WO2014089290A1 - Crispr-based genome modification and regulation - Google Patents

Crispr-based genome modification and regulation Download PDF

Info

Publication number
WO2014089290A1
WO2014089290A1 PCT/US2013/073307 US2013073307W WO2014089290A1 WO 2014089290 A1 WO2014089290 A1 WO 2014089290A1 US 2013073307 W US2013073307 W US 2013073307W WO 2014089290 A1 WO2014089290 A1 WO 2014089290A1
Authority
WO
WIPO (PCT)
Prior art keywords
domain
cell
protein
sequence
fusion protein
Prior art date
Application number
PCT/US2013/073307
Other languages
French (fr)
Inventor
Fuqiang Chen
Gregory D. Davis
Qiaohua KANG
Scott W. KNIGHT
Original Assignee
Sigma-Aldrich Co. Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=50883989&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2014089290(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Sigma-Aldrich Co. Llc filed Critical Sigma-Aldrich Co. Llc
Priority to PL18160519T priority Critical patent/PL3363902T3/en
Priority to EP19201769.7A priority patent/EP3611263A1/en
Priority to AU2013355214A priority patent/AU2013355214B2/en
Priority to SG11201503824SA priority patent/SG11201503824SA/en
Priority to KR1020187001934A priority patent/KR102006880B1/en
Priority to PL13859964T priority patent/PL2928496T3/en
Priority to CN201810574719.4A priority patent/CN108913676B/en
Priority to DK13859964T priority patent/DK2928496T3/en
Priority to KR1020217011309A priority patent/KR102479178B1/en
Priority to EP13859964.2A priority patent/EP2928496B1/en
Priority to IL300199A priority patent/IL300199A/en
Priority to PL16183724T priority patent/PL3138911T3/en
Priority to KR1020237015558A priority patent/KR20230070065A/en
Priority to KR1020157013843A priority patent/KR101844123B1/en
Priority to PL18156734T priority patent/PL3360964T3/en
Priority to EP18156734.8A priority patent/EP3360964B1/en
Priority to EP16183724.0A priority patent/EP3138911B1/en
Priority to KR1020207023229A priority patent/KR102243092B1/en
Priority to BR112015012375A priority patent/BR112015012375A2/en
Priority to KR1020197022305A priority patent/KR102145760B1/en
Priority to CA2891347A priority patent/CA2891347C/en
Priority to JP2015545838A priority patent/JP6620018B2/en
Priority to KR1020227043854A priority patent/KR102531576B1/en
Priority to ES13859964T priority patent/ES2757325T3/en
Priority to US14/649,777 priority patent/US20160017366A1/en
Priority to EP16183725.7A priority patent/EP3138912B1/en
Priority to CN201380072477.4A priority patent/CN105142669B/en
Priority to EP19189913.7A priority patent/EP3617309A3/en
Priority to PL16183725T priority patent/PL3138912T3/en
Priority to EP18160519.7A priority patent/EP3363902B1/en
Publication of WO2014089290A1 publication Critical patent/WO2014089290A1/en
Priority to IL238856A priority patent/IL238856B/en
Priority to HK16106396.9A priority patent/HK1218389A1/en
Priority to US15/188,899 priority patent/US20160298125A1/en
Priority to US15/188,927 priority patent/US20160298136A1/en
Priority to US15/188,933 priority patent/US20160298138A1/en
Priority to US15/188,911 priority patent/US10731181B2/en
Priority to US15/188,909 priority patent/US20160298133A1/en
Priority to US15/188,931 priority patent/US20160298137A1/en
Priority to US15/188,924 priority patent/US10745716B2/en
Priority to US15/188,902 priority patent/US20160298132A1/en
Priority to US15/342,976 priority patent/US20170073705A1/en
Priority to US15/456,204 priority patent/US20170191082A1/en
Priority to AU2017204031A priority patent/AU2017204031B2/en
Priority to IL257178A priority patent/IL257178B/en
Priority to AU2018229489A priority patent/AU2018229489B2/en
Priority to AU2019201344A priority patent/AU2019201344C1/en
Priority to IL267598A priority patent/IL267598B/en
Priority to US16/654,613 priority patent/US20200140897A1/en
Priority to US16/943,792 priority patent/US20210079427A1/en
Priority to US16/943,767 priority patent/US20210207173A1/en
Priority to AU2020230243A priority patent/AU2020230243B2/en
Priority to AU2020230246A priority patent/AU2020230246B2/en
Priority to AU2020273316A priority patent/AU2020273316B2/en
Priority to US17/398,648 priority patent/US20210388396A1/en
Priority to AU2022200330A priority patent/AU2022200330B2/en
Priority to IL291129A priority patent/IL291129B2/en
Priority to AU2023216829A priority patent/AU2023216829A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/10Ophthalmic agents for accommodation disorders, e.g. myopia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/463Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from amphibians
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/21Endodeoxyribonucleases producing 5'-phosphomonoesters (3.1.21)
    • C12Y301/21004Type II site-specific deoxyribonuclease (3.1.21.4)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/09Fusion polypeptide containing a localisation/targetting motif containing a nuclear localisation signal
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/10Fusion polypeptide containing a localisation/targetting motif containing a tag for extracellular membrane crossing, e.g. TAT or VP22
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/80Fusion polypeptide containing a DNA binding domain, e.g. Lacl or Tet-repressor
    • C07K2319/81Fusion polypeptide containing a DNA binding domain, e.g. Lacl or Tet-repressor containing a Zn-finger domain for DNA binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3513Protein; Peptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/22Vectors comprising a coding region that has been codon optimised for expression in a respective host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present disclosure relates targeted genome modification.
  • the disclosure relates to RNA-guided endonucleases or fusion proteins comprising CRISPR/Cas-like protein and methods of using said proteins to modify or regulate targeted chromosomal sequences.
  • Targeted genome modification is a powerful tool for genetic manipulation of eukaryotic cells, embryos, and animals.
  • exogenous sequences can be integrated at targeted genomic locations and/or specific endogenous chromosomal sequences can be deleted, inactivated, or modified.
  • Current methods rely on the use of engineered nuclease enzymes, such as, for example, zinc finger nucleases (ZFNs) or transcription activator-like effector nucleases (TALENs).
  • ZFNs zinc finger nucleases
  • TALENs transcription activator-like effector nucleases
  • These chimeric nucleases contain programmable, sequence-specific DNA-binding modules linked to a nonspecific DNA cleavage domain.
  • Each new genomic target however, requires the design of a new ZFN or TALEN comprising a novel sequence-specific DNA-binding module.
  • these custom designed nucleases tend to be costly and time-consuming to prepare.
  • the specificities of ZFNs and TALENS are such
  • RNA-guided endonuclease wherein the endonuclease comprises at least one nuclear localization signal, at least one nuclease domain, and at least one domain that interacts with a guide RNA to target the endonuclease to a specific nucleotide sequence for cleavage.
  • the endonuclease can be derived from a Cas9 protein.
  • the endonuclease can be modified to lack at least one functional nuclease domain.
  • the endonuclease can further comprise a cell-penetrating domain, a marker domain, or both.
  • the endonuclease can be part of a protein-RNA complex comprising the guide RNA.
  • the guide RNA can be a single molecule comprising a 5' region that is complementary to a target site.
  • an isolated nucleic acid encoding any of the RNA-guided endonucleases disclosed herein.
  • the nucleic acid can be codon optimized for translation in mammalian cells, such as, for example, human cells.
  • the nucleic acid sequence encoding the RNA-guided endonuclease can be operably linked to a promoter control sequence, and optionally, can be part of a vector. In other
  • a vector comprising sequence encoding the RNA-guided endonuclease, which can be operably linked to a promoter control sequence can also comprise sequence encoding a guide RNA, which can be operably linked to a promoter control sequence.
  • Another aspect of the present invention encompasses a method for modifying a chromosomal sequence in a eukaryotic cell or embryo.
  • the method comprises introducing into a eukaryotic cell or embryo (i) at least one RNA-guided endonuclease comprising at least one nuclear localization signal or nucleic acid encoding at least one RNA-guided endonuclease as defined herein, (ii) at least one guide RNA or DNA encoding at least one guide RNA, and, optionally, (iii) at least one donor polynucleotide comprising a donor sequence.
  • the method further comprises culturing the cell or embryo such that each guide RNA directs a RNA-guided
  • the RNA-guided endonuclease can be derived from a Cas9 protein.
  • the nucleic acid encoding the RNA-guided endonuclease introduced into the cell or embryo can be mRNA. In a further embodiment, wherein the nucleic acid encoding the RNA-guided endonuclease introduced into the cell or embryo can be DNA.
  • the DNA encoding the RNA-guided endonuclease can be part of a vector that further comprises a sequence encoding the guide RNA.
  • the eukaryotic cell can be a human cell, a non-human mammalian cell, a stem cell, a non-mammalian vertebrate cell, an invertebrate cell, a plant cell, or a single cell eukaryotic organism.
  • the embryo is a non-human one cell animal embryo.
  • a further aspect of the disclosure provides a fusion protein comprising a CRISP R/Cas-like protein or fragment thereof and an effector domain.
  • the fusion protein comprises at least one nuclear localization signal.
  • the effector domain of the fusion protein can be a cleavage domain, an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain.
  • the CRISPR/Cas-like protein of the fusion protein can be derived from a Cas9 protein.
  • the Cas9 protein can be modified to lack at least one functional nuclease domain.
  • the Cas9 protein can be modified to lack all nuclease activity.
  • the effector domain can be a cleavage domain, such as, for example, a Fokl endonuclease domain or a modified Fokl endonuclease domain.
  • one fusion protein can form a dimer with another fusion protein.
  • the dimer can be a homodimer or a heterodimer.
  • the fusion protein can form a heterodimer with a zinc finger nuclease, wherein the cleavage domain of both the fusion protein and the zinc finger nucleases is a Fokl endonuclease domain or a modified Fokl endonuclease domain.
  • the fusion protein comprises a CRISPR/Cas-like protein derived from a Cas9 protein modified to lack all nuclease activity, and the effector domain is a Fokl endonuclease domain or a modified Fokl endonuclease domain.
  • the fusion protein comprises a CRISPR/Cas-like protein derived from a Cas9 protein modified to lack all nuclease activity, and the effector domain can be an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain.
  • any of the fusion proteins disclosed herein can comprise at least one additional domain chosen from a nuclear localization signal, a cell-penetrating domain, and a marker domain. Also provided are isolated nucleic acids encoding any of the fusion proteins provided herein.
  • Still another aspect of the disclosure encompasses a method for modifying a chromosomal sequence or regulating expression of a chromosomal sequence in a cell or embryo.
  • the method comprises introducing into the cell or embryo (a) at least one fusion protein or nucleic acid encoding at least one fusion protein, wherein the fusion protein comprises a CRISPR/Cas-like protein or a fragment thereof and an effector domain, and (b) at least one guide RNA or DNA encoding at least one guide RNA, wherein the guide RNA guides the CRISPR/Cas-like protein of the fusion protein to a targeted site in the chromosomal sequence and the effector domain of the fusion protein modifies the chromosomal sequence or regulates expression of the chromosomal sequence.
  • the CRISPR/Cas-like protein of the fusion protein can be derived from a Cas9 protein.
  • the CRISPR/Cas- like protein of the fusion protein can be modified to lack at least one functional nuclease domain.
  • the CRISPR/Cas-like protein of the fusion protein can be modified to lack all nuclease activity.
  • the method can comprise introducing into the cell or embryo one fusion protein or nucleic acid encoding one fusion protein and two guide RNAs or DNA encoding two guide RNAs, and wherein one double-stranded break is introduced in the chromosomal sequence.
  • the method can comprise introducing into the cell or embryo two fusion proteins or nucleic acid encoding two fusion proteins and two guide RNAs or DNA encoding two guide RNAs, and wherein two double-stranded breaks are introduced in the chromosomal sequence.
  • the method can comprise introducing into the cell or embryo one fusion protein or nucleic acid encoding one fusion protein, one guide RNA or nucleic acid encoding one guide RNA, and one zinc finger nuclease or nucleic acid encoding one zinc finger nuclease, wherein the zinc finger nuclease comprises a Fokl cleavage domain or a modified a Fokl cleavage domain, and wherein one double- stranded break is introduced into the chromosomal sequence.
  • the method can further comprise introducing into the cell or embryo at least one donor polynucleotide.
  • the fusion protein comprises an effector domain chosen from an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain
  • the fusion protein can comprise a Cas9 protein modified to lack all nuclease activity
  • the method can comprise introducing into the cell or embryo one fusion protein or nucleic acid encoding one fusion protein, and one guide RNA or nucleic acid encoding one guide RNA, and wherein the structure or expression of the targeted chromosomal sequence is modified.
  • the eukaryotic cell can be a human cell, a non-human mammalian cell, a stem cell, a non-mammalian vertebrate cell, an invertebrate cell, a plant cell, or a single cell eukaryotic organism.
  • the embryo is a non-human one cell animal embryo.
  • FIG. 1 diagrams genome modification using protein dimers.
  • A depicts a double stranded break created by a dimer composed of two fusion proteins, each of which comprises a Cas-like protein for DNA binding and a Fokl cleavage domain.
  • B depicts a double stranded break created by a dimer composed of a fusion protein comprising a Cas-like protein and a Fokl cleavage domain and a zinc finger nuclease comprising a zinc finger (ZF) DNA-binding domain and a Fokl cleavage domain.
  • ZF zinc finger
  • FIG. 2 illustrates regulation of gene expression using RNA-guided fusion proteins comprising gene regulatory domains.
  • A depicts a fusion protein comprising a Cas-like protein used for DNA binding and an "A/R" domain that activates or represses gene expression.
  • B diagrams a fusion protein comprising a Cas-like protein for DNA binding and a epigenetic modification domain ("Epi-mod') that affects epigenetic states by covalent modification of proximal DNA or proteins.
  • Epi-mod' epigenetic modification domain
  • (A) depicts a double stranded break created by two RNA-guided endonuclease that have been converted into nickases.
  • (B) depicts two double stranded breaks created by two RNA-guided endonuclease having endonuclease activity.
  • FIG. 4 presents fluorescence-activated cell sorting (FACS) of human K562 cells transfected with Cas9 nucleic acid, Cas9 guiding RNA, and AAVS1 - GFP DNA donor.
  • the Y axis represents the auto fluorescence intensity at a red channel, and the X axis represents the green fluorescence intensity.
  • A K562 cells transfected with 10 ⁇ g of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of pre-annealed crRNA-tracrRNA duplex, and 10 ⁇ g of AAVS1 -GFP plasmid DNA
  • B K562 cells transfected 10 ⁇ g of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of chimeric RNA, and 10 ⁇ g of AAVS1 -GFP plasmid DNA
  • C K562 cells transfected 10 ⁇ g of Cas9 mRNA that was capped by post-transcription capping reaction, 0.3 nmol of chimeric RNA, and 10 ⁇ g of AAVS1 -GFP plasmid DNA
  • D K562 cells transfected with 10 ⁇ g of Cas9 plasmid DNA, 5 ⁇ g of U6-chimeric RNA plasmid DNA, and 10 ⁇ g
  • FIG. 5 presents a junction PCR analysis documenting the targeted integration of GFP into the AAVS1 locus in human cells.
  • Lane M 1 kb DNA molecular markers
  • Lane A K562 cells transfected with 10 ⁇ g of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of pre-annealed crRNA-tracrRNA duplex, and 10 ⁇ g of AAVS1 -GFP plasmid DNA
  • Lane B K562 cells transfected 10 ⁇ g of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of chimeric RNA, and 10 ⁇ g of AAVS1 -GFP plasmid DNA
  • Lane C K562 cells transfected 10 ⁇ g of Cas9 mRNA that was capped by post-transcription capping reaction, 0.3 nmol of chimeric RNA, and 10 ⁇ g of AAVS1 -GFP plasmid DNA
  • Lane C
  • RNA-guided endonucleases which comprise at least one nuclear localization signal, at least one nuclease domain, and at least one domain that interacts with a guide RNA to target the endonuclease to a specific nucleotide sequence for cleavage.
  • nucleic acids encoding the RNA- guided endonucleases as well as methods of using the RNA-guided endonucleases to modify chromosomal sequences of eukaryotic cells or embryos.
  • the RNA-guided endonuclease interacts with specific guide RNAs, each of which directs the
  • RNA-guided endonuclease to a specific targeted site, at which site the RNA-guided endonuclease introduces a double-stranded break that can be repaired by a DNA repair process such that the chromosomal sequence is modified. Since the specificity is provided by the guide RNA, the RNA-based endonuclease is universal and can be used with different guide RNAs to target different genomic sequences. The methods disclosed herein can be used to target and modify specific chromosomal sequences and/or introduce exogenous sequences at targeted locations in the genome of cells or embryos.
  • the targeting is specific with limited off target effects.
  • a fusion protein comprises a CRISPR/Cas-like protein or fragment thereof and an effector domain.
  • Suitable effector domains include, without limit, cleavage domains, epigenetic modification domains, transcriptional activation domains, and transcriptional repressor domains.
  • Each fusion protein is guided to a specific chromosomal sequence by a specific guide RNA, wherein the effector domain mediates targeted genome
  • the fusion proteins can function as dimers thereby increasing the length of the target site and increasing the likelihood of its uniqueness in the genome (thus, reducing off target effects).
  • endogenous CRISPR systems modify genomic locations based on DNA binding word lengths of approximately 13-20 bp (Cong et al., Science, 339:819-823). At this word size, only 5- 7% of the target sites are unique within the genome (Iseli et al, PLos One 2(6):e579).
  • DNA binding word sizes for zinc finger nucleases typically range from 30-36 bp, resulting in target sites that are approximately 85-87% unique within the human genome.
  • the smaller sized DNA binding sites utilized by CRISPR-based systems limits and complicates design of targeted CRISP-based nucleases near desired locations, such as disease SNPs, small exons, start codons, and stop codons, as well as other locations within complex genomes.
  • the present disclosure not only provides means for expanding the CRISPR DNA binding word length (i.e., so as to limit off-target activity), but further provides CRISPR fusion proteins having modified functionality. According, the disclosed CRISPR fusion proteins have increased target specificity and unique functionality(ies). Also provided herein are methods of using the fusion proteins to modify or regulate expression of targeted chromosomal sequences.
  • RNA-guided endonucleases comprising at least one nuclear localization signal, which permits entry of the endonuclease into the nuclei of eukaryotic cells and embryos such as, for example, non-human one cell embryos.
  • RNA-guided endonucleases also comprise at least one nuclease domain and at least one domain that interacts with a guide RNA.
  • An RNA-guided endonuclease is directed to a specific nucleic acid sequence (or target site) by a guide RNA.
  • the guide RNA interacts with the RNA-guided endonuclease as well as the target site such that, once directed to the target site, the RNA-guided
  • the endonuclease is able to introduce a double-stranded break into the target site nucleic acid sequence. Since the guide RNA provides the specificity for the targeted cleavage, the endonuclease of the RNA-guided endonuclease is universal and can be used with different guide RNAs to cleave different target nucleic acid sequences.
  • RNA-guided endonucleases isolated nucleic acids (i.e., RNA or DNA) encoding the RNA-guided endonucleases, vectors comprising nucleic acids encoding the RNA-guided endonucleases, and protein-RNA complexes comprising the RNA- guided endonuclease plus a guide RNA.
  • isolated nucleic acids i.e., RNA or DNA
  • vectors comprising nucleic acids encoding the RNA-guided endonucleases
  • protein-RNA complexes comprising the RNA- guided endonuclease plus a guide RNA.
  • the RNA-guided endonuclease can be derived from a clustered regularly interspersed short p_alindromic repeats (CRISPR)/CRISPR-associated (Cas) system.
  • CRISPR clustered regularly interspersed short p_alindromic repeats
  • Cas CRISPR-associated
  • the CRISPR/Cas system can be a type I, a type II, or a type III system.
  • Non- limiting examples of suitable CRISPR/Cas proteins include Cas3, Cas4, Cas5, Cas5e (or CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8a1 , Cas8a2, Cas8b, Cas8c, Cas9, Cas10, Casl Od, CasF, CasG, CasH, Csy1 , Csy2, Csy3, Cse1 (or CasA), Cse2 (or CasB), Cse3 (or CasE), Cse4 (or CasC), Csc1 , Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1 , Cmr3, Cmr4, Cmr5, Cmr6, Csb1 , Csb2, Csb3,Csx17, Csx14, Csx10, Csx16, CsaX, Cs
  • the RNA-guided endonuclease is derived from a type II CRISPR/Cas system.
  • the RNA-guided endonuclease is derived from a Cas9 protein.
  • the Cas9 protein can be from Streptococcus pyogenes, Streptococcus thermophilus, Streptococcus sp., Nocardiopsis rougevillei,
  • Microscilla marina Burkholderiales bacterium, Polaromonas naphthalenivorans,
  • Polaromonas sp. Crocosphaera watsonii, Cyanothece sp., Microcystis aeruginosa, Synechococcus sp., Acetohalobium arabaticum, Ammonifex degensii,
  • Caldicieriosiruptor becscii Candidatus Desulforudis, Clostridium botulinum, Clostridium difficile, Finegoldia magna, Natranaerobius thermophilus, Pelotomaculum
  • thermopropionicum Acidithiobacillus caldus, Acidithiobacillus ferrooxidans,
  • chthonoplastes Oscillatoria sp., Petrotoga mobilis, Thermosipho africanus, or
  • CRISPR/Cas proteins comprise at least one RNA recognition and/or RNA binding domain. RNA recognition and/or RNA binding domains interact with guide RNAs. CRISPR/Cas proteins can also comprise nuclease domains (i.e., DNase or RNase domains), DNA binding domains, helicase domains, RNAse domains, protein-protein interaction domains, dimerization domains, as well as other domains.
  • the CRISPR/Cas-like protein can be a wild type CRISPR/Cas protein, a modified CRISPR/Cas protein, or a fragment of a wild type or modified
  • the CRISPR/Cas-like protein can be modified to increase nucleic acid binding affinity and/or specificity, alter an enzymatic activity, and/or change another property of the protein.
  • nuclease i.e., DNase, RNase
  • the CRISPR/Cas-like protein can be truncated to remove domains that are not essential for the function of the fusion protein.
  • the CRISPR/Cas-like protein can also be truncated or modified to optimize the activity of the effector domain of the fusion protein.
  • the CRISPR/Cas-like protein can be derived from a wild type Cas9 protein or fragment thereof. In other embodiments, the
  • CRISPR/Cas-like protein can be derived from modified Cas9 protein.
  • the amino acid sequence of the Cas9 protein can be modified to alter one or more properties (e.g., nuclease activity, affinity, stability, etc.) of the protein.
  • domains of the Cas9 protein not involved in RNA-guided cleavage can be eliminated from the protein such that the modified Cas9 protein is smaller than the wild type Cas9 protein.
  • a Cas9 protein comprises at least two nuclease (i.e., DNase) domains.
  • a Cas9 protein can comprise a RuvC-like nuclease domain and a HNH-like nuclease domain. The RuvC and HNH domains work together to cut single strands to make a double-stranded break in DNA. (Jinek et al., Science, 337: 816-821 ).
  • the Cas9-derived protein can be modified to contain only one functional nuclease domain (either a RuvC-like or a HNH-like nuclease domain).
  • the Cas9-derived protein can be modified such that one of the nuclease domains is deleted or mutated such that it is no longer functional (i.e., the nuclease activity is absent).
  • the Cas9-derived protein is able to introduce a nick into a double- stranded nucleic acid (such protein is termed a "nickase"), but not cleave the double- stranded DNA.
  • nickase a double- stranded nucleic acid
  • an aspartate to alanine (D10A) conversion in a RuvC-like domain converts the Cas9-derived protein into a nickase.
  • H840A or H839A a histidine to alanine (H840A or H839A) conversion in a HNH domain converts the Cas9-derived protein into a nickase.
  • Each nuclease domain can be modified using well-known methods, such as site-directed mutagenesis, PCR-mediated mutagenesis, and total gene synthesis, as well as other methods known in the art.
  • the RNA-guided endonuclease disclosed herein comprises at least one nuclear localization signal.
  • an NLS comprises a stretch of basic amino acids. Nuclear localization signals are known in the art (see, e.g., Lange et al., J. Biol. Chem., 2007, 282:5101 -5105).
  • the NLS can be a monopartite sequence, such as PKKKRKV (SEQ ID NO:1 ) or PKKKRRV (SEQ ID NO:2).
  • the NLS can be a bipartite sequence.
  • the NLS can be KRPAATKKAGQAKKKK (SEQ ID NO:3).
  • the NLS can be located at the N-terminus, the C-terminal, or in an internal location of the RNA- guided endonuclease.
  • the RNA-guided endonuclease can further comprise at least one cell-penetrating domain.
  • the cell-penetrating domain can be a cell-penetrating peptide sequence derived from the HIV-1 TAT protein.
  • the TAT cell-penetrating sequence can be
  • the cell- penetrating domain can be TLM (PLSSIFSRIGDPPKKKRKV; SEQ ID NO:5), a cell- penetrating peptide sequence derived from the human hepatitis B virus.
  • the cell-penetrating domain can be MPG
  • the cell-penetrating domain can be Pep-1
  • KETWWETWWTEWSQPKKKRKV SEQ ID NO:8
  • VP22 a cell penetrating peptide from Herpes simplex virus, or a polyarginine peptide sequence.
  • the cell-penetrating domain can be located at the N-terminus, the C-terminus, or in an internal location of the protein.
  • the RNA-guided endonuclease can also comprise at least one marker domain.
  • marker domains include fluorescent proteins, purification tags, and epitope tags.
  • the marker domain can be a fluorescent protein.
  • suitable 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 fluorescent proteins (e.g. EBFP, EBFP2, Azurite, mKalamal , GFPuv, Sapphire, T-sapphire,), cyan fluorescent proteins (e.g. ECFP, Cerulean, CyPet,
  • red fluorescent proteins mKate, mKate2, mPlum, DsRed monomer, mCherry, mRFP1 , DsRed-Express, DsRed2, DsRed-Monomer, HcRed- Tandem, HcRedl , AsRed2, eqFP61 1 , mRasberry, mStrawberry, Jred), and orange fluorescent proteins (mOrange, mKO, Kusabira-Orange, Monomeric Kusabira-Orange, mTangerine, tdTomato) or any other suitable fluorescent protein.
  • red fluorescent proteins mKate, mKate2, mPlum, DsRed monomer, mCherry, mRFP1 , DsRed-Express, DsRed2, DsRed-Monomer, HcRed- Tandem, HcRedl , AsRed2, eqFP61 1 , mRasberry, m
  • the marker domain can be a purification tag and/or an epitope tag.
  • Exemplary tags include, but are not limited to, glutathione-S-transferase (GST), chitin binding protein (CBP), maltose binding protein, thioredoxin (TRX), poly(NANP), tandem affinity purification (TAP) tag, myc, AcV5, AU1 , AU5, E, ECS, E2, FLAG, HA, nus, Softag 1 , Softag 3, Strep, SBP, Glu-Glu, HSV, KT3, S, S1 , T7, V5, VSV-G, 6xHis, biotin carboxyl carrier protein (BCCP), and calmodulin.
  • GST glutathione-S-transferase
  • CBP chitin binding protein
  • TRX thioredoxin
  • poly(NANP) poly(NANP)
  • TAP tandem affinity purification
  • the RNA-guided endonuclease may be part of a protein-RNA complex comprising a guide RNA.
  • the guide RNA interacts with the RNA-guided endonuclease to direct the endonuclease to a specific target site, wherein the 5' end of the guide RNA base pairs with a specific protospacer sequence.
  • a fusion protein comprising a CRISP R/Cas-like protein or fragment thereof and an effector domain.
  • the CRISP R/Cas-like protein is directed to a target site by a guide RNA, at which site the effector domain can modify or effect the targeted nucleic acid sequence.
  • the effector domain can be a cleavage domain, an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain.
  • the fusion protein can further comprise at least one additional domain chosen from a nuclear localization signal, a cell-penetrating domain, or a marker domain.
  • the fusion protein comprises a CRISPR/Cas-like protein or a fragment thereof.
  • CRISPR/Cas-like proteins are detailed above in section (I).
  • the CRISPR/Cas-like protein can be located at the N-terminus, the C-terminus, or in an internal location of the fusion protein
  • the CRISPR/Cas-like protein of the fusion protein can be derived from a Cas9 protein.
  • the Cas9-derived protein can be wild type, modified, or a fragment thereof.
  • the Cas9-derived protein can be modified to contain only one functional nuclease domain (either a RuvC-like or a HNH-like nuclease domain).
