example 1. Targeting of the Y Chromosome Gene S[y Assisted by TALENs or CRISPR
A targeted deletion comprising a lacZ replacement allele for Sry was created with
a ing vector comprising, in order, an upstream homology arm of approximately 700 bp,
a alactosidase coding sequence (lacZ) followed by a polyadenylation signal, a
neomycin resistance cassette flanked by loxP sites comprising a human ubiquitin C promoter,
including the first exon, first intron, and part of the second exon, a neomycin
phosphotransferase coding sequence, and a enylation signal, and a downstream
homology arm of approximately 650 bp. The allele created by t targeting of the Sry
gene with the targeting vector comprises a deletion of the approximately 1 kb Sry open
[Link]
http://www.velocigene.com/komp/detail/12778
[Link]
http://www.velocigene.com/komp/detail/12778
reading frame and replacement with the lacZ-neo cassette such that the beta-galactosidase
coding sequence is fused in-frame at the Sry start codon. The targeting vector was used to
target the Sry gene in both the VGB6 (a.k.a. B6A6) C57BL/6 and the VGF1 (a.k.a.
F1H4) C57BL6/129 F1 hybrid ES cell lines. VGF1 (F1H4) mouse ES cells were derived
from hybrid embryos produced by ng a female C57BL/6NTac mouse to a male
SvEvTac mouse. Therefore, VGF1 ES cells contain a Y some from
129S6/SvEvTac mouse. The female XY mice produced from the VGF1 cell line contain a Y
chromosome derived from 129S6/SvEvTac mouse.
Example 2. TALEN- or CRISPR-induced Mutations in the Y some Gene Sry
] Deletion mutations, presumably the result of non-homologous end g (NHEJ)
repair of double strand DNA breaks, ranging from 3 bp to 1.2 kb and larger were created in
the Sry gene by the action of a TALEN or of CRISPR guide RNAs, in combination with Cas9
DNA endonuclease (see, . ES cells comprising the TALEN- and CRISPR-induced
mutations in the Sry gene also carried random transgenic insertions of the NIH KOMP project
VG12778 LTVEC ( available via internet on the world wide web (www) at the URL
“velocigene.com/komp/detail/12778”), which comprises a deletion of the Sry coding
ce and replacement with an insertion cassette comprising lacZ fused in-frame with the
Sry start codon and a neomycin resistance gene flanked by homology arms of 38 and 37 kb
and based on a BAC from the bMQ library (129S7/SvEv Brd-Hprt b-m2). The LTVEC
comprises in its homology arms all the known control elements for the expression of Sry. Its
lacZ-encoded beta-galactosidase serves as a reporter for the tissue-specific and
pmental specific expression of the Sry gene. TALEN- and CRISPR-induced
mutations accompanied by LTVEC insertions were created in both the VGB6 (a.k.a. B6A6)
and VGF1 (a.k.a. F1H4) ES cell lines.
We obtained a TALEN (TALEN-1) designed to target part of the HMG box DNA
binding motif coding sequence (upstream recognition sequence: 5´-
TCCCGTGGTGAGAGGCAC-3´ (SEQ ID NO. 72); downstream recognition sequence: 5´-
TATTTTGCATGCTGGGAT-3´ (SEQ ID NO. 73) in the Sry gene. TALEN-1 was active in
creating NHEJ ons at the Sry locus in multiple experiments.
Both VGB6 and VGF1 mouse ES cells were created with TALEN-induced
mutations. Table 1 contains a list of all the clones and the sizes of the on mutations
they carry. (ND in Table 1 indicates that a mutation was detected by a qPCR assay, but the
exact molecular nature of the mutation was not determined.) All clones also carry at least one
copy of the NIH KOMP project VG12778 LTVEC.
Table 1
TALEN- and CRISPR-induced mutations in the Sry gene
ES cell Clone Mutation inducing agent Deletion (bp)
VGB6 DE7 TALEN- 1 9
DEl 1 TALEN- 1 303*
DG5 TALEN- 1 627
DH1 TALEN- 1 ND
EA2 TALEN- 1 ND
ED4 TALEN- 1 > 1200
EF4 TALEN- 1 16
EG7 TALEN-l >1200
VGB6 RD3 TALEN- 1 >1200
RE9 TALEN- 1 ND
RF3 TALEN- 1 9
RG7 TALEN- 1 15
SF7 TALEN- 1 3
SGl 1 TALEN- 1 6
SH2 TALEN- 1 >200
SHl 1 TALEN- 1 2
VGFl TB1 TALEN- 1 1 1
TC2 TALEN-l 5
UA5 TALEN- 1 15
UB5 TALEN- 1 1201
UE12 TALEN- 1 9
WEI 1 TALEN- 1 >1200
VGB6 0G6 -2 9
QE8 CRISPR-3 5
VGFl AU-B6 CRISPR-4 5
AU-C12 CRISPR-4 8
AW-H5 -S 22
*Also contained a 50 bp insertion
The results of microinjections of the Sry mutant clones are set forth in Table 2 and
the breeding results of sex-reversed females are set forth in Table 3.