  • the Cas9-derived protein can be modified such that one of the nuclease domains is deleted or mutated such that it is no longer functional (i.e., the nuclease activity is absent).
  • the Cas9-derived protein is able to introduce a nick into a double-stranded nucleic acid (such protein is termed a "nickase"), but not cleave the double-stranded DNA.
  • a nickase such protein is termed a "nickase”
  • an aspartate to alanine (D10A) conversion in a RuvC-like domain converts the Cas9-derived protein into a nickase.
  • a histidine to alanine (H840A or H839A) conversion in a HNH domain converts the Cas9- derived protein into a nickase.
  • both of the RuvC-like nuclease domain and the HNH-like nuclease domain can be modified or eliminated such that the Cas9-derived protein is unable to nick or cleave double stranded nucleic acid.
  • all nuclease domains of the Cas9-derived protein can be modified or eliminated such that the Cas9-derived protein lacks all nuclease activity.
  • any or all of the nuclease domains can be inactivated by one or more deletion mutations, insertion mutations, and/or substitution mutations using well-known methods, such as site- directed mutagenesis, PCR-mediated mutagenesis, and total gene synthesis, as well as other methods known in the art.
  • the CRISPR/Cas-like protein of the fusion protein is derived from a Cas9 protein in which all the nuclease domains have been inactivated or deleted.
  • the fusion protein also comprises an effector domain.
  • the effector domain can be a cleavage domain, an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain.
  • the effector domain can be located at the N-terminus, the C-terminus, or in an internal location of the fusion protein.
  • the effector domain is a cleavage domain.
  • a "cleavage domain” refers to a domain that cleaves DNA.
  • the cleavage domain can be obtained from any endonuclease or exonuclease.
  • Non-limiting examples of endonucleases from which a cleavage domain can be derived include, but are not limited to, restriction endonucleases and homing endonucleases. See, for example, New England Biolabs Catalog or Belfort et al. (1997) Nucleic Acids Res.
  • the cleavage domain can be derived from a type ll-S endonuclease.
  • Type ll-S endonucleases cleave DNA at sites that are typically several base pairs away the recognition site and, as such, have separable recognition and cleavage domains. These enzymes generally are monomers that transiently associate to form dimers to cleave each strand of DNA at staggered locations.
  • suitable type ll-S endonucleases include Bfil, Bpml, Bsal, Bsgl, BsmBI, Bsml, BspMI, Fokl, Mboll, and Sapl.
  • the cleavage domain of the fusion protein is a Fokl cleavage domain or a derivative thereof.
  • the type ll-S cleavage can be modified to facilitate dimerization of two different cleavage domains (each of which is attached to a CRISP R/Cas-like protein or fragment thereof).
  • the cleavage domain of Fokl can be modified by mutating certain amino acid residues.
  • amino acid residues at positions 446, 447, 479, 483, 484, 486, 487, 490, 491 , 496, 498, 499, 500, 531 , 534, 537, and 538 of Fokl cleavage domains are targets for modification.
  • modified cleavage domains of Fokl that form obligate heterodimers include a pair in which a first modified cleavage domain includes mutations at amino acid positions 490 and 538 and a second modified cleavage domain that includes mutations at amino acid positions 486 and 499 (Miller et al., 2007, Nat. Biotechnol, 25:778-785; Szczpek et al., 2007, Nat. Biotechnol, 25:786-793).
  • modified Fokl cleavage domains can include three amino acid changes (Doyon et al. 201 1 , Nat. Methods, 8:74- 81 ).
  • one modified Fokl domain (which is termed ELD) can comprise Q486E, I499L, N496D mutations and the other modified Fokl domain (which is termed KKR) can comprise E490K, I538K, H537R mutations.
  • the effector domain of the fusion protein is a Fokl cleavage domain or a modified Fokl cleavage domain.
  • the Cas9-derived can be modified as discussed herein such that its endonuclease activity is eliminated.
  • the Cas9-derived can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
  • the effector domain of the fusion protein can be an epigenetic modification domain.
  • epigenetic modification domains alter histone structure and/or chromosomal structure without altering the DNA sequence. Changes histone and/or chromatin structure can lead to changes in gene expression.
  • epigenetic modification include, without limit, acetylation or methylation of lysine residues in histone proteins, and methylation of cytosine residues in DNA.
  • suitable epigenetic modification domains include histone
  • acetyltansferase domains histone deacetylase domains, histone methyltransferase domains, histone demethylase domains, DNA methyltransferase domains, and DNA demethylase domains.
  • the HAT domain can be derived from EP300 (i.e., E1 A binding protein p300), CREBBP (i.e., CREB-binding protein), CDY1 , CDY2, CDYL1 , CLOCK, ELP3, ESA1 , GCN5 (KAT2A), HAT1 ,KAT2B, KAT5, MYST1 , MYST2, MYST3, MYST4, NCOA1 , NCOA2, NCOA3, NCOAT, P/CAF, Tip60, TAFII250, or TF3C4.
  • the HAT domain is p300
  • the Cas9-derived can be modified as discussed herein such that its endonuclease activity is eliminated.
  • the Cas9-derived can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
  • the effector domain of the fusion protein can be a transcriptional activation domain.
  • a transcriptional activation domain interacts with transcriptional control elements and/or transcriptional regulatory proteins (i.e., transcription factors, RNA polymerases, etc.) to increase and/or activate
  • the transcriptional activation domain can be, without limit, a herpes simplex virus VP16 activation domain, VP64 (which is a tetrameric derivative of VP16), a N FKB p65 activation domain, p53 activation domains 1 and 2, a CREB (cAMP response element binding protein) activation domain, an E2A activation domain, and an NFAT (nuclear factor of activated T-cells) activation domain.
  • the transcriptional activation domain can be Gal4, Gcn4, MLL, Rtg3, Gln3, Oaf1 , Pip2, Pdr1 , Pdr3, Pho4, and Leu3.
  • the transcriptional activation domain may be wild type, or it may be a modified version of the original transcriptional activation domain.
  • the effector domain of the fusion protein is a VP16 or VP64 transcriptional activation domain.
  • the Cas9-derived protein can be modified as discussed herein such that its endonuclease activity is eliminated.
  • the Cas9-derived can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
  • the effector domain of the fusion protein can be a transcriptional repressor domain.
  • a transcriptional repressor domain interacts with transcriptional control elements and/or transcriptional regulatory proteins (i.e., transcription factors, RNA polymerases, etc.) to decrease and/or terminate transcription of a gene.
  • transcriptional repressor domains include inducible cAMP early repressor (ICER) domains, Kruppel-associated box A (KRAB-A) repressor domains, YY1 glycine rich repressor domains, Sp1 -like repressors, E(spl) repressors, ⁇ repressor, and MeCP2.
  • the Cas9-derived protein can be modified as discussed herein such that its endonuclease activity is eliminated.
  • the cas9 can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
  • the fusion protein further comprises at least one additional domain.
  • suitable additional domains include nuclear localization signals, cell-penetrating or translocation domains, and marker domains.
  • suitable nuclear localization signals, cell-penetrating domains, and marker domains are presented above in section (I).
  • a dimer comprising at least one fusion protein can form.
  • the dimer can be a homodimer or a heterodimer.
  • the heterodimer comprises two different fusion proteins.
  • the heterodimer comprises one fusion protein and an additional protein.
  • the dimer is a homodimer in which the two fusion protein monomers are identical with respect to the primary amino acid sequence.
  • the Cas9-derived proteins are modified such that their endonuclease activity is eliminated, i.e., such that they have no functional nuclease domains.
  • each fusion protein monomer comprises an identical Cas9 like protein and an identical cleavage domain.
  • the cleavage domain can be any cleavage domain, such as any of the exemplary cleavage domains provided herein.
  • the cleavage domain is a Fokl cleavage domain or a modified Fokl cleavage domain.
  • specific guide RNAs would direct the fusion protein monomers to different but closely adjacent sites such that, upon dimer formation, the nuclease domains of the two monomers would create a double stranded break in the target DNA.
  • the dimer is a heterodimer of two different fusion proteins.
  • the CRISP R/Cas-like protein of each fusion protein can be derived from a different CRISPR/Cas protein or from an orthologous CRISPR/Cas protein from a different bacterial species.
  • each fusion protein can comprise a Cas9-like protein, which Cas9-like protein is derived from a different bacterial species.
  • each fusion protein would recognize a different target site (i.e., specified by the protospacer and/or PAM sequence).
  • the guide RNAs could position the heterodimer to different but closely adjacent sites such that their nuclease domains results in an effective double stranded break in the target DNA.
  • the heterodimer can also have modified Cas9 proteins with nicking activity such that the nicking locations are different.
  • each fusion protein of a heterodimer can have different effector domains.
  • the effector domain is a cleavage domain
  • each fusion protein can contain a different modified cleavage domain.
  • each fusion protein can contain a different modified Fokl cleavage domain, as detailed above in section (ll)(b)(i).
  • the Cas-9 proteins can be modified such that their endonuclease activities are eliminated.
  • the two fusion proteins forming a heterodimer can differ in both the CRISPR/Cas-like protein domain and the effector domain.
  • the homodimer or heterodimer can comprise at least one additional domain chosen from nuclear localization signals (NLSs), cell-penetrating, translocation domains and marker domains, as detailed above.
  • NLSs nuclear localization signals
  • cell-penetrating cell-penetrating
  • translocation domains translocation domains
  • marker domains marker domains
  • one or both of the Cas9-derived proteins can be modified such that its endonuclease activity is eliminated or modified.
  • the heterodimer comprises one fusion protein and an additional protein.
  • the additional protein can be a nuclease.
  • the nuclease is a zinc finger nuclease.
  • a zinc finger nuclease comprises a zinc finger DNA binding domain and a cleavage domain.
  • a zinc finger recognizes and binds three (3) nucleotides.
  • a zinc finger DNA binding domain can comprise from about three zinc fingers to about seven zinc fingers.
  • the zinc finger DNA binding domain can be derived from a naturally occurring protein or it can be engineered. See, for example, Beieri et al. (2002) Nat. Biotechnol. 20:135-141 ; Pabo et al. (2001 ) Ann. Rev.
  • the cleavage domain of the zinc finger nuclease can be any cleavage domain detailed above in section (ll)(b)(i).
  • the cleavage domain of the zinc finger nuclease is a Fokl cleavage domain or a modified Fokl cleavage domain.
  • Such a zinc finger nuclease will dimerize with a fusion protein comprising a Fokl cleavage domain or a modified Fokl cleavage domain.
  • the zinc finger nuclease can comprise at least one additional domain chosen from nuclear localization signals, cell-penetrating or translocation domains, which are detailed above.
  • any of the fusion protein detailed above or a dimer comprising at least one fusion protein may be part of a protein-RNA complex comprising at least one guide RNA.
  • a guide RNA interacts with the CRISPR-CasOlike protein of the fusion protein to direct the fusion protein to a specific target site, wherein the 5' end of the guide RNA base pairs with a specific protospacer sequence.
  • nucleic acids encoding any of the RNA-guided endonucleases or fusion proteins described above in sections (I) and (II), respectively.
  • the nucleic acid can be RNA or DNA.
  • the nucleic acid encoding the RNA-guided endonuclease or fusion protein is mRNA.
  • the m RNA can be 5' capped and/or 3' polyadenylated.
  • the nucleic acid encoding the RNA-guided endonuclease or fusion protein is DNA.
  • the DNA can be present in a vector (see below).
  • the nucleic acid encoding the RNA-guided endonuclease or fusion protein can be codon optimized for efficient translation into protein in the eukaryotic cell or animal of interest.
  • codons can be optimized for expression in humans, mice, rats, hamsters, cows, pigs, cats, dogs, fish, amphibians, plants, yeast, insects, and so forth (see Codon Usage Database at www.kazusa.or.jp/codon/).
  • Programs for codon optimization are available as freeware (e.g., OPTIMIZER at
  • endonuclease or fusion protein can be operably linked to at least one promoter control sequence.
  • the DNA coding sequence can be operably linked to a promoter control sequence for expression in the eukaryotic cell or animal of interest.
  • the promoter control sequence can be constitutive, regulated, or tissue-specific.
  • Suitable constitutive promoter control sequences include, but are not limited to, cytomegalovirus immediate early promoter (CMV), simian virus (SV40) promoter, adenovirus major late promoter, Rous sarcoma virus (RSV) promoter, mouse mammary tumor virus (MMTV) promoter, phosphoglycerate kinase (PGK) promoter, elongation factor (EDI )-alpha promoter, ubiquitin promoters, actin promoters, tubulin promoters, immunoglobulin promoters, fragments thereof, or combinations of any of the foregoing.
  • suitable regulated promoter control sequences include without limit those regulated by heat shock, metals, steroids, antibiotics, or alcohol.
  • tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase-1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM- 2 promoter, INF- ⁇ promoter, Mb promoter, Nphsl promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter.
  • the promoter sequence can be wild type or it can be modified for more efficient or efficacious expression.
  • the encoding DNA can be operably linked to a CMV promoter for constitutive expression in mammalian cells.
  • the sequence encoding the RNA-guided endonuclease or fusion protein can be operably linked to a promoter sequence that is recognized by a phage RNA polymerase for in vitro mRNA synthesis.
  • the in w ro-tran scribed RNA can be purified for use in the methods detailed below in sections (IV) and (V).
  • the promoter sequence can be a T7, T3, or SP6 promoter sequence or a variation of a T7, T3, or SP6 promoter sequence.
  • the DNA encoding the fusion protein is operably linked to a T7 promoter for in vitro mRNA synthesis using T7 RNA polymerase.
  • the sequence encoding the RNA-guided endonuclease or fusion protein can be operably linked to a promoter sequence for in vitro expression of the RNA-guided endonuclease or fusion protein in bacterial or eukaryotic cells.
  • the expressed protein can be purified for use in the methods detailed below in sections (IV) and (V).
  • Suitable bacterial promoters include, without limit, T7 promoters, lac operon promoters, trp promoters, variations thereof, and combinations thereof.
  • An exemplary bacterial promoter is tac which is a hybrid of trp and lac promoters.
  • suitable eukaryotic promoters are listed above.
  • endonuclease or fusion protein also can be linked to a polyadenylation signal (e.g., SV40 polyA signal, bovine growth hormone (BGH) polyA signal, etc.) and/or at least one transcriptional termination sequence.
  • a polyadenylation signal e.g., SV40 polyA signal, bovine growth hormone (BGH) polyA signal, etc.
  • BGH bovine growth hormone
  • sequence encoding the RNA- guided endonuclease or fusion protein also can be linked to sequence encoding at least one nuclear localization signal, at least one cell-penetrating domain, and/or at least one marker domain, which are detailed above in section (I).
  • the DNA encoding the RNA-guided endonuclease or fusion protein can be present in a vector.
  • Suitable vectors include plasmid vectors, phagemids, cosmids, artificial/mini-chromosomes, transposons, and viral vectors (e.g., lentiviral vectors, adeno-associated viral vectors, etc.).
  • the DNA encoding the RNA-guided endonuclease or fusion protein is present in a plasmid vector.
  • suitable plasmid vectors include pUC, pBR322, pET, pBluescript, and variants thereof.
  • the vector can comprise additional expression control sequences (e.g., enhancer sequences, Kozak sequences, polyadenylation sequences, transcriptional termination sequences, etc.), selectable marker sequences (e.g., antibiotic resistance genes), origins of replication, and the like. Additional information can be found in “Current Protocols in Molecular Biology” Ausubel et al., John Wiley & Sons, New York, 2003 or "Molecular Cloning: A Laboratory Manual” Sambrook & Russell, Cold Spring Harbor Press, Cold Spring Harbor, NY, 3 rd edition, 2001 .
  • additional expression control sequences e.g., enhancer sequences, Kozak sequences, polyadenylation sequences, transcriptional termination sequences, etc.
  • selectable marker sequences e.g., antibiotic resistance genes
  • the expression vector comprising the sequence encoding the RNA-guided endonuclease or fusion protein can further comprise sequence encoding a guide RNA.
  • the sequence encoding the guide RNA generally is operably linked to at least one transcriptional control sequence for expression of the guide RNA in the cell or embryo of interest.
  • DNA encoding the guide RNA can be operably linked to a promoter sequence that is recognized by RNA polymerase III (Pol III).
  • Pol III RNA polymerase III
  • suitable Pol III promoters include, but are not limited to, mammalian U6, U3, H1 , and 7SL RNA promoters.
  • Another aspect of the present disclosure encompasses a method for modifying a chromosomal sequence in a eukaryotic cell or embryo.
  • the method comprises introducing into a eukaryotic cell or embryo (i) at least one RNA-guided endonuclease comprising at least one nuclear localization signal or nucleic acid encoding at least one RNA-guided endonuclease comprising at least one nuclear localization signal, (ii) at least one guide RNA or DNA encoding at least one guide RNA, and, optionally, (iii) at least one donor polynucleotide comprising a donor sequence.
  • the method further comprises culturing the cell or embryo such that each guide RNA directs an RNA-guided endonuclease to a targeted site in the chromosomal sequence where the RNA-guided endonuclease introduces a double-stranded break in the targeted site, and the double-stranded break is repaired by a DNA repair process such that the chromosomal sequence is modified.
  • the method can comprise introducing one RNA-guided endonuclease (or encoding nucleic acid) and one guide RNA (or encoding DNA) into a cell or embryo, wherein the RNA-guided endonuclease introduces one double-stranded break in the targeted chromosomal sequence.
  • the double-stranded break in the chromosomal sequence can be repaired by a non-homologous end-joining (NHEJ) repair process.
  • NHEJ non-homologous end-joining
  • the targeted chromosomal sequence can be modified or inactivated.
  • a single nucleotide change can give rise to an altered protein product, or a shift in the reading frame of a coding sequence can inactivate or "knock out" the sequence such that no protein product is made.
  • the optional donor can give rise to an altered protein product, or a shift in the reading frame of a coding sequence can inactivate or "knock out" the sequence such that no protein product is made.
  • the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence at the targeted site during repair of the double-stranded break.
  • the donor sequence in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted site in the chromosomal sequence, the donor sequence can be exchanged with or integrated into the chromosomal sequence at the targeted site during repair mediated by homology-directed repair process.
  • the donor sequence can be ligated directly with the cleaved chromosomal sequence by a non-homologous repair process during repair of the double-stranded break.
  • Exchange or integration of the donor sequence into the chromosomal sequence modifies the targeted chromosomal sequence or introduces an exogenous sequence into the chromosomal sequence of the cell or embryo.
  • the method can comprise introducing two RNA-guided endonucleases (or encoding nucleic acid) and two guide RNAs (or encoding DNA) into a cell or embryo, wherein the RNA-guided endonucleases introduce two double-stranded breaks in the chromosomal sequence. See FIG. 3B.
  • the two breaks can be within several base pairs, within tens of base pairs, or can be separated by many thousands of base pairs.
  • the resultant double-stranded breaks can be repaired by a non-homologous repair process such that the sequence between the two cleavage sites is lost and/or deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break(s).
  • the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded breaks by either a homology-based repair process (e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence) or a non-homologous repair process (e.g., in embodiments in which the donor sequence is flanked by compatible overhangs).
  • a homology-based repair process e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence
  • a non-homologous repair process e.g., in embodiments in which the donor sequence is flanked by compatible overhangs.
  • the method can comprise introducing one RNA-guided endonuclease modified to cleave one strand of a double-stranded sequence (or encoding nucleic acid) and two guide RNAs (or encoding DNA) into a cell or embryo, wherein each guide RNA directs the RNA-guided endonuclease to a specific target site, at which site the modified endonuclease cleaves one strand (i.e., nicks) of the double-stranded chromosomal sequence, and wherein the two nicks are in opposite stands and in close enough proximity to constitute a double-stranded break. See FIG. 3A.
  • the resultant double-stranded break can be repaired by a non-homologous repair process such that deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break.
  • the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded break by either a homology-based repair process (e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence) or a non-homologous repair process (e.g., in embodiments in which the donor sequence is flanked by compatible overhangs).
  • a homology-based repair process e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence
  • a non-homologous repair process e.g., in embodiments in which the donor sequence is flanked by compatible overhangs.
  • the method comprises introducing into a cell or embryo at least one RNA-guided endonuclease comprising at least one nuclear localization signal or nucleic acid encoding at least one RNA-guided endonuclease comprising at least one nuclear localization signal.
  • RNA-guided endonucleases and nucleic acids encoding RNA-guided endonucleases are described above in sections (I) and (III), respectively.
  • the RNA-guided endonuclease can be introduced into the cell or embryo as an isolated protein.
  • the RNA-guided endonuclease can further comprise at least one cell-penetrating domain, which facilitates cellular uptake of the protein.
  • the RNA-guided endonuclease can be introduced into the cell or embryo as an mRNA molecule.
  • the RNA-guided endonuclease can be introduced into the cell or embryo as a DNA molecule.
  • DNA sequence encoding the fusion protein is operably linked to a promoter sequence that will function in the cell or embryo of interest.
  • the DNA sequence can be linear, or the DNA sequence can be part of a vector.
  • the fusion protein can be introduced into the cell or embryo as an RNA-protein complex comprising the fusion protein and the guide RNA.
  • DNA encoding the RNA-guided endonuclease can further comprise sequence encoding a guide RNA.
  • each of the sequences encoding the RNA-guided endonuclease and the guide RNA is operably linked to appropriate promoter control sequence that allows expression of the RNA-guided endonuclease and the guide RNA, respectively, in the cell or embryo.
  • the DNA sequence encoding the RNA-guided endonuclease and the guide RNA can further comprise additional expression control, regulatory, and/or processing sequence(s).
  • the DNA sequence encoding the RNA-guided endonuclease and the guide RNA can be linear or can be part of a vector
  • the method also comprises introducing into a cell or embryo at least one guide RNA or DNA encoding at least one guide RNA.
  • a guide RNA interacts with the RNA-guided endonuclease to direct the endonuclease to a specific target site, at which site the 5' end of the guide RNA base pairs with a specific protospacer sequence in the chromosomal sequence.
  • Each guide RNA comprises three regions: a first region at the 5' end that is complementary to the target site in the chromosomal sequence, a second internal region that forms a stem loop structure, and a third 3' region that remains essentially single-stranded.
  • the first region of each guide RNA is different such that each guide RNA guides a fusion protein to a specific target site.
  • the second and third regions of each guide RNA can be the same in all guide RNAs.
  • the first region of the guide RNA is complementary to sequence (i.e., protospacer sequence) at the target site in the chromosomal sequence such that the first region of the guide RNA can base pair with the target site.
  • the first region of the guide RNA can comprise from about 10 nucleotides to more than about 25 nucleotides.
  • the region of base pairing between the first region of the guide RNA and the target site in the chromosomal sequence can be about 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 23, 24, 25, or more than 25 nucleotides in length.
  • the first region of the guide RNA is about 19, 20, or 21 nucleotides in length.
  • the guide RNA also comprises a second region that forms a secondary structure.
  • the secondary structure comprises a stem (or hairpin) and a loop.
  • the length of the loop and the stem can vary.
  • the loop can range from about 3 to about 10 nucleotides in length
  • the stem can range from about 6 to about 20 base pairs in length.
  • the stem can comprise one or more bulges of 1 to about 10 nucleotides.
  • the overall length of the second region can range from about 16 to about 60 nucleotides in length.
  • the loop is about 4 nucleotides in length and the stem comprises about 12 base pairs.
  • the guide RNA also comprises a third region at the 3' end that remains essentially single-stranded.
  • the third region has no complementarity to any chromosomal sequence in the cell of interest and has no complementarity to the rest of the guide RNA.
  • the length of the third region can vary. In general, the third region is more than about 4 nucleotides in length. For example, the length of the third region can range from about 5 to about 60 nucleotides in length.
  • the combined length of the second and third regions (also called the universal or scaffold region) of the guide RNA can range from about 30 to about 120 nucleotides in length. In one aspect, the combined length of the second and third regions of the guide RNA range from about 70 to about 100 nucleotides in length.
  • the guide RNA comprises a single molecule comprising all three regions.
  • the guide RNA can comprise two separate molecules.
  • the first RNA molecule can comprise the first region of the guide RNA and one half of the "stem" of the second region of the guide RNA.
  • the second RNA molecule can comprise the other half of the "stem” of the second region of the guide RNA and the third region of the guide RNA.
  • the first and second RNA molecules each contain a sequence of nucleotides that are complementary to one another.
  • the first and second RNA molecules each comprise a sequence (of about 6 to about 20 nucleotides) that base pairs to the other sequence to form a functional guide RNA.
  • the guide RNA can be introduced into the cell or embryo as a RNA molecule.
  • the RNA molecule can be transcribed in vitro.
  • the RNA molecule can be chemically synthesized.
  • the guide RNA can be introduced into the cell or embryo as a DNA molecule.
  • the DNA encoding the guide RNA can be operably linked to promoter control sequence for expression of the guide RNA in the cell or embryo of interest.
  • the RNA coding sequence can be operably linked to a promoter sequence that is recognized by RNA polymerase III (Pol III).
  • Suitable Pol III promoters include, but are not limited to, mammalian U6 or H1 promoters.
  • the RNA coding sequence is linked to a mouse or human U6 promoter.
  • the RNA coding sequence is linked to a mouse or human H1 promoter.
  • the DNA molecule encoding the guide RNA can be linear or circular.
  • the DNA sequence encoding the guide RNA can be part of a vector.
  • Suitable vectors include plasmid vectors, phagemids, cosmids, artificial/mini-chromosomes, transposons, and viral vectors.
  • the DNA encoding the RNA-guided endonuclease is present in a plasmid vector.
  • suitable plasmid vectors include pUC, pBR322, pET, pBluescript, and variants thereof.
  • the vector can comprise additional expression control sequences (e.g., enhancer sequences, Kozak sequences, polyadenylation sequences, transcriptional termination sequences, etc.), selectable marker sequences (e.g., antibiotic resistance genes), origins of replication, and the like.
  • each can be part of a separate molecule (e.g., one vector containing fusion protein coding sequence and a second vector containing guide RNA coding sequence) or both can be part of the same molecule (e.g., one vector containing coding (and regulatory) sequence for both the fusion protein and the guide RNA).
  • RNA-guided endonuclease in conjunction with a guide RNA is directed to a target site in the chromosomal sequence, wherein the RNA-guided endonuclease introduces a double-stranded break in the chromosomal sequence.
  • the target site has no sequence limitation except that the sequence is immediately followed (downstream) by a consensus sequence.
  • This consensus sequence is also known as a P/otospacer adjacent motif (PAM).
  • PAM P/otospacer adjacent motif
  • Examples of PAM include, but are not limited to, NGG, NGGNG, and NNAGAAW (wherein N is defined as any nucleotide and W is defined as either A or T).
  • the first region (at the 5' end) of the guide RNA is complementary to the protospacer of the target sequence.
  • the first region of the guide RNA is about 19 to 21 nucleotides in length.
  • the sequence of the target site in the chromosomal sequence is 5'-N 19 .2i-/VGG-3'.
  • the PAM is in italics.
  • the target site can be in the coding region of a gene, in an intron of a gene, in a control region of a gene, in a non-coding region between genes, etc.
  • the gene can be a protein coding gene or an RNA coding gene.
  • the gene can be any gene of interest.
  • the method further comprises introducing at least one donor polynucleotide into the embryo.
  • a donor polynucleotide comprises at least one donor sequence.
  • polynucleotide corresponds to an endogenous or native chromosomal sequence.
  • the donor sequence can be essentially identical to a portion of the
  • the donor sequence can comprise a modified version of the wild type sequence at the targeted site such that, upon integration or exchange with the native sequence, the sequence at the targeted chromosomal location comprises at least one nucleotide change.
  • the change can be an insertion of one or more nucleotides, a deletion of one or more nucleotides, a substitution of one or more nucleotides, or combinations thereof.
  • the cell or embryo/animal can produce a modified gene product from the targeted chromosomal sequence.
  • the donor sequence of the donor polynucleotide corresponds to an exogenous sequence.
  • an "exogenous" sequence refers to a sequence that is not native to the cell or embryo, or a sequence whose native location in the genome of the cell or embryo is in a different location.
  • the exogenous sequence can comprise protein coding sequence, which can be operably linked to an exogenous promoter control sequence such that, upon integration into the genome, the cell or embryo/animal is able to express the protein coded by the integrated sequence.
  • the exogenous sequence can be integrated into the chromosomal sequence such that its expression is regulated by an endogenous promoter control sequence.
  • the exogenous sequence can be a transcriptional control sequence, another expression control sequence, an RNA coding sequence, and so forth. Integration of an exogenous sequence into a chromosomal sequence is termed a "knock in.”
  • the length of the donor sequence can and will vary.
  • the donor sequence can vary in length from several nucleotides to hundreds of nucleotides to hundreds of thousands of nucleotides.
  • Donor polynucleotide comprising upstream and downstream sequences.
  • the donor sequence in the donor polynucleotide is flanked by an upstream sequence and a downstream sequence, which have substantial sequence identity to sequences located upstream and downstream, respectively, of the targeted site in the chromosomal sequence. Because of these sequence similarities, the upstream and downstream sequences of the donor polynucleotide permit
  • the upstream sequence refers to a nucleic acid sequence that shares substantial sequence identity with a chromosomal sequence upstream of the targeted site.
  • the downstream sequence refers to a nucleic acid sequence that shares substantial sequence identity with a chromosomal sequence downstream of the targeted site.
  • substantially sequence identity refers to sequences having at least about 75% sequence identity.
  • the upstream and downstream sequences in the donor polynucleotide can have about 75%, 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with sequence upstream or downstream to the targeted site.
  • the upstream and downstream sequences in the donor polynucleotide can have about 95% or 100% sequence identity with chromosomal sequences upstream or downstream to the targeted site.
  • the upstream sequence shares substantial sequence identity with a chromosomal sequence located immediately upstream of the targeted site (i.e., adjacent to the targeted site). In other embodiments, the upstream sequence shares substantial sequence identity with a chromosomal sequence that is located within about one hundred (100) nucleotides upstream from the targeted site. Thus, for example, the upstream sequence can share substantial sequence identity with a chromosomal sequence that is located about 1 to about 20, about 21 to about 40, about 41 to about 60, about 61 to about 80, or about 81 to about 100 nucleotides upstream from the targeted site. In one embodiment, the downstream sequence shares substantial sequence identity with a chromosomal sequence located immediately downstream of the targeted site (i.e., adjacent to the targeted site). In other
  • the downstream sequence shares substantial sequence identity with a chromosomal sequence that is located within about one hundred (100) nucleotides downstream from the targeted site.
  • the downstream sequence can share substantial sequence identity with a chromosomal sequence that is located about 1 to about 20, about 21 to about 40, about 41 to about 60, about 61 to about 80, or about 81 to about 100 nucleotides downstream from the targeted site.
  • Each upstream or downstream sequence can range in length from about 20 nucleotides to about 5000 nucleotides.
  • upstream and downstream sequences can comprise about 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1 100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2800, 3000, 3200, 3400, 3600, 3800, 4000, 4200, 4400, 4600, 4800, or 5000 nucleotides.
  • upstream and downstream sequences can comprise about 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1 100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2800, 3000, 3200, 3400, 3600, 3800, 4000, 4200, 4400, 4600,
  • downstream sequences can range in length from about 50 to about 1500 nucleotides.
  • Donor polynucleotides comprising the upstream and downstream sequences with sequence similarity to the targeted chromosomal sequence can be linear or circular.
  • the donor polynucleotide can be part of a vector.
  • the vector can be a plasmid vector.
  • Donor polynucleotide comprising targeted cleavage site(s).
  • the donor polynucleotide can additionally comprise at least one targeted cleavage site that is recognized by the RNA-guided endonuclease.
  • the targeted cleavage site added to the donor polynucleotide can be placed upstream or
  • the donor sequence can be flanked by targeted cleavage sites such that, upon cleavage by the RNA-guided endonuclease, the donor sequence is flanked by overhangs that are compatible with those in the chromosomal sequence generated upon cleavage by the RNA-guided endonuclease. Accordingly, the donor sequence can be ligated with the cleaved chromosomal sequence during repair of the double stranded break by a nonhomologous repair process.
  • donor polynucleotides comprising the targeted cleavage site(s) will be circular (e.g., can be part of a plasmid vector).
  • Donor polynucleotide comprising a short donor sequence with optional overhangs.
  • the donor polynucleotide can be a linear molecule comprising a short donor sequence with optional short overhangs that are compatible with the overhangs generated by the RNA-guided endonuclease.
  • the donor sequence can be ligated directly with the cleaved chromosomal sequence during repair of the double-stranded break.
  • the donor sequence can be less than about 1 ,000, less than about 500, less than about 250, or less than about 100 nucleotides.
  • the donor polynucleotide can be a linear molecule comprising a short donor sequence with blunt ends. In other iterations, the donor polynucleotide can be a linear molecule comprising a short donor sequence with 5' and/or 3' overhangs. The overhangs can comprise 1 , 2, 3, 4, or 5 nucleotides.
  • the donor polynucleotide will be DNA.
  • the DNA may be single-stranded or double-stranded and/or linear or circular.
  • the donor polynucleotide may be a DNA plasmid, a bacterial artificial chromosome (BAC), a yeast artificial chromosome (YAC), a viral vector, a linear piece of DNA, a PCR fragment, a naked nucleic acid, or a nucleic acid complexed with a delivery vehicle such as a liposome or poloxamer.
  • the donor polynucleotide comprising the donor sequence can be part of a plasmid vector. In any of these situations, the donor polynucleotide comprising the donor sequence can further comprise at least one additional sequence.
  • RNA-targeted endonuclease(s) (or encoding nucleic acid), the guide RNA(s) (or encoding DNA), and the optional donor polynucleotide(s) can be introduced into a cell or embryo by a variety of means.
  • the cell or embryo is transfected. Suitable transfection methods include calcium phosphate- mediated transfection, nucleofection (or electroporation), cationic polymer transfection (e.g., DEAE-dextran or polyethylenimine), viral transduction, virosome transfection, virion transfection, liposome transfection, cationic liposome transfection,
  • the embryo is a fertilized one-cell stage embryo of the species of interest.
  • the molecules can be injected into the pronuclei of one cell embryos.
  • polynucleotide(s) can be introduced into the cell or embryo simultaneously or
  • RNA-targeted endonuclease(s) or encoding nucleic acid
  • guide RNA(s) or encoding DNA
  • DNA encoding an RNA- targeted endonuclease and DNA encoding a guide RNA are delivered together within the plasmid vector.
  • the method further comprises maintaining the cell or embryo under appropriate conditions such that the guide RNA(s) directs the RNA-guided
  • RNA- guided endonuclease(s) introduce at least one double-stranded break in the
  • a double-stranded break can be repaired by a DNA repair process such that the chromosomal sequence is modified by a deletion of at least one nucleotide, an insertion of at least one nucleotide, a substitution of at least one nucleotide, or a combination thereof.
  • the double-stranded break can be repaired via a non-homologous end-joining (NHEJ) repair process.
  • NHEJ non-homologous end-joining
  • sequence at the chromosomal sequence can be modified such that the reading frame of a coding region can be shifted and that the chromosomal sequence is inactivated or "knocked out.”
  • An inactivated protein-coding chromosomal sequence does not give rise to the protein coded by the wild type chromosomal sequence.
  • the double- stranded break can be repaired by a homology-directed repair (HDR) process such that the donor sequence is integrated into the chromosomal sequence.
  • HDR homology-directed repair
  • an exogenous sequence can be integrated into the genome of the cell or embryo, or the targeted chromosomal sequence can be modified by exchange of a modified sequence for the wild type chromosomal sequence.
  • the RNA-guided endonuclease can cleave both the targeted chromosomal sequence and the donor polynucleotide.
  • the linearized donor polynucleotide can be integrated into the chromosomal sequence at the site of the double-stranded break by ligation between the donor polynucleotide and the cleaved chromosomal sequence via a NHEJ process.
  • the short donor sequence can be integrated into the chromosomal sequence at the site of the double-stranded break via a NHEJ process.
  • the integration can proceed via the ligation of blunt ends between the short donor sequence and the chromosomal sequence at the site of the double stranded break.
  • the integration can proceed via the ligation of sticky ends (i.e., having 5' or 3' overhangs) between a short donor sequence that is flanked by overhangs that are compatible with those generated by the RNA-targeting endonuclease in the cleaved chromosomal sequence.
  • the cell is maintained under conditions appropriate for cell growth and/or maintenance. Suitable cell culture conditions are well known in the art and are described, for example, in Santiago et al. (2008) PNAS 105:5809-5814; Moehle et al. (2007) PNAS 104:3055-3060; Urnov et al. (2005) Nature 435:646-651 ; and Lombardo et al (2007) Nat. Biotechnology 25:1298-1306. Those of skill in the art appreciate that methods for culturing cells are known in the art and can and will vary depending on the cell type. Routine optimization may be used, in all cases, to determine the best techniques for a particular cell type.
  • An embryo can be cultured in vitro (e.g., in cell culture). Typically, the embryo is cultured at an appropriate temperature and in appropriate media with the necessary 0 2 /C0 2 ratio to allow the expression of the RNA endonuclease and guide RNA, if necessary. Suitable non-limiting examples of media include M2, M16, KSOM, BMOC, and HTF media. A skilled artisan will appreciate that culture conditions can and will vary depending on the species of embryo. Routine optimization may be used, in all cases, to determine the best culture conditions for a particular species of embryo. In some cases, a cell line may be derived from an in vitro-cultured embryo (e.g., an embryonic stem cell line).
  • an embryo may be cultured in vivo by transferring the embryo into the uterus of a female host.
  • the female host is from the same or similar species as the embryo.
  • the female host is pseudo-pregnant.
  • Methods of preparing pseudo-pregnant female hosts are known in the art.
  • methods of transferring an embryo into a female host are known. Culturing an embryo in vivo permits the embryo to develop and can result in a live birth of an animal derived from the embryo. Such an animal would comprise the modified chromosomal sequence in every cell of the body.
  • the cell can be a human cell, a non-human mammalian cell, a non-mammalian vertebrate cell, an invertebrate cell, an insect cell, a plant cell, a yeast cell, or a single cell eukaryotic organism.
  • the embryo is non-human mammalian embryo.
  • the embryos can be a one cell non- human mammalian embryo.
  • Exemplary mammalian embryos, including one cell embryos include without limit mouse, rat, hamster, rodent, rabbit, feline, canine, ovine, porcine, bovine, equine, and primate embryos.
  • the cell can be a stem cell.
  • Suitable stem cells include without limit embryonic stem cells, ES-like stem cells, fetal stem cells, adult stem cells, pluripotent stem cells, induced pluripotent stem cells, multipotent stem cells, oligopotent stem cells, unipotent stem cells and others.
  • the cell is a mammalian cell.
  • Non-limiting examples of suitable mammalian cells include Chinese hamster ovary (CHO) cells, baby hamster kidney (BHK) cells; mouse myeloma NS0 cells, mouse embryonic fibroblast 3T3 cells (NIH3T3), mouse B lymphoma A20 cells; mouse melanoma B16 cells; mouse myoblast C2C12 cells; mouse myeloma SP2/0 cells; mouse embryonic mesenchymal C3H-10T1 /2 cells; mouse carcinoma CT26 cells, mouse prostate DuCuP cells; mouse breast EMT6 cells; mouse hepatoma Hepa1 c1 c7 cells; mouse myeloma J5582 cells; mouse epithelial MTD-1 A cells; mouse myocardial MyEnd cells; mouse renal RenCa cells; mouse pancreatic RIN-5F cells; mouse melanoma X64 cells; mouse lymphoma YAC-1 cells; rat glioblastoma 9L cells; rat B lymph
  • Another aspect of the present disclosure encompasses a method for modifying a chromosomal sequence or regulating expression of a chromosomal sequence in a cell or embryo.
  • the method comprises introducing into the cell or embryo (a) at least one fusion protein or nucleic acid encoding at least one fusion protein, wherein the fusion protein comprises a CRISPR/Cas-like protein or a fragment thereof and an effector domain, and (b) at least one guide RNA or DNA encoding the guide RNA, wherein the guide RNA guides the CRISPR/Cas-like protein of the fusion protein to a targeted site in the chromosomal sequence and the effector domain of the fusion protein modifies the chromosomal sequence or regulates expression of the chromosomal sequence.
  • Fusion proteins comprising a CRISPR/Cas-like protein or a fragment thereof and an effector domain are detailed above in section (II).
  • the fusion proteins disclosed herein further comprise at least one nuclear localization signal.
  • Nucleic acids encoding fusion proteins are described above in section (III).
  • the fusion protein can be introduced into the cell or embryo as an isolated protein (which can further comprise a cell-penetrating domain).
  • the isolated fusion protein can be part of a protein-RNA complex comprising the guide RNA.
  • the fusion protein can be introduced into the cell or embryo as a RNA molecule (which can be capped and/or polyadenylated).
  • the fusion protein can be introduced into the cell or embryo as a DNA molecule.
  • the fusion protein and the guide RNA can be introduced into the cell or embryo as discrete DNA molecules or as part of the same DNA molecule.
  • DNA molecules can be plasmid vectors.
  • the method further comprises introducing into the cell or embryo at least one zinc finger nuclease.
  • Zinc finger nucleases are described above in section (ll)(d).
  • the method further comprises introducing into the cell or embryo at least one donor polynucleotide. Donor polynucleotides are detailed above in section (IV)(d). Means for introducing molecules into cells or embryos, as well as means for culturing cell or embryos are described above in sections (IV)(e) and (IV)(f), respectively. Suitable cells and embryos are described above in section (IV)(g).
  • the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein) and two guide RNAs (or DNA encoding two guide RNAs).
  • the two guide RNAs direct the fusion protein to two different target sites in the chromosomal sequence, wherein the fusion protein dimerizes (e.g., form a homodimer) such that the two cleavage domains can introduce a double stranded break into the chromosomal sequence. See FIG. 1A.
  • the double-stranded break in the chromosomal sequence can be repaired by a non-homologous end-joining (NHEJ) repair process.
  • NHEJ non-homologous end-joining
  • a single nucleotide change can give rise to an altered protein product, or a shift in the reading frame of a coding sequence can inactivate or "knock out" the sequence such that no protein product is made.
  • the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence at the targeted site during repair of the double-stranded break.
  • the donor sequence can be exchanged with or integrated into the
  • the donor sequence can be ligated directly with the cleaved chromosomal sequence by a non-homologous repair process during repair of the double-stranded break.
  • Exchange or integration of the donor sequence into the chromosomal sequence modifies the targeted chromosomal sequence or introduces an exogenous sequence into the chromosomal sequence of the cell or embryo.
  • the method can comprise introducing into the cell or embryo two different fusion proteins (or nucleic acid encoding two different fusion proteins) and two guide RNAs (or DNA encoding two guide RNAs).
  • the fusion proteins can differ as detailed above in section (II).
  • Each guide RNA directs a fusion protein to a specific target site in the chromosomal sequence, wherein the fusion proteins dimerize (e.g., form a heterodimer) such that the two cleavage domains can introduce a double stranded break into the chromosomal sequence.
  • the resultant double-stranded breaks can be repaired by a non-homologous repair process such that deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break.
  • the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded break by either a homology-based repair process (e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence) or a nonhomologous repair process (e.g., in embodiments in which the donor sequence is flanked by compatible overhangs).
  • a homology-based repair process e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence
  • nonhomologous repair process e.g., in embodiments in which the donor sequence is flanked by compatible overhangs.
  • the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein), one guide RNA (or DNA encoding one guide RNA), and one zinc finger nuclease (or nucleic acid encoding the zinc finger nuclease), wherein the zinc finger nuclease comprises a Fokl cleavage domain or a modified Fokl cleavage domain.
  • the guide RNA directs the fusion protein to a specific chromosomal sequence, and the zinc finger nuclease is directed to another
  • the fusion protein and the zinc finger nuclease dimerize such that the cleavage domain of the fusion protein and the cleavage domain of the zinc finger nuclease can introduce a double stranded break into the chromosomal sequence. See FIG. 1 B.
  • the resultant double-stranded breaks can be repaired by a non-homologous repair process such that deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break.
  • the optional donor polynucleotide is not present
  • the resultant double-stranded breaks can be repaired by a non-homologous repair process such that deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break.
  • the optional donor polynucleotide such that deletions of
  • the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded break by either a homology-based repair process (e.g., in
  • the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein) and one guide RNA (or DNA encoding one guide RNA).
  • the guide RNA directs the fusion protein to a specific chromosomal sequence, wherein the transcriptional activation domain or a transcriptional repressor domain activates or represses expression, respectively, of the targeted chromosomal sequence. See FIG. 2A.
  • the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein) and one guide RNA (or DNA encoding one guide RNA).
  • the guide RNA directs the fusion protein to a specific chromosomal sequence, wherein the epigenetic modification domain modifies the structure of the targeted the chromosomal sequence. See FIG. 2A.
  • Epigenetic modifications include acetylation, methylation of histone proteins and/or nucleotide methylation.
  • structural modification of the chromosomal sequence leads to changes in expression of the chromosomal sequence.
  • the present disclosure encompasses genetically modified cells, non-human embryos, and non-human animals comprising at least one chromosomal sequence that has been modified using an RNA-guided endonuclease-mediated or fusion protein-mediated process, for example, using the methods described herein.
  • the disclosure provides cells comprising at least one DNA or RNA molecule encoding an RNA-guided endonuclease or fusion protein targeted to a chromosomal sequence of interest or a fusion protein, at least one guide RNA, and optionally one or more donor polynucleotide(s).
  • the disclosure also provides non-human embryos comprising at least one DNA or RNA molecule encoding an RNA-guided endonuclease or fusion protein targeted to a chromosomal sequence of interest, at least one guide RNA, and optionally one or more donor polynucleotide(s).
  • the present disclosure provides genetically modified non-human animals, non-human embryos, or animal cells comprising at least one modified chromosomal sequence.
  • the modified chromosomal sequence may be modified such that it is (1 ) inactivated, (2) has an altered expression or produces an altered protein product, or (3) comprises an integrated sequence.
  • the chromosomal sequence is modified with an RNA guided endonuclease-mediated or fusion protein-mediated process, using the methods described herein.
  • one aspect of the present disclosure provides a genetically modified animal in which at least one chromosomal sequence has been modified.
  • the genetically modified animal comprises at least one inactivated chromosomal sequence.
  • the modified chromosomal sequence may be inactivated such that the sequence is not transcribed and/or a functional protein product is not produced.
  • a genetically modified animal comprising an inactivated chromosomal sequence may be termed a "knock out” or a "conditional knock out.”
  • the inactivated chromosomal sequence can include a deletion mutation (i.e., deletion of one or more nucleotides), an insertion mutation (i.e., insertion of one or more nucleotides), or a nonsense mutation (i.e., substitution of a single nucleotide for another nucleotide such that a stop codon is introduced).
  • a deletion mutation i.e., deletion of one or more nucleotides
  • an insertion mutation i.e., insertion of one or more nucleotides
  • a nonsense mutation i.e., substitution of a single nucleotide for another nucleotide such that a stop codon is introduced.
  • the inactivated chromosomal sequence comprises no exogenously introduced sequence.
  • genetically modified animals in which two, three, four, five
  • the modified chromosomal sequence can be altered such that it codes for a variant protein product.
  • a genetically modified animal comprising a modified chromosomal sequence can comprise a targeted point mutation(s) or other modification such that an altered protein product is produced.
  • the chromosomal sequence can be modified such that at least one nucleotide is changed and the expressed protein comprises one changed amino acid residue (missense mutation).
  • the chromosomal sequence can be modified to comprise more than one missense mutation such that more than one amino acid is changed.
  • the chromosomal sequence can be modified to have a three nucleotide deletion or insertion such that the expressed protein comprises a single amino acid deletion or insertion.
  • the altered or variant protein can have altered properties or activities compared to the wild type protein, such as altered substrate specificity, altered enzyme activity, altered kinetic rates, etc.
  • the genetically modified animal can comprise at least one chromosomally integrated sequence.
  • a genetically modified animal comprising an integrated sequence may be termed a "knock in” or a
  • the chromosomally integrated sequence can, for example, encode an orthologous protein, an endogenous protein, or combinations of both.
  • a sequence encoding an orthologous protein or an endogenous protein can be integrated into a chromosomal sequence encoding a protein such that the chromosomal sequence is inactivated, but the exogenous sequence is expressed.
  • the sequence encoding the orthologous protein or endogenous protein may be operably linked to a promoter control sequence.
  • a sequence encoding an orthologous protein or an endogenous protein may be integrated into a chromosomal sequence without affecting expression of a chromosomal sequence.
  • a sequence encoding a protein can be integrated into a "safe harbor" locus, such as the Rosa26 locus, HPRT locus, or AAV locus.
  • a "safe harbor" locus such as the Rosa26 locus, HPRT locus, or AAV locus.
  • the present disclosure also encompasses genetically modified animals in which two, three, four, five, six, seven, eight, nine, or ten or more sequences, including sequences encoding protein(s), are integrated into the genome.
  • the chromosomally integrated sequence encoding a protein can encode the wild type form of a protein of interest or can encode a protein comprising at least one modification such that an altered version of the protein is produced.
  • a chromosomally integrated sequence encoding a protein related to a disease or disorder can comprise at least one modification such that the altered version of the protein produced causes or potentiates the associated disorder.
  • the chromosomally integrated sequence encoding a protein related to a disease or disorder can comprise at least one modification such that the altered version of the protein protects against the development of the associated disorder.
  • the genetically modified animal can be a "humanized" animal comprising at least one chromosomally integrated
  • the functional human protein can have no corresponding ortholog in the genetically modified animal.
  • the wild type animal from which the genetically modified animal is derived may comprise an ortholog corresponding to the functional human protein. In this case, the
  • orthologous sequence in the "humanized” animal is inactivated such that no functional protein is made and the "humanized” animal comprises at least one chromosomally integrated sequence encoding the human protein.
  • the genetically modified animal can comprise at least one modified chromosomal sequence encoding a protein such that the expression pattern of the protein is altered.
  • regulatory regions controlling the expression of the protein such as a promoter or a transcription factor binding site, can be altered such that the protein is over-produced, or the tissue-specific or temporal expression of the protein is altered, or a combination thereof.
  • conditional knockout system includes a Cre-lox recombination system.
  • a Cre-lox recombination system comprises a Cre recombinase enzyme, a site-specific DNA recombinase that can catalyze the
  • telomere sequence between specific sites (lox sites) in a nucleic acid molecule.
  • Methods of using this system to produce temporal and tissue specific expression are known in the art.
  • a genetically modified animal is generated with lox sites flanking a chromosomal sequence.
  • the genetically modified animal comprising the lox-flanked chromosomal sequence can then be crossed with another genetically modified animal expressing Cre recombinase.
  • Progeny animals comprising the lox-flanked chromosomal sequence and the Cre recombinase are then produced, and the lox-flanked chromosomal sequence is recombined, leading to deletion or inversion of the chromosomal sequence encoding the protein.
  • Expression of Cre recombinase can be temporally and conditionally regulated to effect temporally and conditionally regulated recombination of the chromosomal sequence.
  • the genetically modified animal disclosed herein can be heterozygous for the modified chromosomal sequence.
  • the genetically modified animal can be homozygous for the modified chromosomal sequence.
  • the genetically modified animals disclosed herein can be crossbred to create animals comprising more than one modified chromosomal sequence or to create animals that are homozygous for one or more modified chromosomal sequences.
  • two animals comprising the same modified chromosomal sequence can be crossbred to create an animal homozygous for the modified chromosomal sequence.
  • animals with different modified chromosomal sequences can be crossbred to create an animal comprising both modified chromosomal sequences.
  • a f i rst animal comprising an inactivated
  • chromosomal sequence gene "x" can be crossed with a second animal comprising a chromosomally integrated sequence encoding a human gene "X" protein to give rise to "humanized” gene "X” offspring comprising both the inactivated gene "x" chromosomal sequence and the chromosomally integrated human gene "X” sequence.
  • a humanized gene "X” animal can be crossed with a humanized gene ⁇ " animal to create humanized gene X/gene Y offspring.
  • an animal comprising a modified
  • chromosomal sequence can be crossbred to combine the modified chromosomal sequence with other genetic backgrounds.
  • other genetic backgrounds may include wild-type genetic backgrounds, genetic backgrounds with deletion mutations, genetic backgrounds with another targeted integration, and genetic backgrounds with non-targeted integrations.
  • animal refers to a non-human animal.
  • the animal may be an embryo, a juvenile, or an adult.
  • Suitable animals include vertebrates such as mammals, birds, reptiles, amphibians, shellfish, and fish. Examples of suitable mammals include without limit rodents, companion animals, livestock, and primates.
  • rodents include mice, rats, hamsters, gerbils, and guinea pigs.
  • Suitable companion animals include but are not limited to cats, dogs, rabbits, hedgehogs, and ferrets.
  • livestock include horses, goats, sheep, swine, cattle, llamas, and alpacas.
  • Suitable primates include but are not limited to capuchin monkeys, chimpanzees, lemurs, macaques,
  • marmosets tamarins, spider monkeys, squirrel monkeys, and vervet monkeys.
  • birds include chickens, turkeys, ducks, and geese.
  • the animal may be an invertebrate such as an insect, a nematode, and the like.
  • insects include Drosophila and mosquitoes.
  • An exemplary animal is a rat.
  • suitable rat strains include Dahl Salt- Sensitive, Fischer 344, Lewis, Long Evans Hooded, Sprague-Dawley, and Wistar.
  • the animal is not a genetically modified mouse. In each of the foregoing iterations of suitable animals for the invention, the animal does not include exogenously introduced, randomly integrated transposon sequences.
  • a further aspect of the present disclosure provides genetically modified cells or cell lines comprising at least one modified chromosomal sequence.
  • the genetically modified cell or cell line can be derived from any of the genetically modified animals disclosed herein.
  • the chromosomal sequence can be modified in a cell as described herein above (in the paragraphs describing chromosomal sequence modifications in animals) using the methods descried herein.
  • the disclosure also encompasses a lysate of said cells or cell lines.
  • the cells are eukaryotic cells.
  • Suitable host cells include fungi or yeast, such as Pichia, Saccharomyces, or Schizosaccharomyces; insect cells, such as SF9 cells from Spodoptera frugiperda or S2 cells from Drosophila melanogaster; and animal cells, such as mouse, rat, hamster, non-human primate, or human cells.
  • Exemplary cells are mammalian.
  • the mammalian cells can be primary cells. In general, any primary cell that is sensitive to double strand breaks may be used.
  • the cells may be of a variety of cell types, e.g., fibroblast, myoblast, T or B cell, macrophage, epithelial cell, and so forth.
  • the cell line can be any established cell line or a primary cell line that is not yet described.
  • the cell line can be adherent or non-adherent, or the cell line can be grown under conditions that encourage adherent, non-adherent or organotypic growth using standard techniques known to individuals skilled in the art.
  • suitable mammalian cells and cell lines are provided herein in section (IV)(g).
  • the cell can be a stem cell.
  • suitable stem cells are provided in section (IV)(g).
  • the present disclosure also provides a genetically modified non- human embryo comprising at least one modified chromosomal sequence.
  • the chromosomal sequence can be modified in an embryo as described herein above (in the paragraphs describing chromosomal sequence modifications in animals) using the methods descried herein.
  • the embryo is a non-human fertilized one-cell stage embryo of the animal species of interest.
  • Exemplary mammalian embryos, including one cell embryos include without limit, mouse, rat, hamster, rodent, rabbit, feline, canine, ovine, porcine, bovine, equine, and primate embryos.
  • endogenous sequence refers to a chromosomal sequence that is native to the cell.
  • exogenous refers to a sequence that is not native to the cell, or a chromosomal sequence whose native location in the genome of the cell is in a different chromosomal location.
  • a "gene,” as used herein, refers to a DNA region (including exons and introns) encoding a gene product, as well as all DNA regions which regulate the production of the gene product, whether or not such regulatory sequences are adjacent to coding and/or transcribed sequences. Accordingly, a gene includes, but is not necessarily limited to, promoter sequences, terminators, translational regulatory sequences such as ribosome binding sites and internal ribosome entry sites, enhancers, silencers, insulators, boundary elements, replication origins, matrix attachment sites, and locus control regions.
  • heterologous refers to an entity that is not endogenous or native to the cell of interest.
  • a heterologous protein refers to a protein that is derived from or was originally derived from an exogenous source, such as an exogenously introduced nucleic acid sequence. In some instances, the heterologous protein is not normally produced by the cell of interest.
  • nucleic acid and “polynucleotide” refer to a
  • deoxyribonucleotide or ribonucleotide polymer in linear or circular conformation, and in either single- or double-stranded form.
  • these terms are not to be construed as limiting with respect to the length of a polymer.
  • the terms can encompass known analogs of natural nucleotides, as well as nucleotides that are modified in the base, sugar and/or phosphate moieties (e.g., phosphorothioate backbones).
  • an analog of a particular nucleotide has the same base-pairing specificity; i.e., an analog of A will base-pair with T.