Table 2
F0 generation VelociMice produced by njection of Sry mutant ES cell clones into 8—cell embryos
ES Cell Clone Sry mutation Female VM Male VM
VGB6 ED4 >1 kb deletion 2 O
EG7 >1 kb deletion 19 O
GB4 None 3
GGl None 5
DEll 303 bp deletion; 50 bp insertion O
DG5 627 bp deletion ll 0
VGFl TA3 None 0 5
TA4 None 0 l l
TBl 11 bp deletion 2 0
TC2 5 bp deletion 8 O
TH4 None 2 6
UB5 1,201 bp deletion 6 0
WEll >1.2 kb deletion 7 0
UA5 15 bp deletion 4 O
UE12 9 bp deletion 8 O
Table 3
Breeding results of XY female VelociMice with mutations in the Sry gene
2:11 SW Elgéetion
Clone XYIlSeglale 86 Pups born
VGB6 EG7 >1,200 1460403 0
1460404 0
1460405 0
1460406 1 0*
1460408 0
1460409 0
1460410 0
VGB6 DG5 627 1460428 0
1460429 0
1460430 0
1460431 0
1460432 0
1460436 0
1460437 0
1460438 0
1460410 0
VGF1 UB5 1,201 1525585 5 33
6 5 25
1525587 5 32
1525588 3 35
1525589 3 19
VGF1 WE11 >1,200 1525573 3 4
1525574 5 21
4 11
1525576 4 14
1525577 4 16
1525578 2 4
1525579 2 6
VGF1 TB1 11 1525700 5 30
1525701 4 28
VGF1 TC2 5 1525706 1 2
1525707 5 10
1525708 1 6
1525709 4 17
0 1 3
1525711 3 9
1525712 4 12
1525713 2 7
VGF1 UA5 15 1594102 2 5
1594103 2 7
WO 00805
4 1 3
1594105 2 15
VGF1 UE12 9 1594117 1 6
8 2 12
1594119 1 11
1594120 2 8
1594121 2 21
1594122 2 15
1594123 2 10
*XY Female ID# 6 had to be euthanized before birth because she had a near—term crisis and could not
deliver. Her dead pups (4 male, 5 females) were recovered by dissection and none carried the Sry mutation.
] All of the VelociMice with Sry mutations derived from VGB6 ES cells were
female, as ed for inactivation of Sry (Table 2). Those without Sry mutations but
carrying at least one copy of the NIH KOMP VG12778 LTVEC produced only male
VelociMice (Table 2, clones GB4 and GGl). When 17 Sry mutant female B6 VelociMice
were test bred, only one became pregnant after about four months of breeding set-up (Table
3), and that female had to be euthanized before birth because she had a near-term crisis and
could not deliver. Her dead pups (4 male, 5 females) recovered by dissection were all WT;
none d the Sry mutation. It was concluded that nearly all Sry mutant mice made from
VGB6 ES cells are sterile, which is in agreement with the literature on Sry mutations.
However, our data demonstrated very different result with the VGF1 clones.
First, the VGF1 ES cells were maintained, as usual, in our KO-DMEM-like low
osmotic strength growth medium that is zing: some of the microinj ected XY clones
grown in this medium will produce fertile XY females, i.e. an XY female phenomenon, even
though they do not carry mutations. An example is clone TH4, which has no Sry mutation
but carries at least one copy of the NIH KOMP VG12778 LTVEC. This clone produced 2
female and 6 male VelociMice (Table 2). Two other VGF1 clones with no Sry mutations
(TA3 and TA4, Table 2) produced only male VelociMice. We wanted to determine if VGF1
XY ES cells with mutations in Sry might also be feminized by the medium. In other words,
would they, unlike the VGB6 Sry mutant ES cells, produce some fertile XY Sry mutant
females? (Note that VGB6 ES cells cannot be maintained in KO-DMEM-like low c
strength media and retain the ability to produce mice.) The answer is yes as shown in Table
Six VGF1 ES cell clones with TALEN-induced small deletions ranging from 5 bp
to over 1 kb were microinjected. All produced female VelociMice, 32 of which were bred.