  • nucleotide refers to deoxyribonucleotides
  • nucleotides may be standard nucleotides (i.e., adenosine, guanosine, cytidine, thymidine, and uridine) or nucleotide analogs.
  • a nucleotide analog refers to a nucleotide having a modified purine or pyrimidine base or a modified ribose moiety.
  • a nucleotide analog may be a naturally occurring nucleotide (e.g., inosine) or a non-naturally occurring nucleotide.
  • Non-limiting examples of modifications on the sugar or base moieties of a nucleotide include the addition (or removal) of acetyl groups, amino groups, carboxyl groups, carboxymethyl groups, hydroxyl groups, methyl groups, phosphoryl groups, and thiol groups, as well as the substitution of the carbon and nitrogen atoms of the bases with other atoms (e.g., 7-deaza purines).
  • Nucleotide analogs also include dideoxy nucleotides, 2'-0-methyl nucleotides, locked nucleic acids (LNA), peptide nucleic acids (PNA), and morpholinos.
  • polypeptide and “protein” are used interchangeably to refer to a polymer of amino acid residues.
  • nucleic acid and amino acid sequence identity are known in the art. Typically, such techniques include determining the nucleotide sequence of the mRNA for a gene and/or determining the amino acid sequence encoded thereby, and comparing these sequences to a second nucleotide or amino acid sequence. Genomic sequences can also be determined and compared in this fashion. In general, identity refers to an exact nucleotide-to-nucleotide or amino acid-to-amino acid correspondence of two polynucleotides or polypeptide sequences, respectively. Two or more sequences (polynucleotide or amino acid) can be compared by determining their percent identity.
  • the percent identity of two sequences is the number of exact matches between two aligned sequences divided by the length of the shorter sequences and multiplied by 100.
  • An approximate alignment for nucleic acid sequences is provided by the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482- 489 (1981 ). This algorithm can be applied to amino acid sequences by using the scoring matrix developed by Dayhoff, Atlas of Protein Sequences and Structure, M. O. Dayhoff ed., 5 suppl. 3:353-358, National Biomedical Research Foundation,
  • a Cas9 gene from Streptococcus pyogenes strain MGAS15252 was optimized with Homo sapiens codon preference to enhance its translation in mammalian cells.
  • the Cas9 gene also was modified by adding a nuclear localization signal PKKKRKV (SEQ ID NO:1 ) at the C terminus for targeting the protein into the nuclei of mammalian cells.
  • Table 1 presents the modified Cas9 amino acid sequence, with the nuclear localization sequence underlined.
  • Table 2 presents the codon optimized, modified Cas9 DNA sequence.
  • the modified Cas9 DNA sequence was placed under the control of cytomegalovirus (CMV) promoter for constituent expression in mammalian cells.
  • CMV cytomegalovirus
  • the modified Cas9 DNA sequence was also placed under the control T7 promoter for in vitro mRNA synthesis with T7 RNA polymerase.
  • In vitro RNA transcription was performed by using MessageMAX T7 ARCA-Capped Message Transcription Kit and T7 mScript Standard mRNA Production System (Cellscript).
  • the adeno-associated virus integration site 1 (AAVS1 ) locus was used as a target for Cas9-mediated human genome modification.
  • the human AAVS1 locus is located in intron 1 (4427 bp) of protein phosphatase 1 , regulatory subunit 12C (PPP1 R12C).
  • Table 3 presents the first exon (shaded gray) and the first intron of PPP1 R12C.
  • the underlined sequence within the intron is the targeted modification site (i.e., AAVS1 locus).
  • Cas9 guide RNAs were designed for targeting the human AAVS1 locus.
  • a 42 nucleotide RNA (referred to herein as a "crRNA" sequence) comprising (5' to 3') a target recognition sequence (i.e., sequence complementary to the non-coding strand of the target sequence) and protospacer sequence; a 85 nucleotide RNA
  • tracrRNA sequence (referred to herein as a "tracrRNA” sequence) comprising 5' sequence with
  • a chimeric RNA comprising nucleotides 1 -32 of the crRNA, a GAAA loop, and nucleotides 19-45 of the tracrRNA were prepared.
  • the crRNA was chemically synthesized by Sigma-Aldrich.
  • the tracrRNA and chimeric RNA were synthesized by in vitro transcription with T7 RNA polymerase using T7-Scribe Standard RNA IVT Kit (Cellscript).
  • the chimeric RNA coding sequence was also placed under the control of human U6 promoter for in vivo transcription in human cells. Table 4 presents the sequences of the guide RNAs.
  • Targeted integration of a GFP protein into the N terminus of PPP1 R12C was used to monitor Cas9-mediated genome modification.
  • a donor polynucleotide was prepared.
  • the AAVS1 -GFP DNA donor contained a 5' (1 185 bp) AAVS1 locus homologous arm, an RNA splicing receptor, a turbo GFP coding sequence, a 3' transcription terminator, and a 3' (1217 bp) AAVS1 locus homologous arm.
  • Table 5 presents the sequences of the RNA splicing receptor and the GFP coding sequence followed by the 3' transcription terminator.
  • Plasmid DNA was prepared by using GenElute Endotoxin-Free Plasmid Maxiprep Kit (Sigma).
  • Targeted gene integration will result in a fusion protein between the first 107 amino acids of the PPP1 R12C and the turbo GFP.
  • the expected fusion protein contains the first 107 amino acid residues of PPP1 R12C (highlighted in grey) from RNA splicing between the first exon of PPP1 R12C and the engineered splice receptor (see Table 6).
  • the K562 cell line was obtained from American Type Culture Collection (ATCC) and grown in Iscove's Modified Dulbecco's Medium, supplemented with 10% FBS and 2 mM L-glutamine. All media and supplements were obtained from Sigma-Aldrich. Cultures were split one day before transfection (at approximately 0.5 million cells per ml_ before transfection). Cells were transfected with Nucleofector Solution V (Lonza) on a Nucleofector (Lonza) with the T-016 program. Each nucleofection contained approximately 0.6 million cells.
  • Transfection treatments are detailed in Table 7. Cells were grown at 37 °C and 5% CO 2 immediately after nucleofection.
  • FACS Fluorescence-activated cell sorting
  • Genomic DNA was extracted from transfected cells with GenElute Mammalian Genomic DNA Miniprep Kit (Sigma) 12 days after transfection. Genomic DNA was then PCR amplified with a forward primer located outside the 5' homologous arm of the AAVS1 -GFP plasmid donor and a reverse primer located at the 5' region of the GFP.
  • the forward primer was 5'- CCACTCTGTGCTGACCACTCT-3' (SEQ ID NO:18) and reverse primer was 5'- GCGGCACTCGATCTCCA-3' (SEQ ID NO:19).
  • the expected fragment size from the junction PCR was 1388 bp.
  • the amplification was carried out with JumpStart Taq ReadyMix (Sigma), using the following cycling
  • PCR products were resolved on 1 % agarose gel.
  • the mouse Rosa26 locus can be targeted for genome
  • Table 8 presents a portion of the mouse Rosa26 sequence in which potential target sites are shown in bold. Each target site comprises a protospacer.
  • RNAs were designed to target each of the target sites in the mouse Rosa26 locus.
  • the sequences are shown in Table 9, each is 42 nucleotides in length and the 5' region is complementary to the strand that is not presented in Table 8 (i.e., the strand that is complementary to the strand shown in Table 8).
  • the crRNAs were chemically synthesized and pre-annealed to the tracrRNA (SEQ ID NO:13; see Example 2).
  • Pre-annealed crRNA / tracrRNA and in vitro transcribed mRNA encoding modified Cas9 protein (SEQ ID NO. 9; see Example 1 ) can be microinjected into the pronuclei of fertilized mouse embryos.
  • the Cas9 protein cleaves the target site, and the resultant double-stranded break can be repaired via a non-homologous end-joining (NHEJ) repair process.
  • NHEJ non-homologous end-joining
  • the injected embryos can be either incubated at 37 °C, 5% CO 2 overnight or for up to 4 days, followed by genotyping analysis, or the injected embryos can be implanted into recipient female mice such that live born animals can be genotyped.
  • the in w ' fro-incu bated embryos or tissues from live born animals can be screened for the presence of Cas9-induced mutation at the Rosa locus using standard methods.
  • the embryos or tissues from fetus or live-born animals can be harvested for DNA extraction and analysis. DNA ca n b e isolated using standard procedures.
  • the targeted region of the Rosa26 locus can be PCR amplified using appropriate primers.
  • NHEJ is error-prone, deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. Mutations can be detected using PCR-based genotyping methods, such as Cel-I mismatch assays and DNA sequencing.
  • the Rosa26 locus can be modified in mouse embryos by co- injecting a donor polynucleotide, as detailed above in section (IV)(d), along with the pre- annealed crRNA / tracrRNA and mRNA encoding modified Cas9 as described above in Example 6.
  • a donor polynucleotide as detailed above in section (IV)(d)
  • mRNA encoding modified Cas9 as described above in Example 6.
  • w ' fro-incu bated embryos or tissues from live born animals (as described in Example 6) can be screened for a modified Rosa26 locus using PCR-based genotyping methods, such as RFLP assays, junction PCR, and DNA sequencing.
  • the rat Rosa26 locus can be targeted for genome modifications.
  • Table 10 presents a portion of the rat sequence in which potential target sites are shown in bold. Each target site comprises a protospacer.
  • RNAs were designed to target each of the target sites in the rat Rosa26 locus. The sequences are shown in Table 1 1 , each is 42 nucleotides in length and the 5' region is complementary to the strand that is not presented in Table 10 (i.e., the strand that is complementary to the strand shown in Table 10).
  • the crRNAs were chemically synthesized and pre-annealed to the tracrRNA (SEQ ID NO:13; see Example 2).
  • Pre-annealed crRNA / tracrRNA and in vitro transcribed mRNA encoding modified Cas9 protein (SEQ ID NO. 9; see Example 1 ) can be microinjected into the pronuclei of fertilized rat embryos.
  • the Cas9 protein cleaves the target site, and the resultant double-stranded break can be repaired via a non-homologous end-joining (NHEJ) repair process.
  • NHEJ non-homologous end-joining
  • the injected embryos can be either incubated at 37 °C, 5% CO 2 overnight or for up to 4 days, followed by genotyping analysis, or the injected embryos can be implanted into recipient female mice such that live born animals can be genotyped.
  • the in w ' fro-incu bated embryos or tissues from live born animals can be screened for the presence of Cas9-induced mutation at the Rosa locus using standard methods.
  • the embryos or tissues from fetus or live-born animals can be harvested for DNA extraction and analysis. DNA ca n b e isolated using standard procedures.
  • the targeted region of the Rosa26 locus can be PCR amplified using appropriate primers.
  • the Rosa26 locus can be modified in rat embryos by co-injecting a donor polynucleotide, as detailed above in section (IV)(d), along with the pre-annealed crRNA / tracrRNA and mRNA encoding modified Cas9 as described above in Example 8.
  • a donor polynucleotide as detailed above in section (IV)(d)
  • mRNA encoding modified Cas9 as described above in Example 8.
  • In vitro- incubated embryos or tissues from live born rats (as described in Example 8) can be screened for a modified Rosa26 locus using PCR-based genotyping methods, such as RFLP assays, junction PCR, and DNA sequencing.

Abstract

The present invention provides RNA-guided endonucleases, which are engineered for expression in eukaryotic cells or embryos, and methods of using the RNA-guided endonuclease for targeted genome modification in in eukaryotic cells or embryos. Also provided are fusion proteins, wherein each fusion protein comprises a CRISPR/Cas-like protein or fragment thereof and an effector domain. The effector domain can be a cleavage domain, an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain. Also provided are methods for using the fusion proteins to modify a chromosomal sequence or regulate expression of a chromosomal sequence.

Description

CRISPR-BASED GENOME MODIFICATION AND REGULATION
FIELD OF THE INVENTION
[0001 ] The present disclosure relates targeted genome modification. In particular, the disclosure relates to RNA-guided endonucleases or fusion proteins comprising CRISPR/Cas-like protein and methods of using said proteins to modify or regulate targeted chromosomal sequences.
BACKGROUND OF THE INVENTION
[0002] Targeted genome modification is a powerful tool for genetic manipulation of eukaryotic cells, embryos, and animals. For example, exogenous sequences can be integrated at targeted genomic locations and/or specific endogenous chromosomal sequences can be deleted, inactivated, or modified. Current methods rely on the use of engineered nuclease enzymes, such as, for example, zinc finger nucleases (ZFNs) or transcription activator-like effector nucleases (TALENs). These chimeric nucleases contain programmable, sequence-specific DNA-binding modules linked to a nonspecific DNA cleavage domain. Each new genomic target, however, requires the design of a new ZFN or TALEN comprising a novel sequence-specific DNA-binding module. Thus, these custom designed nucleases tend to be costly and time-consuming to prepare. Moreover, the specificities of ZFNs and TALENS are such that they can mediate off-target cleavages.
[0003] Thus, there is a need for a targeted genome modification
technology that does not require the design of a new nuclease for each new targeted genomic location. Additionally, there is a need for a technology with increased specificity with few or no off-target effects.
SUMMARY OF THE INVENTION
[0004] Among the various aspects of the present disclosure is the provision of an isolated RNA-guided endonuclease, wherein the endonuclease comprises at least one nuclear localization signal, at least one nuclease domain, and at least one domain that interacts with a guide RNA to target the endonuclease to a specific nucleotide sequence for cleavage. In one embodiment, the endonuclease can be derived from a Cas9 protein. In another embodiment, the endonuclease can be modified to lack at least one functional nuclease domain. In other embodiments, the endonuclease can further comprise a cell-penetrating domain, a marker domain, or both. In a further embodiment, the endonuclease can be part of a protein-RNA complex comprising the guide RNA. In some instances, the guide RNA can be a single molecule comprising a 5' region that is complementary to a target site. Also provided is an isolated nucleic acid encoding any of the RNA-guided endonucleases disclosed herein. In some embodiments, the nucleic acid can be codon optimized for translation in mammalian cells, such as, for example, human cells. In other embodiments, the nucleic acid sequence encoding the RNA-guided endonuclease can be operably linked to a promoter control sequence, and optionally, can be part of a vector. In other
embodiments, a vector comprising sequence encoding the RNA-guided endonuclease, which can be operably linked to a promoter control sequence, can also comprise sequence encoding a guide RNA, which can be operably linked to a promoter control sequence.
[0005] Another aspect of the present invention encompasses a method for modifying a chromosomal sequence in a eukaryotic cell or embryo. The method comprises introducing into a eukaryotic cell or embryo (i) at least one RNA-guided endonuclease comprising at least one nuclear localization signal or nucleic acid encoding at least one RNA-guided endonuclease as defined herein, (ii) at least one guide RNA or DNA encoding at least one guide RNA, and, optionally, (iii) at least one donor polynucleotide comprising a donor sequence. The method further comprises culturing the cell or embryo such that each guide RNA directs a RNA-guided
endonuclease to a targeted site in the chromosomal sequence where the RNA-guided endonuclease introduces a double-stranded break in the targeted site, and the double- stranded break is repaired by a DNA repair process such that the chromosomal sequence is modified. In one embodiment, the RNA-guided endonuclease can be derived from a Cas9 protein. In another embodiment, the nucleic acid encoding the RNA-guided endonuclease introduced into the cell or embryo can be mRNA. In a further embodiment, wherein the nucleic acid encoding the RNA-guided endonuclease introduced into the cell or embryo can be DNA. In a further embodiment, the DNA encoding the RNA-guided endonuclease can be part of a vector that further comprises a sequence encoding the guide RNA. In certain embodiments, the eukaryotic cell can be a human cell, a non-human mammalian cell, a stem cell, a non-mammalian vertebrate cell, an invertebrate cell, a plant cell, or a single cell eukaryotic organism. In certain other embodiments, the embryo is a non-human one cell animal embryo.
[0006] A further aspect of the disclosure provides a fusion protein comprising a CRISP R/Cas-like protein or fragment thereof and an effector domain. In general, the fusion protein comprises at least one nuclear localization signal. The effector domain of the fusion protein can be a cleavage domain, an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain. In one embodiment, the CRISPR/Cas-like protein of the fusion protein can be derived from a Cas9 protein. In one iteration, the Cas9 protein can be modified to lack at least one functional nuclease domain. In an alternate iteration, the Cas9 protein can be modified to lack all nuclease activity. In one embodiment, the effector domain can be a cleavage domain, such as, for example, a Fokl endonuclease domain or a modified Fokl endonuclease domain. In another embodiment, one fusion protein can form a dimer with another fusion protein. The dimer can be a homodimer or a heterodimer. In another embodiment, the fusion protein can form a heterodimer with a zinc finger nuclease, wherein the cleavage domain of both the fusion protein and the zinc finger nucleases is a Fokl endonuclease domain or a modified Fokl endonuclease domain. In still another embodiment, the fusion protein comprises a CRISPR/Cas-like protein derived from a Cas9 protein modified to lack all nuclease activity, and the effector domain is a Fokl endonuclease domain or a modified Fokl endonuclease domain. In still another embodiment, the fusion protein comprises a CRISPR/Cas-like protein derived from a Cas9 protein modified to lack all nuclease activity, and the effector domain can be an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain. In additional embodiments, any of the fusion proteins disclosed herein can comprise at least one additional domain chosen from a nuclear localization signal, a cell-penetrating domain, and a marker domain. Also provided are isolated nucleic acids encoding any of the fusion proteins provided herein.
[0007] Still another aspect of the disclosure encompasses a method for modifying a chromosomal sequence or regulating expression of a chromosomal sequence in a cell or embryo. The method comprises introducing into the cell or embryo (a) at least one fusion protein or nucleic acid encoding at least one fusion protein, wherein the fusion protein comprises a CRISPR/Cas-like protein or a fragment thereof and an effector domain, and (b) at least one guide RNA or DNA encoding at least one guide RNA, wherein the guide RNA guides the CRISPR/Cas-like protein of the fusion protein to a targeted site in the chromosomal sequence and the effector domain of the fusion protein modifies the chromosomal sequence or regulates expression of the chromosomal sequence. In one embodiment, the CRISPR/Cas-like protein of the fusion protein can be derived from a Cas9 protein. In another embodiment, the CRISPR/Cas- like protein of the fusion protein can be modified to lack at least one functional nuclease domain. In still another embodiment, the CRISPR/Cas-like protein of the fusion protein can be modified to lack all nuclease activity. In one embodiment in which the fusion protein comprises a Cas9 protein modified to lack all nuclease activity and a Fokl cleavage domain or a modified Fokl cleavage domain, the method can comprise introducing into the cell or embryo one fusion protein or nucleic acid encoding one fusion protein and two guide RNAs or DNA encoding two guide RNAs, and wherein one double-stranded break is introduced in the chromosomal sequence. In another embodiment in which the fusion protein comprises a Cas9 protein modified to lack all nuclease activity and a Fokl cleavage domain or a modified Fokl cleavage domain, the method can comprise introducing into the cell or embryo two fusion proteins or nucleic acid encoding two fusion proteins and two guide RNAs or DNA encoding two guide RNAs, and wherein two double-stranded breaks are introduced in the chromosomal sequence. In still another one embodiment in which the fusion protein comprises a Cas9 protein modified to lack all nuclease activity and a Fokl cleavage domain or a modified Fokl cleavage domain, the method can comprise introducing into the cell or embryo one fusion protein or nucleic acid encoding one fusion protein, one guide RNA or nucleic acid encoding one guide RNA, and one zinc finger nuclease or nucleic acid encoding one zinc finger nuclease, wherein the zinc finger nuclease comprises a Fokl cleavage domain or a modified a Fokl cleavage domain, and wherein one double- stranded break is introduced into the chromosomal sequence. In certain embodiments in which the fusion protein comprises a cleavage domain, the method can further comprise introducing into the cell or embryo at least one donor polynucleotide. In embodiments in which the fusion protein comprises an effector domain chosen from an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain, the fusion protein can comprise a Cas9 protein modified to lack all nuclease activity, and the method can comprise introducing into the cell or embryo one fusion protein or nucleic acid encoding one fusion protein, and one guide RNA or nucleic acid encoding one guide RNA, and wherein the structure or expression of the targeted chromosomal sequence is modified. In certain embodiments, the eukaryotic cell can be a human cell, a non-human mammalian cell, a stem cell, a non-mammalian vertebrate cell, an invertebrate cell, a plant cell, or a single cell eukaryotic organism. In certain other embodiments, the embryo is a non-human one cell animal embryo.
[0008] Other aspects and iterations of the disclosure are detailed below.
BRIEF DESCRIPTION OF THE DRAWINGS
[0009] FIG. 1 diagrams genome modification using protein dimers. (A) depicts a double stranded break created by a dimer composed of two fusion proteins, each of which comprises a Cas-like protein for DNA binding and a Fokl cleavage domain. (B) depicts a double stranded break created by a dimer composed of a fusion protein comprising a Cas-like protein and a Fokl cleavage domain and a zinc finger nuclease comprising a zinc finger (ZF) DNA-binding domain and a Fokl cleavage domain.
[0010] FIG. 2 illustrates regulation of gene expression using RNA-guided fusion proteins comprising gene regulatory domains. (A) depicts a fusion protein comprising a Cas-like protein used for DNA binding and an "A/R" domain that activates or represses gene expression. (B) diagrams a fusion protein comprising a Cas-like protein for DNA binding and a epigenetic modification domain ("Epi-mod') that affects epigenetic states by covalent modification of proximal DNA or proteins. [001 1 ] FIG. 3 diagrams genome modification using two RNA-guided endonuclease. (A) depicts a double stranded break created by two RNA-guided endonuclease that have been converted into nickases. (B) depicts two double stranded breaks created by two RNA-guided endonuclease having endonuclease activity.
[0012] FIG. 4 presents fluorescence-activated cell sorting (FACS) of human K562 cells transfected with Cas9 nucleic acid, Cas9 guiding RNA, and AAVS1 - GFP DNA donor. The Y axis represents the auto fluorescence intensity at a red channel, and the X axis represents the green fluorescence intensity. (A) K562 cells transfected with 10 μg of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of pre-annealed crRNA-tracrRNA duplex, and 10 μg of AAVS1 -GFP plasmid DNA; (B) K562 cells transfected 10 μg of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of chimeric RNA, and 10 μg of AAVS1 -GFP plasmid DNA; (C) K562 cells transfected 10 μg of Cas9 mRNA that was capped by post-transcription capping reaction, 0.3 nmol of chimeric RNA, and 10 μg of AAVS1 -GFP plasmid DNA; (D) K562 cells transfected with 10 μg of Cas9 plasmid DNA, 5 μg of U6-chimeric RNA plasmid DNA, and 10 μg of AAVS1 -GFP plasmid DNA; (E) K562 cells transfected with 10 μg of AAVS1 -GFP plasmid DNA; (F) K562 cells transfected with transfection reagents only.
[0013] FIG. 5 presents a junction PCR analysis documenting the targeted integration of GFP into the AAVS1 locus in human cells. Lane M: 1 kb DNA molecular markers; Lane A: K562 cells transfected with 10 μg of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of pre-annealed crRNA-tracrRNA duplex, and 10 μg of AAVS1 -GFP plasmid DNA; Lane B: K562 cells transfected 10 μg of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of chimeric RNA, and 10 μg of AAVS1 -GFP plasmid DNA; Lane C: K562 cells transfected 10 μg of Cas9 mRNA that was capped by post-transcription capping reaction, 0.3 nmol of chimeric RNA, and 10 μg of AAVS1 -GFP plasmid DNA; Lane D: K562 cells transfected with 10 μg of Cas9 plasmid DNA, 5 μg of U6-chimeric RNA plasmid DNA, and 10 μg of AAVS1 -GFP plasmid DNA; Lane E: K562 cells transfected with 10 μg of AAVS1 -GFP plasmid DNA; Lane F: K562 cells transfected with transfection reagents only. DETAILED DESCRIPTION OF THE INVENTION
[0014] Provided herein are RNA-guided endonucleases, which comprise at least one nuclear localization signal, at least one nuclease domain, and at least one domain that interacts with a guide RNA to target the endonuclease to a specific nucleotide sequence for cleavage. Also provided are nucleic acids encoding the RNA- guided endonucleases, as well as methods of using the RNA-guided endonucleases to modify chromosomal sequences of eukaryotic cells or embryos. The RNA-guided endonuclease interacts with specific guide RNAs, each of which directs the
endonuclease to a specific targeted site, at which site the RNA-guided endonuclease introduces a double-stranded break that can be repaired by a DNA repair process such that the chromosomal sequence is modified. Since the specificity is provided by the guide RNA, the RNA-based endonuclease is universal and can be used with different guide RNAs to target different genomic sequences. The methods disclosed herein can be used to target and modify specific chromosomal sequences and/or introduce exogenous sequences at targeted locations in the genome of cells or embryos.
Furthermore, the targeting is specific with limited off target effects.
[0015] The present disclosure provides fusion proteins, wherein a fusion protein comprises a CRISPR/Cas-like protein or fragment thereof and an effector domain. Suitable effector domains include, without limit, cleavage domains, epigenetic modification domains, transcriptional activation domains, and transcriptional repressor domains. Each fusion protein is guided to a specific chromosomal sequence by a specific guide RNA, wherein the effector domain mediates targeted genome
modification or gene regulation. In one aspect, the fusion proteins can function as dimers thereby increasing the length of the target site and increasing the likelihood of its uniqueness in the genome (thus, reducing off target effects). For example, endogenous CRISPR systems modify genomic locations based on DNA binding word lengths of approximately 13-20 bp (Cong et al., Science, 339:819-823). At this word size, only 5- 7% of the target sites are unique within the genome (Iseli et al, PLos One 2(6):e579). In contrast, DNA binding word sizes for zinc finger nucleases typically range from 30-36 bp, resulting in target sites that are approximately 85-87% unique within the human genome. The smaller sized DNA binding sites utilized by CRISPR-based systems limits and complicates design of targeted CRISP-based nucleases near desired locations, such as disease SNPs, small exons, start codons, and stop codons, as well as other locations within complex genomes. The present disclosure not only provides means for expanding the CRISPR DNA binding word length (i.e., so as to limit off-target activity), but further provides CRISPR fusion proteins having modified functionality. According, the disclosed CRISPR fusion proteins have increased target specificity and unique functionality(ies). Also provided herein are methods of using the fusion proteins to modify or regulate expression of targeted chromosomal sequences.
(I) RNA-Guided Endonucleases
[0016] One aspect of the present disclosure provides RNA-guided endonucleases comprising at least one nuclear localization signal, which permits entry of the endonuclease into the nuclei of eukaryotic cells and embryos such as, for example, non-human one cell embryos. RNA-guided endonucleases also comprise at least one nuclease domain and at least one domain that interacts with a guide RNA. An RNA-guided endonuclease is directed to a specific nucleic acid sequence (or target site) by a guide RNA. The guide RNA interacts with the RNA-guided endonuclease as well as the target site such that, once directed to the target site, the RNA-guided
endonuclease is able to introduce a double-stranded break into the target site nucleic acid sequence. Since the guide RNA provides the specificity for the targeted cleavage, the endonuclease of the RNA-guided endonuclease is universal and can be used with different guide RNAs to cleave different target nucleic acid sequences. Provided herein are isolated RNA-guided endonucleases, isolated nucleic acids (i.e., RNA or DNA) encoding the RNA-guided endonucleases, vectors comprising nucleic acids encoding the RNA-guided endonucleases, and protein-RNA complexes comprising the RNA- guided endonuclease plus a guide RNA.
[0017] The RNA-guided endonuclease can be derived from a clustered regularly interspersed short p_alindromic repeats (CRISPR)/CRISPR-associated (Cas) system. The CRISPR/Cas system can be a type I, a type II, or a type III system. Non- limiting examples of suitable CRISPR/Cas proteins include Cas3, Cas4, Cas5, Cas5e (or CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8a1 , Cas8a2, Cas8b, Cas8c, Cas9, Cas10, Casl Od, CasF, CasG, CasH, Csy1 , Csy2, Csy3, Cse1 (or CasA), Cse2 (or CasB), Cse3 (or CasE), Cse4 (or CasC), Csc1 , Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1 , Cmr3, Cmr4, Cmr5, Cmr6, Csb1 , Csb2, Csb3,Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csz1 , Csx15, Csf1 , Csf2, Csf3, Csf4, and Cu1966.
[0018] In one embodiment, the RNA-guided endonuclease is derived from a type II CRISPR/Cas system. In specific embodiments, the RNA-guided endonuclease is derived from a Cas9 protein. The Cas9 protein can be from Streptococcus pyogenes, Streptococcus thermophilus, Streptococcus sp., Nocardiopsis dassonvillei,
Streptomyces pristinaespiralis, Streptomyces viridochromogenes, Streptomyces viridochromogenes, Streptosporangium roseum, Streptosporangium roseum,
AlicyclobacHlus acidocaldarius, Bacillus pseudomycoides, Bacillus selenitireducens, Exiguobacterium sibiricum, Lactobacillus delbrueckii, Lactobacillus salivarius,
Microscilla marina, Burkholderiales bacterium, Polaromonas naphthalenivorans,
Polaromonas sp., Crocosphaera watsonii, Cyanothece sp., Microcystis aeruginosa, Synechococcus sp., Acetohalobium arabaticum, Ammonifex degensii,
Caldicelulosiruptor becscii, Candidatus Desulforudis, Clostridium botulinum, Clostridium difficile, Finegoldia magna, Natranaerobius thermophilus, Pelotomaculum
thermopropionicum, Acidithiobacillus caldus, Acidithiobacillus ferrooxidans,
Allochromatium vinosum, Marinobacter sp., Nitrosococcus halophilus, Nitrosococcus watsoni, Pseudoalteromonas haloplanktis, Ktedonobacter racemifer, Methanohalobium evestigatum, Anabaena variabilis, Nodularia spumigena, Nostoc sp., Arthrospira maxima, Arthrospira platensis, Arthrospira sp., Lyngbya sp., Microcoleus
chthonoplastes, Oscillatoria sp., Petrotoga mobilis, Thermosipho africanus, or
Acaryochloris marina.
[0019] In general, CRISPR/Cas proteins comprise at least one RNA recognition and/or RNA binding domain. RNA recognition and/or RNA binding domains interact with guide RNAs. CRISPR/Cas proteins can also comprise nuclease domains (i.e., DNase or RNase domains), DNA binding domains, helicase domains, RNAse domains, protein-protein interaction domains, dimerization domains, as well as other domains. [0020] The CRISPR/Cas-like protein can be a wild type CRISPR/Cas protein, a modified CRISPR/Cas protein, or a fragment of a wild type or modified
CRISPR/Cas protein. The CRISPR/Cas-like protein can be modified to increase nucleic acid binding affinity and/or specificity, alter an enzymatic activity, and/or change another property of the protein. For example, nuclease (i.e., DNase, RNase) domains of the CRISPR/Cas-like protein can be modified, deleted, or inactivated. Alternatively, the CRISPR/Cas-like protein can be truncated to remove domains that are not essential for the function of the fusion protein. The CRISPR/Cas-like protein can also be truncated or modified to optimize the activity of the effector domain of the fusion protein.
[0021 ] In some embodiments, the CRISPR/Cas-like protein can be derived from a wild type Cas9 protein or fragment thereof. In other embodiments, the
CRISPR/Cas-like protein can be derived from modified Cas9 protein. For example, the amino acid sequence of the Cas9 protein can be modified to alter one or more properties (e.g., nuclease activity, affinity, stability, etc.) of the protein. Alternatively, domains of the Cas9 protein not involved in RNA-guided cleavage can be eliminated from the protein such that the modified Cas9 protein is smaller than the wild type Cas9 protein.
[0022] In general, a Cas9 protein comprises at least two nuclease (i.e., DNase) domains. For example, a Cas9 protein can comprise a RuvC-like nuclease domain and a HNH-like nuclease domain. The RuvC and HNH domains work together to cut single strands to make a double-stranded break in DNA. (Jinek et al., Science, 337: 816-821 ). In some embodiments, the Cas9-derived protein can be modified to contain only one functional nuclease domain (either a RuvC-like or a HNH-like nuclease domain). For example, the Cas9-derived protein can be modified such that one of the nuclease domains is deleted or mutated such that it is no longer functional (i.e., the nuclease activity is absent). In some embodiments in which one of the nuclease domains is inactive, the Cas9-derived protein is able to introduce a nick into a double- stranded nucleic acid (such protein is termed a "nickase"), but not cleave the double- stranded DNA. For example, an aspartate to alanine (D10A) conversion in a RuvC-like domain converts the Cas9-derived protein into a nickase. Likewise, a histidine to alanine (H840A or H839A) conversion in a HNH domain converts the Cas9-derived protein into a nickase. Each nuclease domain can be modified using well-known methods, such as site-directed mutagenesis, PCR-mediated mutagenesis, and total gene synthesis, as well as other methods known in the art.
[0023] The RNA-guided endonuclease disclosed herein comprises at least one nuclear localization signal. In general, an NLS comprises a stretch of basic amino acids. Nuclear localization signals are known in the art (see, e.g., Lange et al., J. Biol. Chem., 2007, 282:5101 -5105). For example, in one embodiment, the NLS can be a monopartite sequence, such as PKKKRKV (SEQ ID NO:1 ) or PKKKRRV (SEQ ID NO:2). In another embodiment, the NLS can be a bipartite sequence. In still another embodiment, the NLS can be KRPAATKKAGQAKKKK (SEQ ID NO:3). The NLS can be located at the N-terminus, the C-terminal, or in an internal location of the RNA- guided endonuclease.
[0024] In some embodiments, the RNA-guided endonuclease can further comprise at least one cell-penetrating domain. In one embodiment, the cell-penetrating domain can be a cell-penetrating peptide sequence derived from the HIV-1 TAT protein. As an example, the TAT cell-penetrating sequence can be
GRKKRRQRRRPPQPKKKRKV (SEQ ID NO:4). In another embodiment, the cell- penetrating domain can be TLM (PLSSIFSRIGDPPKKKRKV; SEQ ID NO:5), a cell- penetrating peptide sequence derived from the human hepatitis B virus. In still another embodiment, the cell-penetrating domain can be MPG
(GALFLGWLGAAGSTMGAPKKKRKV; SEQ ID NO:6 or
GALFLGFLGAAGSTMGAWSQPKKKRKV; SEQ ID NO:7). In an additional
embodiment, the cell-penetrating domain can be Pep-1
(KETWWETWWTEWSQPKKKRKV; SEQ ID NO:8), VP22, a cell penetrating peptide from Herpes simplex virus, or a polyarginine peptide sequence. The cell-penetrating domain can be located at the N-terminus, the C-terminus, or in an internal location of the protein.
[0025] In still other embodiments, the RNA-guided endonuclease can also comprise at least one marker domain. Non-limiting examples of marker domains include fluorescent proteins, purification tags, and epitope tags. In some embodiments, the marker domain can be a fluorescent protein. Non limiting examples of suitable 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 fluorescent proteins (e.g. EBFP, EBFP2, 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, mRFP1 , DsRed-Express, DsRed2, DsRed-Monomer, HcRed- Tandem, HcRedl , AsRed2, eqFP61 1 , mRasberry, mStrawberry, Jred), and orange fluorescent proteins (mOrange, mKO, Kusabira-Orange, Monomeric Kusabira-Orange, mTangerine, tdTomato) or any other suitable fluorescent protein. In other
embodiments, the marker domain can be a purification tag and/or an epitope tag.
Exemplary tags include, but are not limited to, glutathione-S-transferase (GST), chitin binding protein (CBP), maltose binding protein, thioredoxin (TRX), poly(NANP), tandem affinity purification (TAP) tag, myc, AcV5, AU1 , AU5, E, ECS, E2, FLAG, HA, nus, Softag 1 , Softag 3, Strep, SBP, Glu-Glu, HSV, KT3, S, S1 , T7, V5, VSV-G, 6xHis, biotin carboxyl carrier protein (BCCP), and calmodulin.
[0026] In certain embodiments, the RNA-guided endonuclease may be part of a protein-RNA complex comprising a guide RNA. The guide RNA interacts with the RNA-guided endonuclease to direct the endonuclease to a specific target site, wherein the 5' end of the guide RNA base pairs with a specific protospacer sequence.
(II) Fusion Proteins
[0027] Another aspect of the present disclosure provides a fusion protein comprising a CRISP R/Cas-like protein or fragment thereof and an effector domain. The CRISP R/Cas-like protein is directed to a target site by a guide RNA, at which site the effector domain can modify or effect the targeted nucleic acid sequence. The effector domain can be a cleavage domain, an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain. The fusion protein can further comprise at least one additional domain chosen from a nuclear localization signal, a cell-penetrating domain, or a marker domain. (a) CRISPR/Cas-like protein
[0028] The fusion protein comprises a CRISPR/Cas-like protein or a fragment thereof. CRISPR/Cas-like proteins are detailed above in section (I). The CRISPR/Cas-like protein can be located at the N-terminus, the C-terminus, or in an internal location of the fusion protein
[0029] In some embodiments, the CRISPR/Cas-like protein of the fusion protein can be derived from a Cas9 protein. The Cas9-derived protein can be wild type, modified, or a fragment thereof. In some embodiments, the Cas9-derived protein can be modified to contain only one functional nuclease domain (either a RuvC-like or a HNH-like nuclease domain). For example, the Cas9-derived protein can be modified such that one of the nuclease domains is deleted or mutated such that it is no longer functional (i.e., the nuclease activity is absent). In some embodiments in which one of the nuclease domains is inactive, the Cas9-derived protein is able to introduce a nick into a double-stranded nucleic acid (such protein is termed a "nickase"), but not cleave the double-stranded DNA. For example, an aspartate to alanine (D10A) conversion in a RuvC-like domain converts the Cas9-derived protein into a nickase. Likewise, a histidine to alanine (H840A or H839A) conversion in a HNH domain converts the Cas9- derived protein into a nickase. In other embodiments, both of the RuvC-like nuclease domain and the HNH-like nuclease domain can be modified or eliminated such that the Cas9-derived protein is unable to nick or cleave double stranded nucleic acid. In still other embodiments, all nuclease domains of the Cas9-derived protein can be modified or eliminated such that the Cas9-derived protein lacks all nuclease activity.
[0030] In any of the above-described embodiments, any or all of the nuclease domains can be inactivated by one or more deletion mutations, insertion mutations, and/or substitution mutations using well-known methods, such as site- directed mutagenesis, PCR-mediated mutagenesis, and total gene synthesis, as well as other methods known in the art. In an exemplary embodiment, the CRISPR/Cas-like protein of the fusion protein is derived from a Cas9 protein in which all the nuclease domains have been inactivated or deleted. (b) Effector domain
[0031 ] The fusion protein also comprises an effector domain. The effector domain can be a cleavage domain, an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain. The effector domain can be located at the N-terminus, the C-terminus, or in an internal location of the fusion protein.
(i) cleavage domain
[0032] In some embodiments, the effector domain is a cleavage domain. As used herein, a "cleavage domain" refers to a domain that cleaves DNA. The cleavage domain can be obtained from any endonuclease or exonuclease. Non-limiting examples of endonucleases from which a cleavage domain can be derived include, but are not limited to, restriction endonucleases and homing endonucleases. See, for example, New England Biolabs Catalog or Belfort et al. (1997) Nucleic Acids Res.
25:3379-3388. Additional enzymes that cleave DNA are known (e.g., S1 Nuclease; mung bean nuclease; pancreatic DNase I ; micrococcal nuclease; yeast HO
endonuclease). See also Linn et al. (eds.) Nucleases, Cold Spring Harbor Laboratory Press, 1993. One or more of these enzymes (or functional fragments thereof) can be used as a source of cleavage domains.
[0033] In some embodiments, the cleavage domain can be derived from a type ll-S endonuclease. Type ll-S endonucleases cleave DNA at sites that are typically several base pairs away the recognition site and, as such, have separable recognition and cleavage domains. These enzymes generally are monomers that transiently associate to form dimers to cleave each strand of DNA at staggered locations. Non- limiting examples of suitable type ll-S endonucleases include Bfil, Bpml, Bsal, Bsgl, BsmBI, Bsml, BspMI, Fokl, Mboll, and Sapl. In exemplary embodiments, the cleavage domain of the fusion protein is a Fokl cleavage domain or a derivative thereof.
[0034] In certain embodiments, the type ll-S cleavage can be modified to facilitate dimerization of two different cleavage domains (each of which is attached to a CRISP R/Cas-like protein or fragment thereof). For example, the cleavage domain of Fokl can be modified by mutating certain amino acid residues. By way of non-limiting example, amino acid residues at positions 446, 447, 479, 483, 484, 486, 487, 490, 491 , 496, 498, 499, 500, 531 , 534, 537, and 538 of Fokl cleavage domains are targets for modification. For example, modified cleavage domains of Fokl that form obligate heterodimers include a pair in which a first modified cleavage domain includes mutations at amino acid positions 490 and 538 and a second modified cleavage domain that includes mutations at amino acid positions 486 and 499 (Miller et al., 2007, Nat. Biotechnol, 25:778-785; Szczpek et al., 2007, Nat. Biotechnol, 25:786-793). For example, the Glu (E) at position 490 can be changed to Lys (K) and the lie (I) at position 538 can be changed to K in one domain (E490K, I538K), and the Gin (Q) at position 486 can be changed to E and the I at position 499 can be changed to Leu (L) in another cleavage domain (Q486E, I499L). In other embodiments, modified Fokl cleavage domains can include three amino acid changes (Doyon et al. 201 1 , Nat. Methods, 8:74- 81 ). For example, one modified Fokl domain (which is termed ELD) can comprise Q486E, I499L, N496D mutations and the other modified Fokl domain (which is termed KKR) can comprise E490K, I538K, H537R mutations.
[0035] In exemplary embodiments, the effector domain of the fusion protein is a Fokl cleavage domain or a modified Fokl cleavage domain.
[0036] In embodiments wherein the effector domain is a cleavage domain and the CRISP R/Cas-like protein is derived from a Cas9 protein, the Cas9-derived can be modified as discussed herein such that its endonuclease activity is eliminated. For example, the Cas9-derived can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
(ii) epiqenetic modification domain
[0037] In other embodiments, the effector domain of the fusion protein can be an epigenetic modification domain. In general, epigenetic modification domains alter histone structure and/or chromosomal structure without altering the DNA sequence. Changes histone and/or chromatin structure can lead to changes in gene expression. Examples of epigenetic modification include, without limit, acetylation or methylation of lysine residues in histone proteins, and methylation of cytosine residues in DNA. Non- limiting examples of suitable epigenetic modification domains include histone
acetyltansferase domains, histone deacetylase domains, histone methyltransferase domains, histone demethylase domains, DNA methyltransferase domains, and DNA demethylase domains.
[0038] In embodiments in which the effector domain is a histone
acetyltansferase (HAT) domain, the HAT domain can be derived from EP300 (i.e., E1 A binding protein p300), CREBBP (i.e., CREB-binding protein), CDY1 , CDY2, CDYL1 , CLOCK, ELP3, ESA1 , GCN5 (KAT2A), HAT1 ,KAT2B, KAT5, MYST1 , MYST2, MYST3, MYST4, NCOA1 , NCOA2, NCOA3, NCOAT, P/CAF, Tip60, TAFII250, or TF3C4. In one such embodiment, the HAT domain is p300
[0039] In embodiments wherein the effector domain is an epigenetic modification domain and the CRISPR/Cas-like protein is derived from a Cas9 protein, the Cas9-derived can be modified as discussed herein such that its endonuclease activity is eliminated. For example, the Cas9-derived can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
(iii) transcriptional activation domain
[0040] In other embodiments, the effector domain of the fusion protein can be a transcriptional activation domain. In general, a transcriptional activation domain interacts with transcriptional control elements and/or transcriptional regulatory proteins (i.e., transcription factors, RNA polymerases, etc.) to increase and/or activate
transcription of a gene. In some embodiments, the transcriptional activation domain can be, without limit, a herpes simplex virus VP16 activation domain, VP64 (which is a tetrameric derivative of VP16), a N FKB p65 activation domain, p53 activation domains 1 and 2, a CREB (cAMP response element binding protein) activation domain, an E2A activation domain, and an NFAT (nuclear factor of activated T-cells) activation domain. In other embodiments, the transcriptional activation domain can be Gal4, Gcn4, MLL, Rtg3, Gln3, Oaf1 , Pip2, Pdr1 , Pdr3, Pho4, and Leu3. The transcriptional activation domain may be wild type, or it may be a modified version of the original transcriptional activation domain. In some embodiments, the effector domain of the fusion protein is a VP16 or VP64 transcriptional activation domain.
[0041 ] In embodiments wherein the effector domain is a transcriptional activation domain and the CRISPR/Cas-like protein is derived from a Cas9 protein, the Cas9-derived protein can be modified as discussed herein such that its endonuclease activity is eliminated. For example, the Cas9-derived can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
(iv) transcriptional repressor domain
[0042] In still other embodiments, the effector domain of the fusion protein can be a transcriptional repressor domain. In general, a transcriptional repressor domain interacts with transcriptional control elements and/or transcriptional regulatory proteins (i.e., transcription factors, RNA polymerases, etc.) to decrease and/or terminate transcription of a gene. Non-limiting examples of suitable transcriptional repressor domains include inducible cAMP early repressor (ICER) domains, Kruppel-associated box A (KRAB-A) repressor domains, YY1 glycine rich repressor domains, Sp1 -like repressors, E(spl) repressors, ΙκΒ repressor, and MeCP2.
[0043] In embodiments wherein the effector domain is a transcriptional repressor domain and the CRISP R/Cas-like protein is derived from a Cas9 protein, the Cas9-derived protein can be modified as discussed herein such that its endonuclease activity is eliminated. For example, the cas9 can be modified by mutating the RuvC and HNH domains such that they no longer possess nuclease activity.
(c) Additional domains
[0044] In some embodiments, the fusion protein further comprises at least one additional domain. Non-limiting examples of suitable additional domains include nuclear localization signals, cell-penetrating or translocation domains, and marker domains. Non-limiting examples of suitable nuclear localization signals, cell-penetrating domains, and marker domains are presented above in section (I).
(d) Fusion protein dimers
[0045] In embodiments in which the effector domain of the fusion protein is a cleavage domain, a dimer comprising at least one fusion protein can form. The dimer can be a homodimer or a heterodimer. In some embodiments, the heterodimer comprises two different fusion proteins. In other embodiments, the heterodimer comprises one fusion protein and an additional protein.
[0046] In some embodiments, the dimer is a homodimer in which the two fusion protein monomers are identical with respect to the primary amino acid sequence. In one embodiment where the dimer is a homodimer, the Cas9-derived proteins are modified such that their endonuclease activity is eliminated, i.e., such that they have no functional nuclease domains. In certain embodiments wherein the Cas9-derived proteins are modified such that their endonuclease activity is eliminated, each fusion protein monomer comprises an identical Cas9 like protein and an identical cleavage domain. The cleavage domain can be any cleavage domain, such as any of the exemplary cleavage domains provided herein. In one specific embodiment, the cleavage domain is a Fokl cleavage domain or a modified Fokl cleavage domain. In such embodiments, specific guide RNAs would direct the fusion protein monomers to different but closely adjacent sites such that, upon dimer formation, the nuclease domains of the two monomers would create a double stranded break in the target DNA.
[0047] In other embodiments, the dimer is a heterodimer of two different fusion proteins. For example, the CRISP R/Cas-like protein of each fusion protein can be derived from a different CRISPR/Cas protein or from an orthologous CRISPR/Cas protein from a different bacterial species. For example, each fusion protein can comprise a Cas9-like protein, which Cas9-like protein is derived from a different bacterial species. In these embodiments, each fusion protein would recognize a different target site (i.e., specified by the protospacer and/or PAM sequence). For example, the guide RNAs could position the heterodimer to different but closely adjacent sites such that their nuclease domains results in an effective double stranded break in the target DNA. The heterodimer can also have modified Cas9 proteins with nicking activity such that the nicking locations are different.
[0048] Alternatively, two fusion proteins of a heterodimer can have different effector domains. In embodiments in which the effector domain is a cleavage domain, each fusion protein can contain a different modified cleavage domain. For example, each fusion protein can contain a different modified Fokl cleavage domain, as detailed above in section (ll)(b)(i). In these embodiments, the Cas-9 proteins can be modified such that their endonuclease activities are eliminated.
[0049] As will be appreciated by those skilled in the art, the two fusion proteins forming a heterodimer can differ in both the CRISPR/Cas-like protein domain and the effector domain.
[0050] In any of the above-described embodiments, the homodimer or heterodimer can comprise at least one additional domain chosen from nuclear localization signals (NLSs), cell-penetrating, translocation domains and marker domains, as detailed above.
[0051 ] In any of the above-described embodiments, one or both of the Cas9-derived proteins can be modified such that its endonuclease activity is eliminated or modified.
[0052] In still alternate embodiments, the heterodimer comprises one fusion protein and an additional protein. For example, the additional protein can be a nuclease. In one embodiment, the nuclease is a zinc finger nuclease. A zinc finger nuclease comprises a zinc finger DNA binding domain and a cleavage domain. A zinc finger recognizes and binds three (3) nucleotides. A zinc finger DNA binding domain can comprise from about three zinc fingers to about seven zinc fingers. The zinc finger DNA binding domain can be derived from a naturally occurring protein or it can be engineered. See, for example, Beerii et al. (2002) Nat. Biotechnol. 20:135-141 ; Pabo et al. (2001 ) Ann. Rev. Biochem. 70:313-340; Isalan et al. (2001 ) Nat. Biotechnol. 19:656- 660; Segal et al. (2001 ) Curr. Opin. Biotechnol. 12:632-637; Choo et al. (2000) Curr. Opin. Struct. Biol. 10:41 1 -416; Zhang et al. (2000) J. Biol. Chem. 275(43):33850-33860; Doyon et al. (2008) Nat. Biotechnol. 26:702-708; and Santiago et al. (2008) Proc. Natl. Acad. Sci. USA 105:5809-5814. The cleavage domain of the zinc finger nuclease can be any cleavage domain detailed above in section (ll)(b)(i). In exemplary embodiments, the cleavage domain of the zinc finger nuclease is a Fokl cleavage domain or a modified Fokl cleavage domain. Such a zinc finger nuclease will dimerize with a fusion protein comprising a Fokl cleavage domain or a modified Fokl cleavage domain. [0053] In some embodiments, the zinc finger nuclease can comprise at least one additional domain chosen from nuclear localization signals, cell-penetrating or translocation domains, which are detailed above.
[0054] In certain embodiments, any of the fusion protein detailed above or a dimer comprising at least one fusion protein may be part of a protein-RNA complex comprising at least one guide RNA. A guide RNA interacts with the CRISPR-CasOlike protein of the fusion protein to direct the fusion protein to a specific target site, wherein the 5' end of the guide RNA base pairs with a specific protospacer sequence.
(Ill) Nucleic Acids Encoding RNA-Guided Endonucleases or Fusion Proteins
[0055] Another aspect of the present disclosure provides nucleic acids encoding any of the RNA-guided endonucleases or fusion proteins described above in sections (I) and (II), respectively. The nucleic acid can be RNA or DNA. In one embodiment, the nucleic acid encoding the RNA-guided endonuclease or fusion protein is mRNA. The m RNA can be 5' capped and/or 3' polyadenylated. In another embodiment, the nucleic acid encoding the RNA-guided endonuclease or fusion protein is DNA. The DNA can be present in a vector (see below).
[0056] The nucleic acid encoding the RNA-guided endonuclease or fusion protein can be codon optimized for efficient translation into protein in the eukaryotic cell or animal of interest. For example, codons can be optimized for expression in humans, mice, rats, hamsters, cows, pigs, cats, dogs, fish, amphibians, plants, yeast, insects, and so forth (see Codon Usage Database at www.kazusa.or.jp/codon/). Programs for codon optimization are available as freeware (e.g., OPTIMIZER at
genomes.urv.es/OPTIMIZER; OptimumGene™ from GenScript at
www.genscript.com/codon_opt.html). Commercial codon optimization programs are also available.
[0057] In some embodiments, DNA encoding the RNA-guided
endonuclease or fusion protein can be operably linked to at least one promoter control sequence. In some iterations, the DNA coding sequence can be operably linked to a promoter control sequence for expression in the eukaryotic cell or animal of interest. The promoter control sequence can be constitutive, regulated, or tissue-specific. Suitable constitutive promoter control sequences include, but are not limited to, cytomegalovirus immediate early promoter (CMV), simian virus (SV40) promoter, adenovirus major late promoter, Rous sarcoma virus (RSV) promoter, mouse mammary tumor virus (MMTV) promoter, phosphoglycerate kinase (PGK) promoter, elongation factor (EDI )-alpha promoter, ubiquitin promoters, actin promoters, tubulin promoters, immunoglobulin promoters, fragments thereof, or combinations of any of the foregoing. Examples of suitable regulated promoter control sequences include without limit those regulated by heat shock, metals, steroids, antibiotics, or alcohol. Non-limiting examples of tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase-1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM- 2 promoter, INF-β promoter, Mb promoter, Nphsl promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter. The promoter sequence can be wild type or it can be modified for more efficient or efficacious expression. In one exemplary embodiment, the encoding DNA can be operably linked to a CMV promoter for constitutive expression in mammalian cells.
[0058] In certain embodiments, the sequence encoding the RNA-guided endonuclease or fusion protein can be operably linked to a promoter sequence that is recognized by a phage RNA polymerase for in vitro mRNA synthesis. In such
embodiments, the in w ro-tran scribed RNA can be purified for use in the methods detailed below in sections (IV) and (V). For example, the promoter sequence can be a T7, T3, or SP6 promoter sequence or a variation of a T7, T3, or SP6 promoter sequence. In an exemplary embodiment, the DNA encoding the fusion protein is operably linked to a T7 promoter for in vitro mRNA synthesis using T7 RNA polymerase.
[0059] In alternate embodiments, the sequence encoding the RNA-guided endonuclease or fusion protein can be operably linked to a promoter sequence for in vitro expression of the RNA-guided endonuclease or fusion protein in bacterial or eukaryotic cells. In such embodiments, the expressed protein can be purified for use in the methods detailed below in sections (IV) and (V). Suitable bacterial promoters include, without limit, T7 promoters, lac operon promoters, trp promoters, variations thereof, and combinations thereof. An exemplary bacterial promoter is tac which is a hybrid of trp and lac promoters. Non-limiting examples of suitable eukaryotic promoters are listed above.
[0060] In additional aspects, the DNA encoding the RNA-guided
endonuclease or fusion protein also can be linked to a polyadenylation signal (e.g., SV40 polyA signal, bovine growth hormone (BGH) polyA signal, etc.) and/or at least one transcriptional termination sequence. Additionally, the sequence encoding the RNA- guided endonuclease or fusion protein also can be linked to sequence encoding at least one nuclear localization signal, at least one cell-penetrating domain, and/or at least one marker domain, which are detailed above in section (I).
[0061 ] In various embodiments, the DNA encoding the RNA-guided endonuclease or fusion protein can be present in a vector. Suitable vectors include plasmid vectors, phagemids, cosmids, artificial/mini-chromosomes, transposons, and viral vectors (e.g., lentiviral vectors, adeno-associated viral vectors, etc.). In one embodiment, the DNA encoding the RNA-guided endonuclease or fusion protein is present in a plasmid vector. Non-limiting examples of suitable plasmid vectors include pUC, pBR322, pET, pBluescript, and variants thereof. The vector can comprise additional expression control sequences (e.g., enhancer sequences, Kozak sequences, polyadenylation sequences, transcriptional termination sequences, etc.), selectable marker sequences (e.g., antibiotic resistance genes), origins of replication, and the like. Additional information can be found in "Current Protocols in Molecular Biology" Ausubel et al., John Wiley & Sons, New York, 2003 or "Molecular Cloning: A Laboratory Manual" Sambrook & Russell, Cold Spring Harbor Press, Cold Spring Harbor, NY, 3rd edition, 2001 .
[0062] In some embodiments, the expression vector comprising the sequence encoding the RNA-guided endonuclease or fusion protein can further comprise sequence encoding a guide RNA. The sequence encoding the guide RNA generally is operably linked to at least one transcriptional control sequence for expression of the guide RNA in the cell or embryo of interest. For example, DNA encoding the guide RNA can be operably linked to a promoter sequence that is recognized by RNA polymerase III (Pol III). Examples of suitable Pol III promoters include, but are not limited to, mammalian U6, U3, H1 , and 7SL RNA promoters. (IV) Method for Modifying a Chromosomal Sequence Using an RNA-Guided Endonuclease
[0063] Another aspect of the present disclosure encompasses a method for modifying a chromosomal sequence in a eukaryotic cell or embryo. The method comprises introducing into a eukaryotic cell or embryo (i) at least one RNA-guided endonuclease comprising at least one nuclear localization signal or nucleic acid encoding at least one RNA-guided endonuclease comprising at least one nuclear localization signal, (ii) at least one guide RNA or DNA encoding at least one guide RNA, and, optionally, (iii) at least one donor polynucleotide comprising a donor sequence. The method further comprises culturing the cell or embryo such that each guide RNA directs an RNA-guided endonuclease to a targeted site in the chromosomal sequence where the RNA-guided endonuclease introduces a double-stranded break in the targeted site, and the double-stranded break is repaired by a DNA repair process such that the chromosomal sequence is modified.