Remarkably, all of the Sry mutant XY female VelociMice were fertile; each produced at least
one litter (Table 3). Many of the Sry mutant XY females produced multiple litters with
normal litter sizes, while some of the XY females ed only one or two small litters. Out
of 299 F1 mice from these breedings that have been genotyped, approximately half (146,
49%) are normal XY males or normal XX females. 174 (58%) of the F1 mice were
phenotypic females, while 125 (42%) were ypic males. 26 of the females (15% of
females, 8.7% of the total F1 generation) were XY females that inherited a mutant Sry allele.
Because of meiotic non-disjunction events associated with XY oocytes, a number of aberrant
genoytpes — XXY, XYY, XO, XXYY — some of which included mutant Sry alleles were
observed in the F1 progeny of Sry mutant XY female VelociMice.
A method for the efficient on of fertile XY female VelociMice from XY ES
cells has been discovered. If inactivating mutations in the Sry gene in ES cells are created
that have been maintained in the feminizing growth medium, a high tion of fertile XY
female mice are obtained that when bred to males produce mostly male and female mice with
normal X and Y chromosomes.
e 3. Embryo recovery in KO-DMEM or DMEM after TALEN-induced Mutations in
the Y Chromosome Gene Sry
Correct targeting of mouse Sry by LTVEC was confirmed or negated by
genotyping of F1 offspring derived from F0 females, which were XY and carried Sry
mutation. Co-segregation in F1 mice of the LacZ/Neo cassette with the Sry mutation (as
assessed by Sry LOA assays) strongly suggests t targeting. Failure of LacZ/Neo to co-
segregate with the on indicates that the original clone contained an Sry deletion
mutation (induced by TALEN) coupled with a LacZ/Neo transgenic ion elsewhere in
the genome.
Offspring from XY females with Sry mutations exhibited a variety of abnormal
karyotypes at a high frequency (including XXY, XYY, and X0). Sex some count was
ed by using unrelated loss of allele (LOA) assays for genes on X and Y chromosomes.
The copy number of Sry was then ined using LOA assays. The presence of mutant Sry
allele was inferred in mice in which the Y chromosome copy number exceeded the Sry copy
number (for instance, 1 copy of Y and 0 copies of Sry, or 2 copies of Y and 1 copy of Sry).
Lastly the presence of LacZ and Neo were ined using TaqMan assays.
In the original set of clones, which were created by Sry LTVEC er with
TALEN nuclease and grown in KO-DMEM, it was evident that LacZ/Neo cassette was not
co-segregating with the Sry mutation. A sample litter from these clones is shown in Table 4.
2015/038001
Table 4: Screening of clones generated by Sry LTVEC together with TALEN nuclease
Mouse SeX X Chr Y Chr Sry LacZ Neo Genotype Comments
Copy # Copy # Copy
1656721 M 1 1 1 X+Y+
1656722 M 1 1 1 X+Y+ LacZ/Neo
present by Sry
mutation absent
1656723 M 1 1 1 0 0 X+Y+
1656724 M 1 2 1 0 0 X+Y+YA Sry mutation
present but
LacZ/Neo
absent
F 2 0 0 X+X+ LacZ/Neo
present but Sry
mutation absent
6 F 2 1 0 X+X+ Sry mutation
YA t but
LacZ/Neo
absent
1656727 F 2 1 0 X+X+
1656728 F 1 0 0 X+ LacZ/Neo
present but Sry
mutation absent
1656729 F 1 0 0 X+
In the subsequent set of , which were created by Sry LTVEC together with
TALEN nuclease and grown in DMEM, the LacZ/Neo cassette was completely co-
ating with the Sry mutation, indicating correct targeting. A typical litter from these
clones is shown in Table 5.
Table 5: Screening results for clones created by Sry LTVEC together with TALEN nuclease
Mouse SeX X Chr Y Chr Sry LacZ Neo Genotype
Copy # Copy # Copy
1848360 M 1 1 1 0 0 X+Y+
1848361 M 1 1 1 0 0 X+Y+
1848362 M 1 1 1 0 0 X+Y+
1848363 1 1 0 0
1848364 1 1
1848365
1848366
1848367
2015/038001
Example 4: TALEN and CRISPR-Assisted ing of Sry by SmallTVECs or LTVECs
As depicted in a targeted deletion sing a lacZ replacement allele for
Sry was created with either a LTVEC or a small targeting vector (smallTVEC) together with
either TALEN nuclease or CRISPR guide RNAs, in combination with Cas9 DNA
endonuclease. The VEC comprised, in order, an upstream homology arm of
approximately 700-800 bp, a beta-galactosidase coding sequence (lacZ) followed by a
polyadenylation signal, a neomycin resistance cassette flanked by loxP sites comprising a
human ubiquitin C promoter, including the first exon, first intron, and part of the second
exon, a neomycin phosphotransferase coding sequence, and a polyadenylation signal, and a
downstream homology arm of approximately 700-800 bp. The allele created by correct
targeting of the Sry gene with the targeting vector comprises a deletion of the approximately
1 kb Sry open reading frame and replacement with the lacZ-neo cassette such that the beta-
galactosidase coding sequence is fused in-frame at the Sry start codon. The ing vector
was used to target the Sry gene in the VGFl (a.k.a. FlH4) /129 F1 hybrid ES cell
line and in the VGB6 ES cell line (a.k.a. B6A6).