[0064] In some embodiments, the method can comprise introducing one RNA-guided endonuclease (or encoding nucleic acid) and one guide RNA (or encoding DNA) into a cell or embryo, wherein the RNA-guided endonuclease introduces one double-stranded break in the targeted chromosomal sequence. In embodiments in which the optional donor polynucleotide is not present, the double-stranded break in the chromosomal sequence can be repaired by a non-homologous end-joining (NHEJ) repair process. Because NHEJ is error-prone, deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. Accordingly, the targeted chromosomal sequence can be modified or inactivated. For example, a single nucleotide change (SNP) can give rise to an altered protein product, or a shift in the reading frame of a coding sequence can inactivate or "knock out" the sequence such that no protein product is made. In embodiments in which the optional donor
polynucleotide is present, the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence at the targeted site during repair of the double-stranded break. For example, in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted site in the chromosomal sequence, the donor sequence can be exchanged with or integrated into the chromosomal sequence at the targeted site during repair mediated by homology-directed repair process. Alternatively, in embodiments in which the donor sequence is flanked by compatible overhangs (or the compatible overhangs are generated in situ by the RNA-guided endonuclease) the donor sequence can be ligated directly with the cleaved chromosomal sequence by a non-homologous repair process during repair of the double-stranded break. Exchange or integration of the donor sequence into the chromosomal sequence modifies the targeted chromosomal sequence or introduces an exogenous sequence into the chromosomal sequence of the cell or embryo.
[0065] In other embodiments, the method can comprise introducing two RNA-guided endonucleases (or encoding nucleic acid) and two guide RNAs (or encoding DNA) into a cell or embryo, wherein the RNA-guided endonucleases introduce two double-stranded breaks in the chromosomal sequence. See FIG. 3B. The two breaks can be within several base pairs, within tens of base pairs, or can be separated by many thousands of base pairs. In embodiments in which the optional donor polynucleotide is not present, the resultant double-stranded breaks can be repaired by a non-homologous repair process such that the sequence between the two cleavage sites is lost and/or deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break(s). In embodiments in which the optional donor polynucleotide is present, the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded breaks by either a homology-based repair process (e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence) or a non-homologous repair process (e.g., in embodiments in which the donor sequence is flanked by compatible overhangs). [0066] In still other embodiments, the method can comprise introducing one RNA-guided endonuclease modified to cleave one strand of a double-stranded sequence (or encoding nucleic acid) and two guide RNAs (or encoding DNA) into a cell or embryo, wherein each guide RNA directs the RNA-guided endonuclease to a specific target site, at which site the modified endonuclease cleaves one strand (i.e., nicks) of the double-stranded chromosomal sequence, and wherein the two nicks are in opposite stands and in close enough proximity to constitute a double-stranded break. See FIG. 3A. In embodiments in which the optional donor polynucleotide is not present, the resultant double-stranded break can be repaired by a non-homologous repair process such that deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. In embodiments in which the optional donor polynucleotide is present, the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded break by either a homology-based repair process (e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence) or a non-homologous repair process (e.g., in embodiments in which the donor sequence is flanked by compatible overhangs).
(a) RNA-guided endonuclease
[0067] The method comprises introducing into a cell or embryo at least one RNA-guided endonuclease comprising at least one nuclear localization signal or nucleic acid encoding at least one RNA-guided endonuclease comprising at least one nuclear localization signal. Such RNA-guided endonucleases and nucleic acids encoding RNA-guided endonucleases are described above in sections (I) and (III), respectively.
[0068] In some embodiments, the RNA-guided endonuclease can be introduced into the cell or embryo as an isolated protein. In such embodiments, the RNA-guided endonuclease can further comprise at least one cell-penetrating domain, which facilitates cellular uptake of the protein. In other embodiments, the RNA-guided endonuclease can be introduced into the cell or embryo as an mRNA molecule. In still other embodiments, the RNA-guided endonuclease can be introduced into the cell or embryo as a DNA molecule. In general, DNA sequence encoding the fusion protein is operably linked to a promoter sequence that will function in the cell or embryo of interest. The DNA sequence can be linear, or the DNA sequence can be part of a vector. In still other embodiments, the fusion protein can be introduced into the cell or embryo as an RNA-protein complex comprising the fusion protein and the guide RNA.
[0069] In alternate embodiments, DNA encoding the RNA-guided endonuclease can further comprise sequence encoding a guide RNA. In general, each of the sequences encoding the RNA-guided endonuclease and the guide RNA is operably linked to appropriate promoter control sequence that allows expression of the RNA-guided endonuclease and the guide RNA, respectively, in the cell or embryo. The DNA sequence encoding the RNA-guided endonuclease and the guide RNA can further comprise additional expression control, regulatory, and/or processing sequence(s). The DNA sequence encoding the RNA-guided endonuclease and the guide RNA can be linear or can be part of a vector
(b) Guide RNA
[0070] The method also comprises introducing into a cell or embryo at least one guide RNA or DNA encoding at least one guide RNA. A guide RNA interacts with the RNA-guided endonuclease to direct the endonuclease to a specific target site, at which site the 5' end of the guide RNA base pairs with a specific protospacer sequence in the chromosomal sequence.
[0071 ] Each guide RNA comprises three regions: a first region at the 5' end that is complementary to the target site in the chromosomal sequence, a second internal region that forms a stem loop structure, and a third 3' region that remains essentially single-stranded. The first region of each guide RNA is different such that each guide RNA guides a fusion protein to a specific target site. The second and third regions of each guide RNA can be the same in all guide RNAs.
[0072] The first region of the guide RNA is complementary to sequence (i.e., protospacer sequence) at the target site in the chromosomal sequence such that the first region of the guide RNA can base pair with the target site. In various embodiments, the first region of the guide RNA can comprise from about 10 nucleotides to more than about 25 nucleotides. For example, the region of base pairing between the first region of the guide RNA and the target site in the chromosomal sequence can be about 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 22, 23, 24, 25, or more than 25 nucleotides in length. In an exemplary embodiment, the first region of the guide RNA is about 19, 20, or 21 nucleotides in length.
[0073] The guide RNA also comprises a second region that forms a secondary structure. In some embodiments, the secondary structure comprises a stem (or hairpin) and a loop. The length of the loop and the stem can vary. For example, the loop can range from about 3 to about 10 nucleotides in length, and the stem can range from about 6 to about 20 base pairs in length. The stem can comprise one or more bulges of 1 to about 10 nucleotides. Thus, the overall length of the second region can range from about 16 to about 60 nucleotides in length. In an exemplary embodiment, the loop is about 4 nucleotides in length and the stem comprises about 12 base pairs.
[0074] The guide RNA also comprises a third region at the 3' end that remains essentially single-stranded. Thus, the third region has no complementarity to any chromosomal sequence in the cell of interest and has no complementarity to the rest of the guide RNA. The length of the third region can vary. In general, the third region is more than about 4 nucleotides in length. For example, the length of the third region can range from about 5 to about 60 nucleotides in length.
[0075] The combined length of the second and third regions (also called the universal or scaffold region) of the guide RNA can range from about 30 to about 120 nucleotides in length. In one aspect, the combined length of the second and third regions of the guide RNA range from about 70 to about 100 nucleotides in length.
[0076] In some embodiments, the guide RNA comprises a single molecule comprising all three regions. In other embodiments, the guide RNA can comprise two separate molecules. The first RNA molecule can comprise the first region of the guide RNA and one half of the "stem" of the second region of the guide RNA. The second RNA molecule can comprise the other half of the "stem" of the second region of the guide RNA and the third region of the guide RNA. Thus, in this embodiment, the first and second RNA molecules each contain a sequence of nucleotides that are complementary to one another. For example, in one embodiment, the first and second RNA molecules each comprise a sequence (of about 6 to about 20 nucleotides) that base pairs to the other sequence to form a functional guide RNA.
[0077] In some embodiments, the guide RNA can be introduced into the cell or embryo as a RNA molecule. The RNA molecule can be transcribed in vitro. Alternatively, the RNA molecule can be chemically synthesized.
[0078] In other embodiments, the guide RNA can be introduced into the cell or embryo as a DNA molecule. In such cases, the DNA encoding the guide RNA can be operably linked to promoter control sequence for expression of the guide RNA in the cell or embryo of interest. For example, the RNA coding sequence can be operably linked to a promoter sequence that is recognized by RNA polymerase III (Pol III).
Examples of suitable Pol III promoters include, but are not limited to, mammalian U6 or H1 promoters. In exemplary embodiments, the RNA coding sequence is linked to a mouse or human U6 promoter. In other exemplary embodiments, the RNA coding sequence is linked to a mouse or human H1 promoter.
[0079] The DNA molecule encoding the guide RNA can be linear or circular. In some embodiments, the DNA sequence encoding the guide RNA can be part of a vector. Suitable vectors include plasmid vectors, phagemids, cosmids, artificial/mini-chromosomes, transposons, and viral vectors. In an exemplary
embodiment, the DNA encoding the RNA-guided endonuclease is present in a plasmid vector. Non-limiting examples of suitable plasmid vectors include pUC, pBR322, pET, pBluescript, and variants thereof. The vector can comprise additional expression control sequences (e.g., enhancer sequences, Kozak sequences, polyadenylation sequences, transcriptional termination sequences, etc.), selectable marker sequences (e.g., antibiotic resistance genes), origins of replication, and the like.
[0080] In embodiments in which both the RNA-guided endonuclease and the guide RNA are introduced into the cell as DNA molecules, each can be part of a separate molecule (e.g., one vector containing fusion protein coding sequence and a second vector containing guide RNA coding sequence) or both can be part of the same molecule (e.g., one vector containing coding (and regulatory) sequence for both the fusion protein and the guide RNA).
(c) Target site
[0081 ] An RNA-guided endonuclease in conjunction with a guide RNA is directed to a target site in the chromosomal sequence, wherein the RNA-guided endonuclease introduces a double-stranded break in the chromosomal sequence. The target site has no sequence limitation except that the sequence is immediately followed (downstream) by a consensus sequence. This consensus sequence is also known as a P/otospacer adjacent motif (PAM). Examples of PAM include, but are not limited to, NGG, NGGNG, and NNAGAAW (wherein N is defined as any nucleotide and W is defined as either A or T). As detailed above in section (IV)(b), the first region (at the 5' end) of the guide RNA is complementary to the protospacer of the target sequence. Typically, the first region of the guide RNA is about 19 to 21 nucleotides in length.
Thus, in certain aspects, the sequence of the target site in the chromosomal sequence is 5'-N19.2i-/VGG-3'. The PAM is in italics.
[0082] The target site can be in the coding region of a gene, in an intron of a gene, in a control region of a gene, in a non-coding region between genes, etc. The gene can be a protein coding gene or an RNA coding gene. The gene can be any gene of interest.
(d) Optional donor polynucleotide
[0083] In some embodiments, the method further comprises introducing at least one donor polynucleotide into the embryo. A donor polynucleotide comprises at least one donor sequence. In some aspects, a donor sequence of the donor
polynucleotide corresponds to an endogenous or native chromosomal sequence. For example, the donor sequence can be essentially identical to a portion of the
chromosomal sequence at or near the targeted site, but which comprises at least one nucleotide change. Thus, the donor sequence can comprise a modified version of the wild type sequence at the targeted site such that, upon integration or exchange with the native sequence, the sequence at the targeted chromosomal location comprises at least one nucleotide change. For example, the change can be an insertion of one or more nucleotides, a deletion of one or more nucleotides, a substitution of one or more nucleotides, or combinations thereof. As a consequence of the integration of the modified sequence, the cell or embryo/animal can produce a modified gene product from the targeted chromosomal sequence.
[0084] In other aspects, the donor sequence of the donor polynucleotide corresponds to an exogenous sequence. As used herein, an "exogenous" sequence refers to a sequence that is not native to the cell or embryo, or a sequence whose native location in the genome of the cell or embryo is in a different location. For example, the exogenous sequence can comprise protein coding sequence, which can be operably linked to an exogenous promoter control sequence such that, upon integration into the genome, the cell or embryo/animal is able to express the protein coded by the integrated sequence. Alternatively, the exogenous sequence can be integrated into the chromosomal sequence such that its expression is regulated by an endogenous promoter control sequence. In other iterations, the exogenous sequence can be a transcriptional control sequence, another expression control sequence, an RNA coding sequence, and so forth. Integration of an exogenous sequence into a chromosomal sequence is termed a "knock in."
[0085] As can be appreciated by those skilled in the art, the length of the donor sequence can and will vary. For example, the donor sequence can vary in length from several nucleotides to hundreds of nucleotides to hundreds of thousands of nucleotides.
[0086] Donor polynucleotide comprising upstream and downstream sequences. In some embodiments, the donor sequence in the donor polynucleotide is flanked by an upstream sequence and a downstream sequence, which have substantial sequence identity to sequences located upstream and downstream, respectively, of the targeted site in the chromosomal sequence. Because of these sequence similarities, the upstream and downstream sequences of the donor polynucleotide permit
homologous recombination between the donor polynucleotide and the targeted chromosomal sequence such that the donor sequence can be integrated into (or exchanged with) the chromosomal sequence. [0087] The upstream sequence, as used herein, refers to a nucleic acid sequence that shares substantial sequence identity with a chromosomal sequence upstream of the targeted site. Similarly, the downstream sequence refers to a nucleic acid sequence that shares substantial sequence identity with a chromosomal sequence downstream of the targeted site. As used herein, the phrase "substantial sequence identity" refers to sequences having at least about 75% sequence identity. Thus, the upstream and downstream sequences in the donor polynucleotide can have about 75%, 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with sequence upstream or downstream to the targeted site. In an exemplary embodiment, the upstream and downstream sequences in the donor polynucleotide can have about 95% or 100% sequence identity with chromosomal sequences upstream or downstream to the targeted site. In one embodiment, the upstream sequence shares substantial sequence identity with a chromosomal sequence located immediately upstream of the targeted site (i.e., adjacent to the targeted site). In other embodiments, the upstream sequence shares substantial sequence identity with a chromosomal sequence that is located within about one hundred (100) nucleotides upstream from the targeted site. Thus, for example, the upstream sequence can share substantial sequence identity with a chromosomal sequence that is located about 1 to about 20, about 21 to about 40, about 41 to about 60, about 61 to about 80, or about 81 to about 100 nucleotides upstream from the targeted site. In one embodiment, the downstream sequence shares substantial sequence identity with a chromosomal sequence located immediately downstream of the targeted site (i.e., adjacent to the targeted site). In other
embodiments, the downstream sequence shares substantial sequence identity with a chromosomal sequence that is located within about one hundred (100) nucleotides downstream from the targeted site. Thus, for example, the downstream sequence can share substantial sequence identity with a chromosomal sequence that is located about 1 to about 20, about 21 to about 40, about 41 to about 60, about 61 to about 80, or about 81 to about 100 nucleotides downstream from the targeted site.
[0088] Each upstream or downstream sequence can range in length from about 20 nucleotides to about 5000 nucleotides. In some embodiments, upstream and downstream sequences can comprise about 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1 100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2100, 2200, 2300, 2400, 2500, 2600, 2800, 3000, 3200, 3400, 3600, 3800, 4000, 4200, 4400, 4600, 4800, or 5000 nucleotides. In exemplary embodiments, upstream and
downstream sequences can range in length from about 50 to about 1500 nucleotides.
[0089] Donor polynucleotides comprising the upstream and downstream sequences with sequence similarity to the targeted chromosomal sequence can be linear or circular. In embodiments in which the donor polynucleotide is circular, it can be part of a vector. For example, the vector can be a plasmid vector.
[0090] Donor polynucleotide comprising targeted cleavage site(s). In other embodiments, the donor polynucleotide can additionally comprise at least one targeted cleavage site that is recognized by the RNA-guided endonuclease. The targeted cleavage site added to the donor polynucleotide can be placed upstream or
downstream or both upstream and downstream of the donor sequence. For example, the donor sequence can be flanked by targeted cleavage sites such that, upon cleavage by the RNA-guided endonuclease, the donor sequence is flanked by overhangs that are compatible with those in the chromosomal sequence generated upon cleavage by the RNA-guided endonuclease. Accordingly, the donor sequence can be ligated with the cleaved chromosomal sequence during repair of the double stranded break by a nonhomologous repair process. Generally, donor polynucleotides comprising the targeted cleavage site(s) will be circular (e.g., can be part of a plasmid vector).
[0091 ] Donor polynucleotide comprising a short donor sequence with optional overhangs. In still alternate embodiments, the donor polynucleotide can be a linear molecule comprising a short donor sequence with optional short overhangs that are compatible with the overhangs generated by the RNA-guided endonuclease. In such embodiments, the donor sequence can be ligated directly with the cleaved chromosomal sequence during repair of the double-stranded break. In some instances, the donor sequence can be less than about 1 ,000, less than about 500, less than about 250, or less than about 100 nucleotides. In certain cases, the donor polynucleotide can be a linear molecule comprising a short donor sequence with blunt ends. In other iterations, the donor polynucleotide can be a linear molecule comprising a short donor sequence with 5' and/or 3' overhangs. The overhangs can comprise 1 , 2, 3, 4, or 5 nucleotides.
[0092] Typically, the donor polynucleotide will be DNA. The DNA may be single-stranded or double-stranded and/or linear or circular. The donor polynucleotide may be a DNA plasmid, a bacterial artificial chromosome (BAC), a yeast artificial chromosome (YAC), a viral vector, a linear piece of DNA, a PCR fragment, a naked nucleic acid, or a nucleic acid complexed with a delivery vehicle such as a liposome or poloxamer. In certain embodiments, the donor polynucleotide comprising the donor sequence can be part of a plasmid vector. In any of these situations, the donor polynucleotide comprising the donor sequence can further comprise at least one additional sequence.
(e) Introducing into the cell or embryo
[0093] The RNA-targeted endonuclease(s) (or encoding nucleic acid), the guide RNA(s) (or encoding DNA), and the optional donor polynucleotide(s) can be introduced into a cell or embryo by a variety of means. In some embodiments, the cell or embryo is transfected. Suitable transfection methods include calcium phosphate- mediated transfection, nucleofection (or electroporation), cationic polymer transfection (e.g., DEAE-dextran or polyethylenimine), viral transduction, virosome transfection, virion transfection, liposome transfection, cationic liposome transfection,
immunoliposome transfection, nonliposomal lipid transfection, dendrimer transfection, heat shock transfection, magnetofection, lipofection, gene gun delivery, impalefection, sonoporation, optical transfection, and proprietary agent-enhanced uptake of nucleic acids. Transfection methods are well known in the art {see, e.g., "Current Protocols in Molecular Biology" Ausubel et al., John Wiley & Sons, New York, 2003 or "Molecular Cloning: A Laboratory Manual" Sambrook & Russell, Cold Spring Harbor Press, Cold Spring Harbor, NY, 3rd edition, 2001 ). In other embodiments, the molecules are introduced into the cell or embryo by microinjection. Typically, the embryo is a fertilized one-cell stage embryo of the species of interest. For example, the molecules can be injected into the pronuclei of one cell embryos. [0094] The RNA-targeted endonuclease(s) (or encoding nucleic acid), the guide RNA(s) (or DNAs encoding the guide RNA), and the optional donor
polynucleotide(s) can be introduced into the cell or embryo simultaneously or
sequentially. The ratio of the RNA-targeted endonuclease(s) (or encoding nucleic acid) to the guide RNA(s) (or encoding DNA) generally will be about stoichiometric such that they can form an RNA-protein complex. In one embodiment, DNA encoding an RNA- targeted endonuclease and DNA encoding a guide RNA are delivered together within the plasmid vector.
(f) Culturing the cell or embryo
[0095] The method further comprises maintaining the cell or embryo under appropriate conditions such that the guide RNA(s) directs the RNA-guided
endonuclease(s) to the targeted site(s) in the chromosomal sequence, and the RNA- guided endonuclease(s) introduce at least one double-stranded break in the
chromosomal sequence. A double-stranded break can be repaired by a DNA repair process such that the chromosomal sequence is modified by a deletion of at least one nucleotide, an insertion of at least one nucleotide, a substitution of at least one nucleotide, or a combination thereof.
[0096] In embodiments in which no donor polynucleotide is introduced into the cell or embryo, the double-stranded break can be repaired via a non-homologous end-joining (NHEJ) repair process. Because NHEJ is error-prone, deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break.
Accordingly, the sequence at the chromosomal sequence can be modified such that the reading frame of a coding region can be shifted and that the chromosomal sequence is inactivated or "knocked out." An inactivated protein-coding chromosomal sequence does not give rise to the protein coded by the wild type chromosomal sequence.
[0097] In embodiments in which a donor polynucleotide comprising upstream and downstream sequences is introduced into the cell or embryo, the double- stranded break can be repaired by a homology-directed repair (HDR) process such that the donor sequence is integrated into the chromosomal sequence. Accordingly, an exogenous sequence can be integrated into the genome of the cell or embryo, or the targeted chromosomal sequence can be modified by exchange of a modified sequence for the wild type chromosomal sequence.
[0098] In embodiments in which a donor polynucleotide comprising the targeted cleave site is introduced into the cell or embryo, the RNA-guided endonuclease can cleave both the targeted chromosomal sequence and the donor polynucleotide. The linearized donor polynucleotide can be integrated into the chromosomal sequence at the site of the double-stranded break by ligation between the donor polynucleotide and the cleaved chromosomal sequence via a NHEJ process.
[0099] In embodiments in which a linear donor polynucleotide comprising a short donor sequence is introduced into the cell or embryo, the short donor sequence can be integrated into the chromosomal sequence at the site of the double-stranded break via a NHEJ process. The integration can proceed via the ligation of blunt ends between the short donor sequence and the chromosomal sequence at the site of the double stranded break. Alternatively, the integration can proceed via the ligation of sticky ends (i.e., having 5' or 3' overhangs) between a short donor sequence that is flanked by overhangs that are compatible with those generated by the RNA-targeting endonuclease in the cleaved chromosomal sequence.
[0100] In general, the cell is maintained under conditions appropriate for cell growth and/or maintenance. Suitable cell culture conditions are well known in the art and are described, for example, in Santiago et al. (2008) PNAS 105:5809-5814; Moehle et al. (2007) PNAS 104:3055-3060; Urnov et al. (2005) Nature 435:646-651 ; and Lombardo et al (2007) Nat. Biotechnology 25:1298-1306. Those of skill in the art appreciate that methods for culturing cells are known in the art and can and will vary depending on the cell type. Routine optimization may be used, in all cases, to determine the best techniques for a particular cell type.
[0101 ] An embryo can be cultured in vitro (e.g., in cell culture). Typically, the embryo is cultured at an appropriate temperature and in appropriate media with the necessary 02/C02 ratio to allow the expression of the RNA endonuclease and guide RNA, if necessary. Suitable non-limiting examples of media include M2, M16, KSOM, BMOC, and HTF media. A skilled artisan will appreciate that culture conditions can and will vary depending on the species of embryo. Routine optimization may be used, in all cases, to determine the best culture conditions for a particular species of embryo. In some cases, a cell line may be derived from an in vitro-cultured embryo (e.g., an embryonic stem cell line).
[0102] Alternatively, an embryo may be cultured in vivo by transferring the embryo into the uterus of a female host. Generally speaking the female host is from the same or similar species as the embryo. Preferably, the female host is pseudo-pregnant. Methods of preparing pseudo-pregnant female hosts are known in the art. Additionally, methods of transferring an embryo into a female host are known. Culturing an embryo in vivo permits the embryo to develop and can result in a live birth of an animal derived from the embryo. Such an animal would comprise the modified chromosomal sequence in every cell of the body.
(g) Cell and embryo types
[0103] A variety of eukaryotic cells and embryos are suitable for use in the method. For example, the cell can be a human cell, a non-human mammalian cell, a non-mammalian vertebrate cell, an invertebrate cell, an insect cell, a plant cell, a yeast cell, or a single cell eukaryotic organism. In general, the embryo is non-human mammalian embryo. In specific embodiments, the embryos can be a one cell non- human mammalian embryo. Exemplary mammalian embryos, including one cell embryos, include without limit mouse, rat, hamster, rodent, rabbit, feline, canine, ovine, porcine, bovine, equine, and primate embryos. In still other embodiments, the cell can be a stem cell. Suitable stem cells include without limit embryonic stem cells, ES-like stem cells, fetal stem cells, adult stem cells, pluripotent stem cells, induced pluripotent stem cells, multipotent stem cells, oligopotent stem cells, unipotent stem cells and others. In exemplary embodiments, the cell is a mammalian cell.
[0104] Non-limiting examples of suitable mammalian cells include Chinese hamster ovary (CHO) cells, baby hamster kidney (BHK) cells; mouse myeloma NS0 cells, mouse embryonic fibroblast 3T3 cells (NIH3T3), mouse B lymphoma A20 cells; mouse melanoma B16 cells; mouse myoblast C2C12 cells; mouse myeloma SP2/0 cells; mouse embryonic mesenchymal C3H-10T1 /2 cells; mouse carcinoma CT26 cells, mouse prostate DuCuP cells; mouse breast EMT6 cells; mouse hepatoma Hepa1 c1 c7 cells; mouse myeloma J5582 cells; mouse epithelial MTD-1 A cells; mouse myocardial MyEnd cells; mouse renal RenCa cells; mouse pancreatic RIN-5F cells; mouse melanoma X64 cells; mouse lymphoma YAC-1 cells; rat glioblastoma 9L cells; rat B lymphoma RBL cells; rat neuroblastoma B35 cells; rat hepatoma cells (HTC); buffalo rat liver BRL 3A cells; canine kidney cells (MDCK); canine mammary (CMT) cells; rat osteosarcoma D17 cells; rat monocyte/macrophage DH82 cells; monkey kidney SV-40 transformed fibroblast (COS7) cells; monkey kidney CVI-76 cells; African green monkey kidney (VERO-76) cells; human embryonic kidney cells (HEK293, HEK293T); human cervical carcinoma cells (HELA); human lung cells (W138); human liver cells (Hep G2); human U2-OS osteosarcoma cells, human A549 cells, human A-431 cells, and human K562 cells. An extensive list of mammalian cell lines may be found in the American Type Culture Collection catalog (ATCC, Mamassas, VA).
(V) Method for Using a Fusion Protein to Modify a Chromosomal Sequence or Regulate Expression of a Chromosomal Sequence
[0105] Another aspect of the present disclosure encompasses a method for modifying a chromosomal sequence or regulating expression of a chromosomal sequence in a cell or embryo. The method comprises introducing into the cell or embryo (a) at least one fusion protein or nucleic acid encoding at least one fusion protein, wherein the fusion protein comprises a CRISPR/Cas-like protein or a fragment thereof and an effector domain, and (b) at least one guide RNA or DNA encoding the guide RNA, wherein the guide RNA guides the CRISPR/Cas-like protein of the fusion protein to a targeted site in the chromosomal sequence and the effector domain of the fusion protein modifies the chromosomal sequence or regulates expression of the chromosomal sequence.
[0106] Fusion proteins comprising a CRISPR/Cas-like protein or a fragment thereof and an effector domain are detailed above in section (II). In general, the fusion proteins disclosed herein further comprise at least one nuclear localization signal. Nucleic acids encoding fusion proteins are described above in section (III). In some embodiments, the fusion protein can be introduced into the cell or embryo as an isolated protein (which can further comprise a cell-penetrating domain). Furthermore, the isolated fusion protein can be part of a protein-RNA complex comprising the guide RNA. In other embodiments, the fusion protein can be introduced into the cell or embryo as a RNA molecule (which can be capped and/or polyadenylated). In still other embodiments, the fusion protein can be introduced into the cell or embryo as a DNA molecule. For example, the fusion protein and the guide RNA can be introduced into the cell or embryo as discrete DNA molecules or as part of the same DNA molecule. Such DNA molecules can be plasmid vectors.
[0107] In some embodiments, the method further comprises introducing into the cell or embryo at least one zinc finger nuclease. Zinc finger nucleases are described above in section (ll)(d). In still other embodiments, the method further comprises introducing into the cell or embryo at least one donor polynucleotide. Donor polynucleotides are detailed above in section (IV)(d). Means for introducing molecules into cells or embryos, as well as means for culturing cell or embryos are described above in sections (IV)(e) and (IV)(f), respectively. Suitable cells and embryos are described above in section (IV)(g).
[0108] In certain embodiments in which the effector domain of the fusion protein is a cleavage domain (e.g., a Fokl cleavage domain or a modified Fokl cleavage domain), the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein) and two guide RNAs (or DNA encoding two guide RNAs). The two guide RNAs direct the fusion protein to two different target sites in the chromosomal sequence, wherein the fusion protein dimerizes (e.g., form a homodimer) such that the two cleavage domains can introduce a double stranded break into the chromosomal sequence. See FIG. 1A. In embodiments in which the optional donor polynucleotide is not present, the double-stranded break in the chromosomal sequence can be repaired by a non-homologous end-joining (NHEJ) repair process. Because NHEJ is error-prone, deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. Accordingly, the targeted chromosomal sequence can be modified or inactivated. For example, a single nucleotide change (SNP) can give rise to an altered protein product, or a shift in the reading frame of a coding sequence can inactivate or "knock out" the sequence such that no protein product is made. In embodiments in which the optional donor polynucleotide is present, the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence at the targeted site during repair of the double-stranded break. For example, in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted site in the chromosomal sequence, the donor sequence can be exchanged with or integrated into the
chromosomal sequence at the targeted site during repair mediated by homology- directed repair process. Alternatively, in embodiments in which the donor sequence is flanked by compatible overhangs (or the compatible overhangs are generated in situ by the RNA-guided endonuclease) the donor sequence can be ligated directly with the cleaved chromosomal sequence by a non-homologous repair process during repair of the double-stranded break. Exchange or integration of the donor sequence into the chromosomal sequence modifies the targeted chromosomal sequence or introduces an exogenous sequence into the chromosomal sequence of the cell or embryo.
[0109] In other embodiments in which the effector domain of the fusion protein is a cleavage domain (e.g., a Fokl cleavage domain or a modified Fokl cleavage domain), the method can comprise introducing into the cell or embryo two different fusion proteins (or nucleic acid encoding two different fusion proteins) and two guide RNAs (or DNA encoding two guide RNAs). The fusion proteins can differ as detailed above in section (II). Each guide RNA directs a fusion protein to a specific target site in the chromosomal sequence, wherein the fusion proteins dimerize (e.g., form a heterodimer) such that the two cleavage domains can introduce a double stranded break into the chromosomal sequence. In embodiments in which the optional donor polynucleotide is not present, the resultant double-stranded breaks can be repaired by a non-homologous repair process such that deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. In embodiments in which the optional donor polynucleotide is present, the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded break by either a homology-based repair process (e.g., in embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence) or a nonhomologous repair process (e.g., in embodiments in which the donor sequence is flanked by compatible overhangs).
[01 10] In still other embodiments in which the effector domain of the fusion protein is a cleavage domain (e.g., a Fokl cleavage domain or a modified Fokl cleavage domain), the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein), one guide RNA (or DNA encoding one guide RNA), and one zinc finger nuclease (or nucleic acid encoding the zinc finger nuclease), wherein the zinc finger nuclease comprises a Fokl cleavage domain or a modified Fokl cleavage domain. The guide RNA directs the fusion protein to a specific chromosomal sequence, and the zinc finger nuclease is directed to another
chromosomal sequence, wherein the fusion protein and the zinc finger nuclease dimerize such that the cleavage domain of the fusion protein and the cleavage domain of the zinc finger nuclease can introduce a double stranded break into the chromosomal sequence. See FIG. 1 B. In embodiments in which the optional donor polynucleotide is not present, the resultant double-stranded breaks can be repaired by a non-homologous repair process such that deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. In embodiments in which the optional donor
polynucleotide is present, the donor sequence in the donor polynucleotide can be exchanged with or integrated into the chromosomal sequence during repair of the double-stranded break by either a homology-based repair process (e.g., in
embodiments in which the donor sequence is flanked by upstream and downstream sequences having substantial sequence identity with upstream and downstream sequences, respectively, of the targeted sites in the chromosomal sequence) or a nonhomologous repair process (e.g., in embodiments in which the donor sequence is flanked by compatible overhangs). [01 1 1 ] In still other embodiments in which the effector domain of the fusion protein is a transcriptional activation domain or a transcriptional repressor domain, the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein) and one guide RNA (or DNA encoding one guide RNA). The guide RNA directs the fusion protein to a specific chromosomal sequence, wherein the transcriptional activation domain or a transcriptional repressor domain activates or represses expression, respectively, of the targeted chromosomal sequence. See FIG. 2A.
[01 12] In alternate embodiments in which the effector domain of the fusion protein is an epigenetic modification domain, the method can comprise introducing into the cell or embryo one fusion protein (or nucleic acid encoding one fusion protein) and one guide RNA (or DNA encoding one guide RNA). The guide RNA directs the fusion protein to a specific chromosomal sequence, wherein the epigenetic modification domain modifies the structure of the targeted the chromosomal sequence. See FIG. 2A. Epigenetic modifications include acetylation, methylation of histone proteins and/or nucleotide methylation. In some instances, structural modification of the chromosomal sequence leads to changes in expression of the chromosomal sequence.
(VI) Genetically Modified Cells and Animals
[01 13] The present disclosure encompasses genetically modified cells, non-human embryos, and non-human animals comprising at least one chromosomal sequence that has been modified using an RNA-guided endonuclease-mediated or fusion protein-mediated process, for example, using the methods described herein. The disclosure provides cells comprising at least one DNA or RNA molecule encoding an RNA-guided endonuclease or fusion protein targeted to a chromosomal sequence of interest or a fusion protein, at least one guide RNA, and optionally one or more donor polynucleotide(s). The disclosure also provides non-human embryos comprising at least one DNA or RNA molecule encoding an RNA-guided endonuclease or fusion protein targeted to a chromosomal sequence of interest, at least one guide RNA, and optionally one or more donor polynucleotide(s). [01 14] The present disclosure provides genetically modified non-human animals, non-human embryos, or animal cells comprising at least one modified chromosomal sequence. The modified chromosomal sequence may be modified such that it is (1 ) inactivated, (2) has an altered expression or produces an altered protein product, or (3) comprises an integrated sequence. The chromosomal sequence is modified with an RNA guided endonuclease-mediated or fusion protein-mediated process, using the methods described herein.
[01 15] As discussed, one aspect of the present disclosure provides a genetically modified animal in which at least one chromosomal sequence has been modified. In one embodiment, the genetically modified animal comprises at least one inactivated chromosomal sequence. The modified chromosomal sequence may be inactivated such that the sequence is not transcribed and/or a functional protein product is not produced. Thus, a genetically modified animal comprising an inactivated chromosomal sequence may be termed a "knock out" or a "conditional knock out." The inactivated chromosomal sequence can include a deletion mutation (i.e., deletion of one or more nucleotides), an insertion mutation (i.e., insertion of one or more nucleotides), or a nonsense mutation (i.e., substitution of a single nucleotide for another nucleotide such that a stop codon is introduced). As a consequence of the mutation, the targeted chromosomal sequence is inactivated and a functional protein is not produced. The inactivated chromosomal sequence comprises no exogenously introduced sequence. Also included herein are genetically modified animals in which two, three, four, five, six, seven, eight, nine, or ten or more chromosomal sequences are inactivated.
[01 16] In another embodiment, the modified chromosomal sequence can be altered such that it codes for a variant protein product. For example, a genetically modified animal comprising a modified chromosomal sequence can comprise a targeted point mutation(s) or other modification such that an altered protein product is produced. In one embodiment, the chromosomal sequence can be modified such that at least one nucleotide is changed and the expressed protein comprises one changed amino acid residue (missense mutation). In another embodiment, the chromosomal sequence can be modified to comprise more than one missense mutation such that more than one amino acid is changed. Additionally, the chromosomal sequence can be modified to have a three nucleotide deletion or insertion such that the expressed protein comprises a single amino acid deletion or insertion. The altered or variant protein can have altered properties or activities compared to the wild type protein, such as altered substrate specificity, altered enzyme activity, altered kinetic rates, etc.
[01 17] In another embodiment, the genetically modified animal can comprise at least one chromosomally integrated sequence. A genetically modified animal comprising an integrated sequence may be termed a "knock in" or a
"conditional knock in." The chromosomally integrated sequence can, for example, encode an orthologous protein, an endogenous protein, or combinations of both. In one embodiment, a sequence encoding an orthologous protein or an endogenous protein can be integrated into a chromosomal sequence encoding a protein such that the chromosomal sequence is inactivated, but the exogenous sequence is expressed. In such a case, the sequence encoding the orthologous protein or endogenous protein may be operably linked to a promoter control sequence. Alternatively, a sequence encoding an orthologous protein or an endogenous protein may be integrated into a chromosomal sequence without affecting expression of a chromosomal sequence. For example, a sequence encoding a protein can be integrated into a "safe harbor" locus, such as the Rosa26 locus, HPRT locus, or AAV locus. The present disclosure also encompasses genetically modified animals in which two, three, four, five, six, seven, eight, nine, or ten or more sequences, including sequences encoding protein(s), are integrated into the genome.
[01 1 8] The chromosomally integrated sequence encoding a protein can encode the wild type form of a protein of interest or can encode a protein comprising at least one modification such that an altered version of the protein is produced. For example, a chromosomally integrated sequence encoding a protein related to a disease or disorder can comprise at least one modification such that the altered version of the protein produced causes or potentiates the associated disorder.
Alternatively, the chromosomally integrated sequence encoding a protein related to a disease or disorder can comprise at least one modification such that the altered version of the protein protects against the development of the associated disorder.
[01 19] In an additional embodiment, the genetically modified animal can be a "humanized" animal comprising at least one chromosomally integrated
sequence encoding a functional human protein. The functional human protein can have no corresponding ortholog in the genetically modified animal. Alternatively, the wild type animal from which the genetically modified animal is derived may comprise an ortholog corresponding to the functional human protein. In this case, the
orthologous sequence in the "humanized" animal is inactivated such that no functional protein is made and the "humanized" animal comprises at least one chromosomally integrated sequence encoding the human protein.
[0120] In yet another embodiment, the genetically modified animal can comprise at least one modified chromosomal sequence encoding a protein such that the expression pattern of the protein is altered. For example, regulatory regions controlling the expression of the protein, such as a promoter or a transcription factor binding site, can be altered such that the protein is over-produced, or the tissue-specific or temporal expression of the protein is altered, or a combination thereof.
Alternatively, the expression pattern of the protein can be altered using a conditional knockout system. A non-limiting example of a conditional knockout system includes a Cre-lox recombination system. A Cre-lox recombination system comprises a Cre recombinase enzyme, a site-specific DNA recombinase that can catalyze the
recombination of a nucleic acid sequence between specific sites (lox sites) in a nucleic acid molecule. Methods of using this system to produce temporal and tissue specific expression are known in the art. In general, a genetically modified animal is generated with lox sites flanking a chromosomal sequence. The genetically modified animal comprising the lox-flanked chromosomal sequence can then be crossed with another genetically modified animal expressing Cre recombinase. Progeny animals comprising the lox-flanked chromosomal sequence and the Cre recombinase are then produced, and the lox-flanked chromosomal sequence is recombined, leading to deletion or inversion of the chromosomal sequence encoding the protein. Expression of Cre recombinase can be temporally and conditionally regulated to effect temporally and conditionally regulated recombination of the chromosomal sequence.
[0121 ] In any of these embodiments, the genetically modified animal disclosed herein can be heterozygous for the modified chromosomal sequence.
Alternatively, the genetically modified animal can be homozygous for the modified chromosomal sequence.
[0122] The genetically modified animals disclosed herein can be crossbred to create animals comprising more than one modified chromosomal sequence or to create animals that are homozygous for one or more modified chromosomal sequences. For example, two animals comprising the same modified chromosomal sequence can be crossbred to create an animal homozygous for the modified chromosomal sequence. Alternatively, animals with different modified chromosomal sequences can be crossbred to create an animal comprising both modified chromosomal sequences.
[0123] For example, a f i rst animal comprising an inactivated
chromosomal sequence gene "x" can be crossed with a second animal comprising a chromosomally integrated sequence encoding a human gene "X" protein to give rise to "humanized" gene "X" offspring comprising both the inactivated gene "x" chromosomal sequence and the chromosomally integrated human gene "X" sequence. Also, a humanized gene "X" animal can be crossed with a humanized gene Ύ" animal to create humanized gene X/gene Y offspring. Those of skill in the art will appreciate that many combinations are possible.
[0124] In other embodiments, an animal comprising a modified
chromosomal sequence can be crossbred to combine the modified chromosomal sequence with other genetic backgrounds. By way of non-limiting example, other genetic backgrounds may include wild-type genetic backgrounds, genetic backgrounds with deletion mutations, genetic backgrounds with another targeted integration, and genetic backgrounds with non-targeted integrations.
[0125] The term "animal," as used herein, refers to a non-human animal. The animal may be an embryo, a juvenile, or an adult. Suitable animals include vertebrates such as mammals, birds, reptiles, amphibians, shellfish, and fish. Examples of suitable mammals include without limit rodents, companion animals, livestock, and primates. Non-limiting examples of rodents include mice, rats, hamsters, gerbils, and guinea pigs. Suitable companion animals include but are not limited to cats, dogs, rabbits, hedgehogs, and ferrets. Non-limiting examples of livestock include horses, goats, sheep, swine, cattle, llamas, and alpacas. Suitable primates include but are not limited to capuchin monkeys, chimpanzees, lemurs, macaques,
marmosets, tamarins, spider monkeys, squirrel monkeys, and vervet monkeys. Non- limiting examples of birds include chickens, turkeys, ducks, and geese. Alternatively, the animal may be an invertebrate such as an insect, a nematode, and the like. Non- limiting examples of insects include Drosophila and mosquitoes. An exemplary animal is a rat. Non-limiting examples of suitable rat strains include Dahl Salt- Sensitive, Fischer 344, Lewis, Long Evans Hooded, Sprague-Dawley, and Wistar. In one embodiment, the animal is not a genetically modified mouse. In each of the foregoing iterations of suitable animals for the invention, the animal does not include exogenously introduced, randomly integrated transposon sequences.
[0126] A further aspect of the present disclosure provides genetically modified cells or cell lines comprising at least one modified chromosomal sequence. The genetically modified cell or cell line can be derived from any of the genetically modified animals disclosed herein. Alternatively, the chromosomal sequence can be modified in a cell as described herein above (in the paragraphs describing chromosomal sequence modifications in animals) using the methods descried herein. The disclosure also encompasses a lysate of said cells or cell lines.
[0127] In general, the cells are eukaryotic cells. Suitable host cells include fungi or yeast, such as Pichia, Saccharomyces, or Schizosaccharomyces; insect cells, such as SF9 cells from Spodoptera frugiperda or S2 cells from Drosophila melanogaster; and animal cells, such as mouse, rat, hamster, non-human primate, or human cells. Exemplary cells are mammalian. The mammalian cells can be primary cells. In general, any primary cell that is sensitive to double strand breaks may be used. The cells may be of a variety of cell types, e.g., fibroblast, myoblast, T or B cell, macrophage, epithelial cell, and so forth.
[0128] When mammalian cell lines are used, the cell line can be any established cell line or a primary cell line that is not yet described. The cell line can be adherent or non-adherent, or the cell line can be grown under conditions that encourage adherent, non-adherent or organotypic growth using standard techniques known to individuals skilled in the art. Non-limiting examples of suitable mammalian cells and cell lines are provided herein in section (IV)(g). In still other embodiments, the cell can be a stem cell. Non-limiting examples of suitable stem cells are provided in section (IV)(g).
[0129] The present disclosure also provides a genetically modified non- human embryo comprising at least one modified chromosomal sequence. The chromosomal sequence can be modified in an embryo as described herein above (in the paragraphs describing chromosomal sequence modifications in animals) using the methods descried herein. In one embodiment, the embryo is a non-human fertilized one-cell stage embryo of the animal species of interest. Exemplary mammalian embryos, including one cell embryos, include without limit, mouse, rat, hamster, rodent, rabbit, feline, canine, ovine, porcine, bovine, equine, and primate embryos.
DEFINITIONS
[0130] Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991 ); and Hale & Marham, The Harper Collins Dictionary of Biology (1991 ). As used herein, the following terms have the meanings ascribed to them unless specified otherwise.
[0131 ] When introducing elements of the present disclosure or the preferred embodiments(s) thereof, the articles "a", "an", "the" and "said" are intended to mean that there are one or more of the elements. The terms "comprising", "including" and "having" are intended to be inclusive and mean that there may be additional elements other than the listed elements.
[0132] As used herein, the term "endogenous sequence" refers to a chromosomal sequence that is native to the cell.
[0133] The term "exogenous," as used herein, refers to a sequence that is not native to the cell, or a chromosomal sequence whose native location in the genome of the cell is in a different chromosomal location.
[0134] A "gene," as used herein, refers to a DNA region (including exons and introns) encoding a gene product, as well as all DNA regions which regulate the production of the gene product, whether or not such regulatory sequences are adjacent to coding and/or transcribed sequences. Accordingly, a gene includes, but is not necessarily limited to, promoter sequences, terminators, translational regulatory sequences such as ribosome binding sites and internal ribosome entry sites, enhancers, silencers, insulators, boundary elements, replication origins, matrix attachment sites, and locus control regions.
[0135] The term "heterologous" refers to an entity that is not endogenous or native to the cell of interest. For example, a heterologous protein refers to a protein that is derived from or was originally derived from an exogenous source, such as an exogenously introduced nucleic acid sequence. In some instances, the heterologous protein is not normally produced by the cell of interest.
[0136] The terms "nucleic acid" and "polynucleotide" refer to a
deoxyribonucleotide or ribonucleotide polymer, in linear or circular conformation, and in either single- or double-stranded form. For the purposes of the present disclosure, these terms are not to be construed as limiting with respect to the length of a polymer. The terms can encompass known analogs of natural nucleotides, as well as nucleotides that are modified in the base, sugar and/or phosphate moieties (e.g., phosphorothioate backbones). In general, an analog of a particular nucleotide has the same base-pairing specificity; i.e., an analog of A will base-pair with T.
[0137] The term "nucleotide" refers to deoxyribonucleotides or
ribonucleotides. The nucleotides may be standard nucleotides (i.e., adenosine, guanosine, cytidine, thymidine, and uridine) or nucleotide analogs. A nucleotide analog refers to a nucleotide having a modified purine or pyrimidine base or a modified ribose moiety. A nucleotide analog may be a naturally occurring nucleotide (e.g., inosine) or a non-naturally occurring nucleotide. Non-limiting examples of modifications on the sugar or base moieties of a nucleotide include the addition (or removal) of acetyl groups, amino groups, carboxyl groups, carboxymethyl groups, hydroxyl groups, methyl groups, phosphoryl groups, and thiol groups, as well as the substitution of the carbon and nitrogen atoms of the bases with other atoms (e.g., 7-deaza purines). Nucleotide analogs also include dideoxy nucleotides, 2'-0-methyl nucleotides, locked nucleic acids (LNA), peptide nucleic acids (PNA), and morpholinos.
[0138] The terms "polypeptide" and "protein" are used interchangeably to refer to a polymer of amino acid residues.
[0139] Techniques for determining nucleic acid and amino acid sequence identity are known in the art. Typically, such techniques include determining the nucleotide sequence of the mRNA for a gene and/or determining the amino acid sequence encoded thereby, and comparing these sequences to a second nucleotide or amino acid sequence. Genomic sequences can also be determined and compared in this fashion. In general, identity refers to an exact nucleotide-to-nucleotide or amino acid-to-amino acid correspondence of two polynucleotides or polypeptide sequences, respectively. Two or more sequences (polynucleotide or amino acid) can be compared by determining their percent identity. The percent identity of two sequences, whether nucleic acid or amino acid sequences, is the number of exact matches between two aligned sequences divided by the length of the shorter sequences and multiplied by 100. An approximate alignment for nucleic acid sequences is provided by the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482- 489 (1981 ). This algorithm can be applied to amino acid sequences by using the scoring matrix developed by Dayhoff, Atlas of Protein Sequences and Structure, M. O. Dayhoff ed., 5 suppl. 3:353-358, National Biomedical Research Foundation,
Washington, D.C., USA, and normalized by Gribskov, Nucl. Acids Res. 14(6):6745-6763 (1986). An exemplary implementation of this algorithm to determine percent identity of a sequence is provided by the Genetics Computer Group (Madison, Wis.) in the "BestFit" utility application. Other suitable programs for calculating the percent identity or similarity between sequences are generally known in the art, for example, another alignment program is BLAST, used with default parameters. For example, BLASTN and BLASTP can be used using the following default parameters: genetic code=standard; filter=none; strand=both; cutoff=60; expect=10; Matrix=BLOSUM62; Descriptions=50 sequences; sort by=HIGH SCORE; Databases=non-redundant,
GenBank+EMBL+DDBJ+PDB+GenBank CDS translations+Swiss
protein+Spupdate+PIR. Details of these programs can be found on the GenBank website.
[0140] As various changes could be made in the above-described cells and methods without departing from the scope of the invention, it is intended that all matter contained in the above description and in the examples given below, shall be interpreted as illustrative and not in a limiting sense.
EXAMPLES
[0141 ] The following examples illustrate certain aspects of the invention.
Example 1: Modification of Cas9 Gene for Mammalian Expression
[0142] A Cas9 gene from Streptococcus pyogenes strain MGAS15252 (Accession number YP_005388840.1 ) was optimized with Homo sapiens codon preference to enhance its translation in mammalian cells. The Cas9 gene also was modified by adding a nuclear localization signal PKKKRKV (SEQ ID NO:1 ) at the C terminus for targeting the protein into the nuclei of mammalian cells. Table 1 presents the modified Cas9 amino acid sequence, with the nuclear localization sequence underlined. Table 2 presents the codon optimized, modified Cas9 DNA sequence.
Table 1 . Modified Cas9 Amino Acid Sequence
MDKKYSIGLDIGTNSVGWAVITDDYKVPSKKFKVLGNTDRHSIKKNLIGALLFGSGETAE
ATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIF
GNIVDEVAYHEKYPTIYHLRKKLADSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNS
DVDKLFIQLVQIYNQLFEENPINASRVDAKAILSARLSKSRRLENLIAQLPGEKRNGLFG
NLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDA ILLSDILRVNSEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYA
GYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELH
AILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEE
VVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPA
FLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGAYHDLL
KIIKDKDFLDNEENEDILEDIVLTLTLFEDRGMIEERLKTYAHLFDDKVMKQLKRRRYTG
WGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQG
HSLHEQIANLAGSPAIKKGILQTVKIVDELVKVMGHKPENIVIEMARENQTTQKGQKNSR
ERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYD
VDHIVPQSFIKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRK
FDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVI
TLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKV
YDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDK
GRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGF
DSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDL
IIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNE
QKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTL
TNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDPKKKRKV
(SEQ ID N0:9)
Table 2. Optimized Cas9 DNA Sequence (5'-3')
ATGGACAAGAAGTACAGCATCGGCCTGGACATCGGCACCAACTCTGTGGGCTGGG
CCGTGATCACCGACGACTACAAGGTGCCCAGCAAGAAATTCAAGGTGCTGGGCAA
CACCGACCGGCACAGCATCAAGAAGAACCTGATCGGCGCCCTGCTGTTCGGCTCT
GGCGAAACAGCCGAGGCCACCCGGCTGAAGAGAACCGCCAGAAGAAGATACACC
AGACGGAAGAACCGGATCTGCTATCTGCAAGAGATCTTCAGCAACGAGATGGCCA
AGGTGGACGACAGCTTCTTCCACAGACTGGAAGAGTCCTTCCTGGTGGAAGAGGA
TAAGAAGCACGAGCGGCACCCCATCTTCGGCAACATCGTGGACGAGGTGGCCTAC
CACGAGAAGTACCCCACCATCTACCACCTGAGAAAGAAGCTGGCCGACAGCACCG
ACAAGGCCGACCTGAGACTGATCTACCTGGCCCTGGCCCACATGATCAAGTTCCG
GGGCCACTTCCTGATCGAGGGCGACCTGAACCCCGACAACAGCGACGTGGACAA
GCTGTTCATCCAGCTGGTGCAGATCTACAATCAGCTGTTCGAGGAAAACCCCATCA
ACGCCAGCAGAGTGGACGCCAAGGCCATCCTGAGCGCCAGACTGAGCAAGAGCA
GACGGCTGGAAAATCTGATCGCCCAGCTGCCCGGCGAGAAGCGGAATGGCCTGT
TCGGCAACCTGATTGCCCTGAGCCTGGGCCTGACCCCCAACTTCAAGAGCAACTT
CGACCTGGCCGAGGATGCCAAACTGCAGCTGAGCAAGGACACCTACGACGACGA
CCTGGACAACCTGCTGGCCCAGATCGGCGACCAGTACGCCGACCTGTTTCTGGCC
GCCAAGAACCTGTCCGACGCCATCCTGCTGAGCGACATCCTGAGAGTGAACAGCG
AGATCACCAAGGCCCCCCTGTCCGCCTCTATGATCAAGAGATACGACGAGCACCA
CCAGGACCTGACCCTGCTGAAAGCTCTCGTGCGGCAGCAGCTGCCTGAGAAGTAC
AAAGAGATTTTCTTCGACCAGAGCAAGAACGGCTACGCCGGCTACATCGATGGCG
GAGCCAGCCAGGAAGAGTTCTACAAGTTCATCAAGCCCATCCTGGAAAAGATGGA
CGGCACCGAGGAACTGCTCGTGAAGCTGAACAGAGAGGACCTGCTGCGGAAGCA
GCGGACCTTCGACAACGGCAGCATCCCCCACCAGATCCACCTGGGAGAGCTGCA CGCCATTCTGCGGCGGCAGGAAGATTTTTACCCATTCCTGAAGGACAACCGGGAA
AAGATCGAGAAGATCCTGACCTTCAGAATCCCCTACTACGTGGGCCCTCTGGCCA
GGGGAAACAGCAGATTCGCCTGGATGACCAGAAAGAGCGAGGAAACCATCACCC
CCTGGAACTTCGAGGAAGTGGTGGACAAGGGCGCCAGCGCCCAGAGCTTCATCG
AGCGGATGACCAACTTCGATAAGAACCTGCCCAACGAGAAGGTGCTGCCCAAGCA
CAGCCTGCTGTACGAGTACTTCACCGTGTACAACGAGCTGACCAAAGTGAAATAC
GTGACCGAGGGAATGCGGAAGCCCGCCTTTCTGAGCGGCGAGCAGAAAAAGGCC
ATCGTGGACCTGCTGTTCAAGACCAACCGGAAAGTGACCGTGAAGCAGCTGAAAG
AGGACTACTTCAAGAAAATCGAGTGCTTCGACAGCGTGGAAATCAGCGGCGTGGA
AGATCGGTTCAACGCCTCCCTGGGCGCCTATCACGATCTGCTGAAAATTATCAAGG
ACAAGGACTTCCTGGACAATGAGGAAAACGAGGACATTCTGGAAGATATCGTGCT
GACCCTGACACTGTTTGAGGACCGGGGCATGATCGAGGAACGGCTGAAAACCTAT
GCCCACCTGTTCGACGACAAAGTGATGAAGCAGCTGAAGCGGCGGAGATACACC
GGCTGGGGCAGGCTGAGCCGGAAGCTGATCAACGGCATCCGGGACAAGCAGTCC
GGCAAGACAATCCTGGATTTCCTGAAGTCCGACGGCTTCGCCAACAGAAACTTCAT
GCAGCTGATCCACGACGACAGCCTGACCTTTAAAGAGGACATCCAGAAAGCCCAG
GTGTCCGGCCAGGGACACTCTCTGCACGAGCAGATCGCCAATCTGGCCGGATCC
CCCGCCATTAAGAAGGGCATCCTGCAGACAGTGAAGATTGTGGACGAGCTCGTGA
AAGTGATGGGCCACAAGCCCGAGAACATCGTGATCGAAATGGCCAGAGAGAACCA
GACCACCCAGAAGGGACAGAAGAACAGCCGCGAGAGAATGAAGCGGATCGAAGA
GGGCATCAAAGAGCTGGGCAGCCAGATCCTGAAAGAACACCCCGTGGAAAACACC
CAGCTGCAGAACGAGAAGCTGTACCTGTACTACCTGCAGAATGGGCGGGATATGT
ACGTGGACCAGGAACTGGACATCAACCGGCTGTCCGACTACGATGTGGACCACAT
TGTGCCCCAGTCCTTCATCAAGGACGACTCCATCGATAACAAAGTGCTGACTCGGA
GCGACAAGAACCGGGGCAAGAGCGACAACGTGCCCTCCGAAGAGGTCGTGAAGA
AGATGAAGAACTACTGGCGCCAGCTGCTGAATGCCAAGCTGATTACCCAGAGGAA
GTTCGACAATCTGACCAAGGCCGAGAGAGGCGGCCTGAGCGAACTGGATAAGGC
CGGCTTCATTAAGCGGCAGCTGGTGGAAACCCGGCAGATCACAAAGCACGTGGCA
CAGATCCTGGACTCCCGGATGAACACTAAGTACGACGAGAACGACAAACTGATCC
GGGAAGTGAAAGTGATCACCCTGAAGTCCAAGCTGGTGTCCGACTTCAGAAAGGA
TTTCCAGTTTTACAAAGTGCGCGAGATCAACAACTACCACCACGCCCACGACGCCT
ACCTGAACGCCGTCGTGGGAACCGCCCTGATCAAAAAGTACCCTAAGCTGGAAAG
CGAGTTCGTGTACGGCGATTACAAGGTGTACGACGTGCGGAAGATGATCGCCAAG
AGCGAGCAGGAAATCGGCAAGGCTACCGCCAAGTACTTCTTCTACAGCAACATCA
TGAACTTTTTCAAGACCGAGATCACACTGGCCAACGGCGAGATCAGAAAGCGGCC
TCTGATCGAGACAAACGGCGAAACCGGGGAGATCGTGTGGGATAAGGGCCGGGA
TTTTGCCACAGTGCGGAAAGTGCTGTCCATGCCCCAAGTGAATATCGTGAAAAAGA
CCGAGGTGCAGACCGGCGGCTTCAGCAAAGAGTCTATCCTGCCCAAGAGGAACTC
CGACAAGCTGATCGCCAGAAAGAAGGATTGGGACCCTAAGAAGTACGGCGGCTTT
GACAGCCCCACCGTGGCCTACTCTGTGCTGGTGGTGGCCAAAGTGGAAAAGGGC
AAGTCCAAGAAACTGAAGAGTGTGAAAGAGCTGCTGGGGATCACCATCATGGAAA
GAAGCAGCTTCGAGAAGAATCCCATCGACTTTCTGGAAGCCAAGGGCTACAAAGA
AGTGAAAAAGGACCTGATCATCAAGCTGCCTAAGTACTCCCTGTTCGAGCTGGAAA
ACGGCCGGAAGCGGATGCTGGCTTCTGCCGGCGAACTGCAGAAGGGAAACGAGC
TGGCCCTGCCCTCCAAATATGTGAACTTCCTGTACCTGGCCAGCCACTATGAGAAG
CTGAAGGGCTCCCCCGAGGATAATGAGCAGAAACAGCTGTTTGTGGAACAGCACA AGCACTACCTGGACGAGATCATCGAGCAGATTAGCGAGTTCTCCAAGCGCGTGAT
CCTGGCCGATGCCAACCTGGACAAGGTGCTGAGCGCCTACAACAAGCACCGGGA
TAAGCCCATCAGAGAGCAGGCCGAGAATATCATCCACCTGTTTACCCTGACCAACC
TGGGAGCCCCTGCCGCCTTCAAGTACTTTGACACCACCATCGACCGGAAGAGGTA
CACCAGCACCAAAGAGGTGCTGGACGCCACCCTGATCCACCAGAGCATCACCGG
CCTGTACGAGACACGGATCGACCTGTCTCAGCTGGGAGGCGACCCCAAGAAAAAG
CGCAAAGTG (SEQ ID NO:10)
[0143] The modified Cas9 DNA sequence was placed under the control of cytomegalovirus (CMV) promoter for constituent expression in mammalian cells. The modified Cas9 DNA sequence was also placed under the control T7 promoter for in vitro mRNA synthesis with T7 RNA polymerase. In vitro RNA transcription was performed by using MessageMAX T7 ARCA-Capped Message Transcription Kit and T7 mScript Standard mRNA Production System (Cellscript).