As illustrated in Table 6, clones produced using four different gRNAs and one TALEN pair
were produced and screened for cleavage and loss of allele by TaqMan assays.
Table 6: Screening results for cleavage and loss of allele
Small TVEC
Targeting
Location Clones Total targ. Eff.
Screened (%)
HMG box 192 2.1
HMG box 192 2.6
3’ end 192 1.6
3’ end gRNA 5 192 2.6
HMG box TALEN pair 1 384 0.3
The LTVEC transgenic clones produced embryos with the same lacZ pattern. rates LacZ expression in the embryos.
Table 7 reports the fertility results of XY Females derived from ES cells grown in
conventional DMEM-based medium that had TALEN-assisted LTVEC targeted deletion-
replacement mutations of Sry. Unexpectedly compared with the results for a similar
ment with ES cells grown in KO-DEMEM-based medium (Table 3), LTVEC targeting
in DMEM-based medium produced clones with tly targeted Sry deletions and lacZ—neo
ions. Forty out of 41 XYS'WQCZ) females derived from four targeted clones produced live
born pups upon mating — a 98% fertility rate. Thus, we have d two new ways to
produce highly fertile XY females from mutant ES cells: (1) TALEN-induced inactivating
mutations in Sry in ES cells grown in a KO-DMEM-based medium; and (2) TALEN-assisted
LTVEC targeted precise on-replacement mutations in ES cells grown in DMEM-based
medium.
Table 7: Production of Sry TALEN Mutant XY Females
Clone ES Allele XY female XY Fertile Fertility
cell description VelocMice females XY rates
line bred females (%)
X-C4 VGFl lacZ—neo
2 2 100
targeted
X-El 0 VGFl lacZ—neo
1 1 1 100
XOF3 VGFl lacZ—neo
2 5 3 3 100
m targeted
2 X-G3 VGFl lacZ—neo
Q 9 3 2 67
targeted
VGFl Total 53 41 40 98
Example 5: Large on on the Y Chromosome Mediated by ZFNs
As illustrated in large deletions, 500 kb or greater, were made on the Y
chromosome using ZFNs targeting the Kdm5d and the Usp9y genes. Table 8 provides
examples of zinc finger sequences on the Y chromosome.
Table 8: Zinc Finger Sequence on the Y Chromosome
Target SEQ ID
Y CHR Plate ch Fmger Sequence_ _ ZFN#
Name N0:
ZFN1 42
NMOll419—r43102al ttAGGTAGGTAGACAGGGATgttttctg
KDMSD NMOll419—43108al atCCAGTCtCTGAAGGAAGCTctgacta
19—rl9880al caAAAGCTTCAGGGGGActcttacactc
NMOll419—19887al ttTGAGCAgGCTACACAGGAGtatactt
ZFNS 46
—rl7347al aaGCGGTGgCAATAGGCAaaagatgtgg
011419—17353al ctGAAGTCCCCAAGGGAGTAtggagatg
Oll4lg—rl7350al agAAAGCGGTGGCAaTAGGCAaaagatg
011419—17356al aaGTCCCCAAGGGAGTAtggagatgccc
012008—r8130al acTCCAACGACTATGACcactccgttca
012008—8136al acAGATCAGATGAAGATgactggtcaaa
012008—r7l72al ctTTCAAGGAAAAAAAGaacaaaaccca
DDX3Y 012008—7178al ggTCTGTGATAAGGACAGTTcaggatgg
—r20472al taAATCTGACTGAGAATGGGtagtagaa
012008—20479al caGATGGTCCAGGAGAGGCTttgaaggc
012008—r7267al atTGGGCTTCCCTCTGGAatcacgagat
OlZOO8—7274al ttTCAGTGATCGTGGAAGTGgatccagg
l48943—r9256lal ctGGTTTGGAAATCGTActgtaaaagac
l48943—92567al gcAAAGAGGTTGAGGATttggacatatt
l48943—r11830al gaGGAGTTGTTGGAGAAGTthattgga
[JSPQY l48943—ll836al atATGAACAAGGCCAAthgatgctcca
l48943—r108581al aCTCAGAAGAAGGATTAGGAatgctttg
148943—108588al AGaAATGTATCAGTTcatcttg
l48943—rl6244al tcCATAAGGATTTTGGAaaaagacacag
ZFN8 65
l48943—l6251al agGCTGTGAGTGGATGGAAGtttgaaat
In one experiment, 3.3 million ES cells from VGB6 clones D-G5 and E-G7
(Table 3) were electroporated with the ZFN mRNA pairs Kdm5d-ZFN5(NM01 1419-
rl7347al)/ZFN6(NM01l4l9-l7353al) and Usp9y-ZFN3(NM148943-
al)/ZFN4(NM148943-l1836al) (10 ug each) and with an LTVEC targeting the Ch25h
gene (0.67 ug), to provide selection for cin resistance. Puromycin resistant colonies
were picked and screened for the deletion. The results are shown in Table 9.
Table 9: Screening Results for large Y chromosome deletion in 12778D-G5 and 12778E-G7
Parental Clone # of Puromycin- # of Colonies # Confirmed d
resistant Colonies Screened Clones
12778E-G7 638 384 8
2015/038001
Table 10 shows the exact sizes of the r than 500 kb deletions that were
precisely determined for one deletion clone (4306A-D5) derived from the E-G7 parental
clone (Table 3) and two deletion clones (4306E-C4 and 4306F-A12) derived from the D-G5
parental clone (Table 3).
Table 10: ZEN-mediated deletions of Kdm5d and Usp9y
Deletion Coordinates
Clone Size (bp)
on Y some
4306A-D5 -785404 534835
4306E-C4 520363-785402 535039
4306F—A12 250373-785404 535031
on of the Kdm5d, Eif2s3y, Uty, Ddx3y, and Usp9y genes ( was
confirmed in the deletion loss-of-allele assays and DNA sequencing as shown in
Clone 4306A-D5 produced nine XY female fully ES cell-derived VelociMice upon
microinjection into 8-cell stage embryos and transfer to surrogate mothers. None of the XY
females from clone 4306A-D5 were fertile.
Example 6: Large on on the Y Chromosome Mediated by CRISPR/Cas
A large deletion of the on the Y chromosome targeting the region n the
Kdm5d and the Usp9y genes was made utilizing CRISPR guide RNAs in combination with
Cas9 DNA endonuclease. gRNAs were designed to target the Kdm5d gene and the Usp9y
gene. The following gRNAs were designed to target Kdm5d: Kdm5dgA (Guide #1)
UUUGCCGAAUAUGCUCUCGU (SEQ ID NO:66); Kdm5ng (Guide #2)
UUGCCGAAUAUGCUCUCGUG (SEQ ID NO:67); and Kdm5dgC (Guide #5)
CGGGCAUCUCCAUACUCCCU (SEQ ID NO:68). The following gRNAs were designed to
target Usp9y: Usp9ygA (Guide #1) UAGCUCGUUGUGUAGCACCU (SEQ ID ;
Usp9ygB (Guide #1) UAUAGUUUCUUCGGGGUAAC (SEQ ID NO:70); and Usp9ng
(Guide #2) CCUUCUAUAGGCCC (SEQ ID NO:7l).
VGFl mouse ES cells were electroporated with 5 ug of a plasmid that expressed
Cas9 and 10 ug each of plasmids that expressed the Kdm5d gRNA B and Usp9y gRNA C
and with an LTVEC targeting the Ch25h gene (0.67 ug), to provide selection for puromycin
resistance. .
As illustrated in Kdm5ng (gRNA B) and Usp9ng (gRNA C) were used
to target the deletion of the Kdm5d and Usp9y genes. The resulting clones were ed for
deletion by loss-of-allele assays for sequences at the Kdm5d and Usp9y genes and the genes
in between (Eif2s3y, Uly, and Ddx3y) and for genes outside the targeted deletion (ny2 and
Sry). As shown in Table 11, four clones comprising the large deletion were obtained. Clone
R-AS produced seven XY male and 3 XY female fully ES cell-derived VelociMice upon
microinjection into 8-cell stage s and transfer to surrogate mothers.
Table 11: TaqMan assay confirming large on mediated by CRISPR guide
RNAs and Cas9
Loss-of—allele Copy Number Determination
Clone 19178TD 16697TD F12 Note
Zjfi/Z Sry
(1317253y)
Large
Q_F1 0 1
deletion
R-A8 o 1 Large
deletion
Large
R-C2 0 1 deletion,
partial loss Y
clone add as
R4511 1 1
WT l
All publications and patent applications ned in the specification are
indicative of the level of those skilled in the art to which this invention pertains. All
ations and patent applications are herein incorporated by reference to the same extent
as if each individual publication or patent application was specifically and individually
indicated to be incorporated by reference. If the information associated with a citation, such
as a deposit number changes with time, the version of the information in effect at the
effective filing date of the application is intended, the effective filing date meaning the actual
filing date or date of a priority application first providing the citation. Unless otherwise
apparent from the context of any embodiment, aspect, step or feature of the invention can be
used in ation with any other. nce to a range includes any integers within the
range, any subrange within the range. Reference to multiple ranges includes composites of
such ranges.
Claims (22)
1. A method for making a mouse XY embryonic stem (ES) cell line capable of producing a fertile XY female mouse in an F0 generation, sing: (a) modifying a mouse XY ES cell to comprise a c modification comprising a deletion that inactivates an endogenous Sry gene, n the mouse XY ES cell comprises a Y chromosome from a 129S6 strain; and (b) culturing the modified mouse XY ES cell under conditions to produce the mouse XY ES cell line e of producing the fertile XY female non-human mammal in the F0 generation.
2. The method of claim 1, wherein the genetic modification is generated by introducing into the mouse XY ES cell a nuclease agent that induces a nick or a double-strand break at a recognition site on the Y chromosome or a polynucleotide encoding the nuclease agent.
3. The method of claim 2, wherein the nuclease agent is a zinc finger se (ZFN), a Transcription Activator-Like Effector Nuclease (TALEN), a clease, or a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated (Cas) protein and a guide RNA (gRNA).
4. The method of claim 3, wherein the nuclease agent is the Cas protein and the gRNA, wherein the Cas protein is a Cas9 protein, and wherein the gRNA comprises: (a) a CRISPR RNA (crRNA) that targets the recognition site, wherein the recognition site is immediately flanked by a Protospacer Adjacent Motif (PAM) sequence; and (b) a activating CRISPR RNA (tracrRNA), optionally wherein the gRNA targets a sequence comprising SEQ ID NO: 10, SEQ ID NO: 11, or SEQ ID NO: 12.
5. The method of claim 3, n the se agent is the TALEN, optionally wherein the TALEN targets a sequence comprising SEQ ID NO: 72 and a sequence sing SEQ ID NO: 73.
6. The method of any one of claims 1-5, wherein the genetic modification is generated by introducing into the mouse XY ES cell a targeting vector comprising an insert polynucleotide flanked by first and second gy arms corresponding to first and second target sites at a target genomic locus on the Y chromosome, wherein the mouse XY ES cell is modified to comprise the insert polynucleotide at the target genomic locus.
7. The method of claim 6, wherein the first homology arm is from about 400- 1000 base pairs, and the second homology arm is from about 00 base pairs.
8. The method of claim 6, wherein the sum total of the first homology arm and the second gy arm is at least 10 kb.
9. The method of any one of claims 6-8, wherein the genetic modification is generated by introducing into the mouse XY ES cell: (i) a nuclease agent that induces the nick or a double-strand break at the recognition site on the Y chromosome or a polynucleotide encoding the nuclease agent; and (ii) the targeting vector comprising the insert polynucleotide flanked by first and second homology arms corresponding to first and second target sites at the target genomic locus on the Y chromosome, wherein the mouse XY ES cell is modified to comprise the insert polynucleotide at the target genomic locus.
10. The method of any one of claims 1-9, wherein the mouse XY ES cell is not cultured in a feminizing medium.
11. The method of any one of claims 1-9, wherein the culturing step comprises culturing the modified mouse XY ES cell in a medium comprising: (1) a base medium comprising about 3 mg/mL sodium chloride and about 2.2 mg/mL sodium bicarbonate and having an osmolality of about 218 mOsm/kg; and (2) supplements that maintain the modified mouse XY ES cell in culture.
12. The method of any one of claims 1-11, wherein upon introduction of the modified mouse XY ES cell into a mouse host embryo and following ion of the mouse host embryo to e F0 mice, at least 60% of the F0 mice are XY s which upon attaining sexual maturity are fertile.
13. A method for making a fertile XY female mouse in an F0 tion, comprising: (a) ucing a mouse XY embryonic stem (ES) cell into a mouse host embryo, wherein the mouse XY ES cell comprises a Y chromosome from a 129S6 strain and a genetic modification comprising a deletion that vates an endogenous mouse Sry gene, and ally wherein the mouse host embryo is a rula stage embryo; (b) gestating the mouse host embryo; and (c) obtaining F0 mice following gestation of the mouse host embryo, wherein at least 60% of the F0 mice are XY females which upon attaining sexual maturity are fertile.
14. The method of claim 13, wherein the F0 XY female mouse is fertile when crossed to a wild-type mouse, optionally wherein the wild-type mouse is a C57BL/6 mouse.
15. The method of any one of claims 1-14, wherein the mouse XY ES cell is isolated from a mouse that is a cross between a 129S6 strain and a C57BL/6 strain.
16. The method of claim 15, wherein the mouse XY ES cell is isolated from a hybrid embryo produced by ng a female C57BL/6NTac mouse to a male 129S6/SvEvTac mouse.
17. The method of claim 16, wherein the mouse XY ES cell is a VGF1 mouse ES cell.
18. The method of any one of claims 1-17, wherein the genetic modification further comprises an insertion of one or more nucleotides, a substitution of one or more nucleotides, or a combination thereof, optionally wherein: (I) the genetic modification further comprises a knockout; a knockin; a replacement of an endogenous nucleic acid sequence with a homologous, orthologous, or heterologous sequence; or a combination thereof; (II) the genetic modification further comprises an ion of a nucleic acid encoding a able marker and/or a nucleic acid ng a reporter gene operably linked to a promoter active in the mouse XY ES cell; or (III) the genetic modification further comprises an insertion of a nucleic acid encoding a reporter gene ly linked to an endogenous Sry promoter.
19. The method of any one of claims 1-18, further comprising modifying the mouse XY ES cell to comprise at least one additional ed genetic modification of a polynucleotide of interest.
20. The method of any one of claims 12-19, wherein upon introduction of the modified mouse XY ES cell into the mouse host embryo and following gestation of the mouse host embryo, at least 80% of the F0 mice are XY females which upon attaining sexual maturity are fertile, optionally wherein at least 90% of the F0 mice are XY females which upon attaining sexual maturity are fertile, and optionally wherein at least 95% of the F0 mice are XY females which upon attaining sexual maturity are fertile.
21. A cell produced by the method of any one of claims 1-12.
22. A mouse produced by the method of any one of claims 13-20. B * E F C E !LX" - + / Z` D DZ`EC, DSaO WP @96<$ SVN]MON NOTO\SWV B^]TT!((+" DZ`EC,!?83" 7 E F QC@2*!G8$*" 4K[0 MTOK^ON MTWVO[ - / + , DZ`EC+ E 7 DZ`EC( DXRT!0'" Z` D >EG64 \KZQO\ON MTWVO[ ' ' V%K% ' ( 0 DZ`EB( W_ B + L 8 5S[\KVMO PZWU 2 WP 2E8 !LX" )* +/ ('. ' ( , *,+! " E Z` 14 ? D % 14 % ( 9 7 DZ`EC) E D Z` * E B + QC@2)!G8$)" Z ` D D Z`E7 DZ`E7* " $( 8 DZ`EB) ( !G 2 @ C Q EKZQO\ DOY]OVMO !B2?" DZ`E7) ,c$442E822E842EEE2E88E8!E88"$* b E88E828288424228E!E88"$* b ,c$842284284E8882E84288!E88"$* c QC@2 G8$( G8$) G8$* QC@2+ QC@2, $ * 6 @ > E 2 =A?B ?2;5 ()../ >EG64 " .03$ !2 O X M W Z O O V ] V d Y [O X ZW Q 4 L S V F MW N R *,+! # D Z ` EKY?KV K[[K`[ \W [MZOOV PWZ QO KVN >A2 [\KZ\ \W [\WX MWNWV NOTO\SWV " .03$ Z X!2 [UKTTEG64 .' & ZO ) $ ) @ 6 4C;DBC QC@2[ $ ( 6 @ > W _ QC@2) QC@2* * E 2 L 8 ) ? 9 C < ( '-306+1 )0,.- */41- (4736 *,+! $ .')& VQ 8&##"% $# 6+01 54206-5 IY6 (#,''#''' JP`) IY5 IY4 * ) IY4 (#'''#''' J7@*&+ QC@2 ` IY4 ( #"!! -( *+.+2,0/ *,+! % F[X0` 5N_*` IY3 IY2 ) F\` ,''#''' 6SP)[*` E[X`$X[ ; IY2 ( J7@,&- QC@2 3$=NU,N FLK(` =NU,N MRZI !Y2(" MRZI1 JP`( ""%%"##"#"%#!%%%%$##%"%%$!##"!##$#%%$"$$"$%!%$%%$$"$""$%#%#"%%$$""!$""%#"$"%$" $"$$"$%%$%%$$"$""$%#%#"%%$$ !!!%!$$"$""$%#%#"%%$$ !!!!!!!!!%!$$"$""$%#%#"%$$$ !!!!!!!!!%!$##$""$%#%#"%%$# !!!!!!!!!%$$$"$""$%#%#"%%$$ *,+! &' *,+! &( %#"$"%"$$"%"%%%%"$"$%%%%!#"%"%%$%"%$$%%"%"$ %#"$"%"$$"%"%%%%"$"$%%%%!#"%"%%$%!!!!!!!!!! %#"$"%"$$$%"%%%%"$"$%%%%!#"%"%%$%!!!!!!!!!! %###"%"$$"%"%%%%"$"$%%#%!#"%"%%$#!!!!!!!!!! %%%%"$"$%%%%%#"%"%%$%!!!!!!!!!! *,+! &) =NU, FB KVN F[X`0 5AH@ ""%%"##"#"%#%%%%%$##%"%%$%##""!!!!!!!!!!!!!"%"#%$$"$""$%#%#"%%$#""#$""%#"$"%$" ""%%"##"#"%#!%%%%$##%"%%$"#"""!!!!!!!!!!!!!"%$%%$$"$""$%#%#"%$$$""!$""%#"""%%$ #%"%#"$"%"$$"%"%%%%"$"$%%!%%#"%"%%$%"%$$"$$"$%%$%%$$"$""$%#%#"%%$$""$""%#"$"%$"%$"$"%#%%""% #%"%#"$"%"$$"%"%%%%"$"$%%$%%!!!!!!!!!!!!!!!!!!!!!!$$"$""$%#%#"%%$$""$""%#"$"%$"%$"$"%#%%""% #%"%#"$"%"$$"%"%%%%"$"$%%$%%!!!!!!!!!!!!!!!!!!!!!!$$"$""$%#%#"%%$$""$""%#"$"%$"%$"$"%#%%""% #%"%#"#"%"$$"%"%%%%"$"$%%$%%!!!!!!!!!!!!!!!!!!!!!!$$#$""$%#%#"%%$$""$""%#"$"%$"%$"$"%#%%""% =NU, FB KVN FX[` 5AH@ (,''7 ($5, (,''7 ($5, ('''C ('''C ('''7 (,''C ('''7 =NU, FB KVN F[X` 5AH@ (,''7 ('''C CE LISTING <110> Frendewey, David Droguett, Gustavo Gagliardi, Anthony Kuno, Junko Auerbach, Wojtek Valenzuela, David M. <120> METHODS AND COMPOSITIONS FOR TARGETED GENETIC MODIFICATIONS AND METHODS OF USE <130> 057766-463545 <150> US ,582 <151> 201426 <150> US
Applications Claiming Priority (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201462017582P | 2014-06-26 | 2014-06-26 | |
US201462017627P | 2014-06-26 | 2014-06-26 | |
US62/017,627 | 2014-06-26 | ||
US62/017,582 | 2014-06-26 | ||
NZ728561A NZ728561B2 (en) | 2014-06-26 | 2015-06-26 | Methods and compositions for targeted genetic modifications and methods of use |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ765592A true NZ765592A (en) | 2021-02-26 |
NZ765592B2 NZ765592B2 (en) | 2021-05-27 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2021201239B2 (en) | Methods and compositions for targeted genetic modifications and methods of use | |
JP7101211B2 (en) | Methods and compositions of gene modification by targeting using a pair of guide RNAs | |
JP7095066B2 (en) | Methods and compositions for targeted gene modification through multiple targets in a single step | |
JP2020137521A (en) | Methods and compositions for modifying targeted locus | |
US11730150B2 (en) | Fibrillin-1 mutations for modeling neonatal progeroid syndrome with congenital lipodystrophy | |
NZ765592A (en) | Methods and compositions for targeted genetic modifications and methods of use | |
NZ765592B2 (en) | Methods and compositions for targeted genetic modifications and methods of use | |
NZ728561B2 (en) | Methods and compositions for targeted genetic modifications and methods of use | |
BR112016030147B1 (en) | METHOD FOR MODIFYING A GENOMIC TARGET LOCUS ON A Y CHROMOSOME IN A MOUSE EMBRYONIC STEM (ES) CELL |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PSEA | Patent sealed | ||
RENW | Renewal (renewal fees accepted) |
Free format text: PATENT RENEWED FOR 1 YEAR UNTIL 26 JUN 2023 BY ANAQUA SERVICES Effective date: 20220523 |
|
RENW | Renewal (renewal fees accepted) |
Free format text: PATENT RENEWED FOR 1 YEAR UNTIL 26 JUN 2024 BY ANAQUA SERVICES Effective date: 20230524 |
|
RENW | Renewal (renewal fees accepted) |
Free format text: PATENT RENEWED FOR 1 YEAR UNTIL 26 JUN 2025 BY ANAQUA SERVICES Effective date: 20240521 |