Example 2: Targeting Cas9
[0144] The adeno-associated virus integration site 1 (AAVS1 ) locus was used as a target for Cas9-mediated human genome modification. The human AAVS1 locus is located in intron 1 (4427 bp) of protein phosphatase 1 , regulatory subunit 12C (PPP1 R12C). Table 3 presents the first exon (shaded gray) and the first intron of PPP1 R12C. The underlined sequence within the intron is the targeted modification site (i.e., AAVS1 locus).
Figure imgf000055_0001
CCCCTCTTCCGATGTTGAGCCCCTCCAGCCGGTCCTGGACTTTGTCTCCTTCCCTG
CCCTGCCCTCTCCTGAACCTGAGCCAGCTCCCATAGCTCAGTCTGGTCTATCTGCC
TGGCCCTGGCCATTGTCACTTTGCGCTGCCCTCCTCTCGCCCCCGAGTGCCCTTG
CTGTGCCGCCGGAACTCTGCCCTCTAACGCTGCCGTCTCTCTCCTGAGTCCGGAC
CACTTTGAGCTCTACTGGCTTCTGCGCCGCCTCTGGCCCACTGTTTCCCCTTCCCA
GGCAGGTCCTGCTTTCTCTGACCTGCATTCTCTCCCCTGGGCCTGTGCCGCTTTCT
GTCTGCAGCTTGTGGCCTGGGTCACCTCTACGGCTGGCCCAGATCCTTCCCTGCC
GCCTCCTTCAGGTTCCGTCTTCCTCCACTCCCTCTTCCCCTTGCTCTCTGCTGTGT
TGCTGCCCAAGGATGCTCTTTCCGGAGCACTTCCTTCTCGGCGCTGCACCACGTG
ATGTCCTCTGAGCGGATCCTCCCCGTGTCTGGGTCCTCTCCGGGCATCTCTCCTC
CCTCACCCAACCCCATGCCGTCTTCACTCGCTGGGTTCCCTTTTCCTTCTCCTTCT
GGGGCCTGTGCCATCTCTCGTTTCTTAGGATGGCCTTCTCCGACGGATGTCTCCCT
TGCGTCCCGCCTCCCCTTCTTGTAGGCCTGCATCATCACCGTTTTTCTGGACAACC
CCAAAGTACCCCGTCTCCCTGGCTTTAGCCACCTCTCCATCCTCTTGCTTTCTTTG
CCTGGACACCCCGTTCTCCTGTGGATTCGGGTCACCTCTCACTCCTTTCATTTGGG
CAGCTCCCCTACCCCCCTTACCTCTCTAGTCTGTGCTAGCTCTTCCAGCCCCCTGT
CATGGCATCTTCCAGGGGTCCGAGAGCTCAGCTAGTCTTCTTCCTCCAACCCGGG
CCCCTATGTCCACTTCAGGACAGCATGTTTGCTGCCTCCAGGGATCCTGTGTCCCC
GAGCTGGGACCACCTTATATTCCCAGGGCCGGTTAATGTGGCTCTGGTTCTGGGT
ACTTTTATCTGTCCCCTCCACCCCACAGTGGGGCCACTAGGGACAGGATTGGTGA
CAGAAAAGCCCCATCCTTAGGCCTCCTCCTTCCTAGTCTCCTGATATTGGGTCTAA
CCCCCACCTCCTGTTAGGCAGATTCCTTATCTGGTGACACACCCCCATTTCCTGGA
GCCATCTCTCTCCTTGCCAGAACCTCTAAGGTTTGCTTACGATGGAGCCAGAGAGG
ATCCTGGGAGGGAGAGCTTGGCAGGGGGTGGGAGGGAAGGGGGGGATGCGTGA
CCTGCCCGGTTCTCAGTGGCCACCCTGCGCTACCCTCTCCCAGAACCTGAGCTGC
TCTGACGCGGCCGTCTGGTGCGTTTCACTGATCCTGGTGCTGCAGCTTCCTTACA
CTTCCCAAGAGGAGAAGCAGTTTGGAAAAACAAAATCAGAATAAGTTGGTCCTGAG
TTCTAACTTTGGCTCTTCACCTTTCTAGTCCCCAATTTATATTGTTCCTCCGTGCGT
CAGTTTTACCTGTGAGATAAGGCCAGTAGCCAGCCCCGTCCTGGCAGGGCTGTGG
TGAGGAGGGGGGTGTCCGTGTGGAAAACTCCCTTTGTGAGAATGGTGCGTCCTAG
GTGTTCACCAGGTCGTGGCCGCCTCTACTCCCTTTCTCTTTCTCCATCCTTCTTTCC
TTAAAGAGTCCCCAGTGCTATCTGGGACATATTCCTCCGCCCAGAGCAGGGTCCC
GCTTCCCTAAGGCCCTGCTCTGGGCTTCTGGGTTTGAGTCCTTGGCAAGCCCAGG
AGAGGCGCTCAGGCTTCCCTGTCCCCCTTCCTCGTCCACCATCTCATGCCCCTGG
CTCTCCTGCCCCTTCCCTACAGGGGTTCCTGGCTCTGCTCTTCAGACTGAGCCCC
GTTCCCCTGCATCCCCGTTCCCCTGCATCCCCCTTCCCCTGCATCCCCCAGAGGC
CCCAGGCCACCTACTTGGCCTGGACCCCACGAGAGGCCACCCCAGCCCTGTCTA
CCAGGCTGCCTTTTGGGTGGATTCTCCTCCAACTGTGGGGTGACTGCTTGGCAAA
CTCACTCTTCGGGGTATCCCAGGAGGCCTGGAGCATTGGGGTGGGCTGGGGTTC
AGAGAGGAGGGATTCCCTTCTCAGGTTACGTGGCCAAGAAGCAGGGGAGCTGGG
TTTGGGTCAGGTCTGGGTGTGGGGTGACCAGCTTATGCTGTTTGCCCAGGACAGC
CTAGTTTTAGCACTGAAACCCTCAGTCCTAGGAAAACAGGGATGGTTGGTCACTGT
CTCTGGGTGACTCTTGATTCCCGGCCAGTTTCTCCACCTGGGGCTGTGTTTCTCGT
CCTGCATCCTTCTCCAGGCAGGTCCCCAAGCATCGCCCCCCTGCTGTGGCTGTTC
CCAAGTTCTTAGGGTACCCCACGTGGGTTTATCAACCACTTGGTGAGGCTGGTACC
CTGCCCCCATTCCTGCACCCCAATTGCCTTAGTGGCTAGGGGGTTGGGGGCTAGA GTAGGAGGGGCTGGAGCCAGGATTCTTAGGGCTGAACAGAGAAGAGCTGGGGGC
CTGGGCTCCTGGGTTTGAGAGAGGAGGGGCTGGGGCCTGGACTCCTGGGTCCGA
GGGAGGAGGGGCTGGGGCCTGGACTCCTGGGTCTGAGGGTGGAGGGACTGGGG
GCCTGGACTCCTGGGTCCGAGGGAGGAGGGGCTGGGGCCTGGACTCGTGGGTC
TGAGGGAGGAGGGGCTGGGGGCCTGGACTTCTGGGTCTTAGGGAGGCGGGGCT
GGGCCTGGACCCCTGGGTCTGAATGGGGAGAGGCTGGGGGCCTGGACTCCTTCA
TCTGAGGGCGGAAGGGCTGGGGCCTGGCCTCCTGGGTTGAATGGGGAGGGGTTG
GGCCTGGACTCTGGAGTCCCTGGTGCCCAGGCCTCAGGCATCTTTCACAGGGATG
CCTGTACTGGGCAGGTCCTTGAAAGGGAAAGGCCCATTGCTCTCCTTGCCCCCCT
CCCCTATCGCCATGACAACTGGGTGGAAATAAACGAGCCGAGTTCATCCCGTTCC
CAGGGCACGTGCGGCCCCTTCACAGCCCGAGTTTCCATGACCTCATGCTCTTGGC
CCTCGTAGCTCCCTCCCGCCTCCTCCAGATGGGCAGCTTTGGAGAGGTGAGGGAC
TTGGGGGGTAATTTATCCCGTGGATCTAGGAGTTTAGCTTCACTCCTTCCTCAGCT
CCAGTTCAGGTCCCGGAGCCCACCCAGTGTCCACAAGGCCTGGGGCAAGTCCCT
CCTCCGACCCCCTGGACTTCGGCTTTTGTCCCCCCAAGTTTTGGACCCCTAAGGG
AAGAATGAGAAACGGTGGCCCGTGTCAGCCCCTGGCTGCAGGGCCCCGTGCAGA
GGGGGCCTCAGTGAACTGGAGTGTGACAGCCTGGGGCCCAGGCACACAGGTGTG
CAGCTGTCTCACCCCTCTGGGAGTCCCGCCCAGGCCCCTGAGTCTGTCCCAGCAC
AGGGTGGCCTTCCTCCACCCTGCATAGCCCTGGGCCCACGGCTTCGTTCCTGCAG
AGTATCTGCTGGGGTGGTTTCCGAGCTTGACCCTTGGAAGGACCTGGCTGGGTTT
AAGGCAGGAGGGGCTGGGGGCCAGGACTCCTGGCTCTGAAGGAGGAGGGGCTG
GAACCTCTTCCCTAGTCTGAGCACTGGAAGCGCCACCTGTGGGTGGTGACGGGG
GTTTTGCCGTGTCTAACAGGTACCATGTGGGGTTCCCGCACCCAGATGAGAAGCC
CCCTCCCTTCCCCGTTCACTTCCTGTTTGCAGATAGCCAGGAGTCCTTTCGTGGTT
TCCACTGAGCACTGAAGGCCTGGCCGGCCTGACCACTGGGCAACCAGGCGTATC
TTAAACAGCCAGTGGCCAGAGGCTGTTGGGTCATTTTCCCCACTGTCCTAGCACC
GTGTCCCTGGATCTGTTTTCGTGGCTCCCTCTGGAGTCCCGACTTGCTGGGACAC
CGTGGCTGGGGTAGGTGCGGCTGACGGCTGTTTCCCACCCCCAG (SEQ I D
N0:1 1 )
[0145] Cas9 guide RNAs were designed for targeting the human AAVS1 locus. A 42 nucleotide RNA (referred to herein as a "crRNA" sequence) comprising (5' to 3') a target recognition sequence (i.e., sequence complementary to the non-coding strand of the target sequence) and protospacer sequence; a 85 nucleotide RNA
(referred to herein as a "tracrRNA" sequence) comprising 5' sequence with
complementarity to the 3' sequence of the crRNA and additional hairpin sequence; and a chimeric RNA comprising nucleotides 1 -32 of the crRNA, a GAAA loop, and nucleotides 19-45 of the tracrRNA were prepared. The crRNA was chemically synthesized by Sigma-Aldrich. The tracrRNA and chimeric RNA were synthesized by in vitro transcription with T7 RNA polymerase using T7-Scribe Standard RNA IVT Kit (Cellscript). The chimeric RNA coding sequence was also placed under the control of human U6 promoter for in vivo transcription in human cells. Table 4 presents the sequences of the guide RNAs.
Figure imgf000058_0001
Example 3: Preparation of Donor Polynucleotide to Monitor Genome Modification
[0146] Targeted integration of a GFP protein into the N terminus of PPP1 R12C was used to monitor Cas9-mediated genome modification. To mediate integration by homologous recombination a donor polynucleotide was prepared. The AAVS1 -GFP DNA donor contained a 5' (1 185 bp) AAVS1 locus homologous arm, an RNA splicing receptor, a turbo GFP coding sequence, a 3' transcription terminator, and a 3' (1217 bp) AAVS1 locus homologous arm. Table 5 presents the sequences of the RNA splicing receptor and the GFP coding sequence followed by the 3' transcription terminator. Plasmid DNA was prepared by using GenElute Endotoxin-Free Plasmid Maxiprep Kit (Sigma).
Figure imgf000058_0002
ACGAGAACCCCTTCCTGCACGCCATCAACAACGGCGGCTA
CACCAACACCCGCATCGAGAAGTACGAGGACGGCGGCGT
GCTGCACGTGAGCTTCAGCTACCGCTACGAGGCCGGCCG
CGTGATCGGCGACTTCAAGGTGATGGGCACCGGCTTCCCC
GAGGACAGCGTGATCTTCACCGACAAGATCGTCCGCAGCA
ACGCCACCGTGGAGCACCTGCACCCCATGGGCGATAACG
ATCTGGATGGCAGCTTCACCCGCACCTTCAGCCTGCGCGA
CGGCGGCTACTACAGCTCCGTGGTGGACAGCCACATGCAC
TTCAAGAGCGCCATCCACCCCAGCATCCTGCAGAACGGGG
GCCCCATGTTCGCCTTCCGCCGCGTGGAGGAGGATCACA
GCAACACCGAGCTGGGCATCGTGGAGTACCAGCACGCCTT
CAAGACCCCGGATGCAGATGCCGGTGAAGAATGAAGATCT
CTGTGCCTTCTAGTTGCCAGCCATCTGTTGTTTGCCCCTCC
CCCGTGCCTTCCTTGACCCTGGAAGGTGCCACTCCCACTG
TCCTTTCCTAATAAAATGAGGAAATTGCATCGCATTGTCTGA
GTAGGTGTCATTCTATTCTGGGGGGTGGGGTGGGGCAGG
ACAGCAAGGGGGAGGATTGGGAAGACAATAGCAGGCATG
CTGGGGATGCGGTGGGCTCTATGGACTCGAGGTTTAAACG
TCGACGCGGCCGCGT
[0147] Targeted gene integration will result in a fusion protein between the first 107 amino acids of the PPP1 R12C and the turbo GFP. The expected fusion protein contains the first 107 amino acid residues of PPP1 R12C (highlighted in grey) from RNA splicing between the first exon of PPP1 R12C and the engineered splice receptor (see Table 6).
Table 6. Predicted amino acid sequence of the PPP1 R12C-GFP fusion protein.
MSGEDGPAAGPGAAAAAARERRREQLRQWGARAGAEPGPGERRARTVRFERAAEF
LAACAGGDLDEARLMLRAADPGPGAELDPAAPPPARAVLDSTNADGISALHQATMDY
KDDDDKVDSRAAESDESGLPAMEIECRITGTLNGVEFELVGGGEGTPEQGRMTNKMK STKGALTFSPYLLSHVMGYGFYHFGTYPSGYENPFLHAINNGGYTNTRIEKYEDGGVL HVSFSYRYEAGRVIGDFKVMGTGFPEDSVIFTDKIVRSNATVEHLHPMGDNDLDGSFT RTFSLRDGGYYSSVVDSHMHFKSAIHPSILQNGGPMFAFRRVEEDHSNTELGIVEYQH AFKTPDADAGEE (SEQ ID NO:17) Example 4: Cas9-Mediated Targeted Integration
[0148] Transfection was performed on human K562 cells. The K562 cell line was obtained from American Type Culture Collection (ATCC) and grown in Iscove's Modified Dulbecco's Medium, supplemented with 10% FBS and 2 mM L-glutamine. All media and supplements were obtained from Sigma-Aldrich. Cultures were split one day before transfection (at approximately 0.5 million cells per ml_ before transfection). Cells were transfected with Nucleofector Solution V (Lonza) on a Nucleofector (Lonza) with the T-016 program. Each nucleofection contained approximately 0.6 million cells.
Transfection treatments are detailed in Table 7. Cells were grown at 37 °C and 5% CO2 immediately after nucleofection.
Figure imgf000060_0001
[0149] Fluorescence-activated cell sorting (FACS) was performed 4 days after transfection. FACS data are presented in FIG. 4. The percent GFP detected in each of the four experimental treatments (A-D) was greater than in the control treatments (E, F), confirming integration of the donor sequence and expression of the fusion protein. Example 5: PCR Confirmation of Targeted Integration
[0150] Genomic DNA was extracted from transfected cells with GenElute Mammalian Genomic DNA Miniprep Kit (Sigma) 12 days after transfection. Genomic DNA was then PCR amplified with a forward primer located outside the 5' homologous arm of the AAVS1 -GFP plasmid donor and a reverse primer located at the 5' region of the GFP. The forward primer was 5'- CCACTCTGTGCTGACCACTCT-3' (SEQ ID NO:18) and reverse primer was 5'- GCGGCACTCGATCTCCA-3' (SEQ ID NO:19). The expected fragment size from the junction PCR was 1388 bp. The amplification was carried out with JumpStart Taq ReadyMix (Sigma), using the following cycling
conditions: 98 °C for 2 minutes for initial denaturation; 35 cycles of 98 °C for 15 seconds, 62 °C for 30 seconds, and 72 °C for 1 minutes and 30 seconds; and a final extension at 72 °C for 5 minutes. PCR products were resolved on 1 % agarose gel.
[0151 ] Cells transfected with 10 μg of Cas9 mRNA transcribed with an Anti-Reverse Cap Analog, 0.3 nmol of pre-annealed crRNA-tracrRNA duplex, and 10 μg of AAVS1 -GFP plasmid DNA displayed a PCR product of the expected size (see lane A, FIG. 5).
Example 6: Cas9-Based Genome Editing in Mouse Embryos
[0152] The mouse Rosa26 locus can be targeted for genome
modifications. Table 8 presents a portion of the mouse Rosa26 sequence in which potential target sites are shown in bold. Each target site comprises a protospacer.
Table 8. Mouse Rosa26 Sequence
GAGCGGCTGCGGGGCGGGTGCAAGCACGTTTCCGACTTGAGTTGCCTCAAGAGG
GGCGTGCTGAGCCAGACCTCCATCGCGCACTCCGGGGAGTGGAGGGAAGGAGC
GAGGGCTCAGTTGGGCTGTTTTGGAGGCAGGAAGCACTTGCTCTCCCAAAGTCGC
TCTGAGTTGTTATCAGTAAGGGAGCTGCAGTGGAGTAGGCGGGGAGAAGGCCGC
ACCCTTCTCCGGAGGGGGGAGGGGAGTGTTGCAATACCTTTCTGGGAGTTCTCTG
CTGCCTCCTGGCTTCTGAGGACCGCCCTGGGCCTGGGAGAATCCCTTCCCCCTCT
TCCCTCGTGATCTGCAACTCCAGTCTTTCTAGAAGATGGGCGGGAGTCTTCTGGG
CAGGCTTAAAGGCTAACCTGGTGTGTGGGCGTTGTCCTGCAGGGGAATTGAACAG
GTGTAAAATTGGAGGGACAAGACTTCCCACAGATTTTCGGTTTTGTCGGGAAGTTT
TTTAATAGGGGCAAATAAGGAAAATGGGAGGATAGGTAGTCATCTGGGGTTTTATG CAGCAAAACTACAGGTTATTATTGCTTGTGATCCGCCTCGGAGTATTTTCCATCGA GGTAGATTAAAGACATGCTCACCCGAGTTTTATACTCTCCTGCTTGAGATCCTTACT ACAGTATGAAATTACAGTGTCGCGAGTTAGACTATGTAAGCAGAATTTTA (SEQ ID NO:20)
[0153] Guide RNAs were designed to target each of the target sites in the mouse Rosa26 locus. The sequences are shown in Table 9, each is 42 nucleotides in length and the 5' region is complementary to the strand that is not presented in Table 8 (i.e., the strand that is complementary to the strand shown in Table 8).
Figure imgf000062_0001
[0154] The crRNAs were chemically synthesized and pre-annealed to the tracrRNA (SEQ ID NO:13; see Example 2). Pre-annealed crRNA / tracrRNA and in vitro transcribed mRNA encoding modified Cas9 protein (SEQ ID NO. 9; see Example 1 ) can be microinjected into the pronuclei of fertilized mouse embryos. Upon guidance to the target set by the crRNA, the Cas9 protein cleaves the target site, and the resultant double-stranded break can be repaired via a non-homologous end-joining (NHEJ) repair process. The injected embryos can be either incubated at 37 °C, 5% CO2 overnight or for up to 4 days, followed by genotyping analysis, or the injected embryos can be implanted into recipient female mice such that live born animals can be genotyped. The in w'fro-incu bated embryos or tissues from live born animals can be screened for the presence of Cas9-induced mutation at the Rosa locus using standard methods. For example, the embryos or tissues from fetus or live-born animals can be harvested for DNA extraction and analysis. DNA ca n b e isolated using standard procedures. The targeted region of the Rosa26 locus can be PCR amplified using appropriate primers. Because NHEJ is error-prone, deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. Mutations can be detected using PCR-based genotyping methods, such as Cel-I mismatch assays and DNA sequencing.
Example 7: Cas9-Based Genome Modification in Mouse Embryos
[0155] The Rosa26 locus can be modified in mouse embryos by co- injecting a donor polynucleotide, as detailed above in section (IV)(d), along with the pre- annealed crRNA / tracrRNA and mRNA encoding modified Cas9 as described above in Example 6. In w'fro-incu bated embryos or tissues from live born animals (as described in Example 6) can be screened for a modified Rosa26 locus using PCR-based genotyping methods, such as RFLP assays, junction PCR, and DNA sequencing.
Example 8: Cas9-Based Genome Editing in Rat Embryos
[0156] The rat Rosa26 locus can be targeted for genome modifications. Table 10 presents a portion of the rat sequence in which potential target sites are shown in bold. Each target site comprises a protospacer.
Table 10. Rat Rosa26 Sequence
GGGATTCCTCCTTGAGTTGTGGCACTGAGGAACGTGCTGAACAAGACCTACATTG
CACTCCAGGGAGTGGATGAAGGAGTTGGGGCTCAGTCGGGTTGTATTGGAGACAA
GAAGCACTTGCTCTCCAAAAGTCGGTTTGAGTTATCATTAAGGGAGCTGCAGTGGA
GTAGGCGGAGAAAAGGCCGCACCCTTCTCAGGACGGGGGAGGGGAGTGTTGCAA
TACCTTTCTGGGAGTTCTCTGCTGCCTCCTGTCTTCTGAGGACCGCCCTGGGCCT
GGAAGATTCCCTTCCCCCTTCTTCCCTCGTGATCTGCAACTGGAGTCTTTCTGGAA
GATAGGCGGGAGTCTTCTGGGCAGGCTTAAAGGCTAACCTGGTGCGTGGGGCGT
TGTCCTGCAGAGGAATTGAACAGGTGTAAAATTGGAGGGGCAAGACTTCCCACAG
ATTTTCGATTGTGTTGTTAAGTATTGTAATAGGGGCAAATAAGGGAAATAGACTAGG
CACTCACCTGGGGTTTTATGCAGCAAAACTACAGGTTATTATTGCTTGTGATCCGC
CCTGGAGAATTTTTCACCGAGGTAGATTGAAGACATGCCCACCCAAATTTTAATATT
CTTCCACTTGCGATCCTTGCTACAGTATGAAA (SEQ ID NO:24)
[0157] Guide RNAs were designed to target each of the target sites in the rat Rosa26 locus. The sequences are shown in Table 1 1 , each is 42 nucleotides in length and the 5' region is complementary to the strand that is not presented in Table 10 (i.e., the strand that is complementary to the strand shown in Table 10).
Figure imgf000064_0001
[0158] The crRNAs were chemically synthesized and pre-annealed to the tracrRNA (SEQ ID NO:13; see Example 2). Pre-annealed crRNA / tracrRNA and in vitro transcribed mRNA encoding modified Cas9 protein (SEQ ID NO. 9; see Example 1 ) can be microinjected into the pronuclei of fertilized rat embryos. Upon guidance to the target site by the crRNA, the Cas9 protein cleaves the target site, and the resultant double-stranded break can be repaired via a non-homologous end-joining (NHEJ) repair process. The injected embryos can be either incubated at 37 °C, 5% CO2 overnight or for up to 4 days, followed by genotyping analysis, or the injected embryos can be implanted into recipient female mice such that live born animals can be genotyped. The in w'fro-incu bated embryos or tissues from live born animals can be screened for the presence of Cas9-induced mutation at the Rosa locus using standard methods. For example, the embryos or tissues from fetus or live-born animals can be harvested for DNA extraction and analysis. DNA ca n b e isolated using standard procedures. The targeted region of the Rosa26 locus can be PCR amplified using appropriate primers. Because NHEJ is error-prone, deletions of at least one nucleotide, insertions of at least one nucleotide, substitutions of at least one nucleotide, or combinations thereof can occur during the repair of the break. Mutations can be detected using PCR-based genotyping methods, such as Cel-I mismatch assays and DNA sequencing. Example 9: Cas9-Based Genome Modification in Rat Embryos
[0159] The Rosa26 locus can be modified in rat embryos by co-injecting a donor polynucleotide, as detailed above in section (IV)(d), along with the pre-annealed crRNA / tracrRNA and mRNA encoding modified Cas9 as described above in Example 8. In vitro- incubated embryos or tissues from live born rats (as described in Example 8) can be screened for a modified Rosa26 locus using PCR-based genotyping methods, such as RFLP assays, junction PCR, and DNA sequencing.

Claims

CLAIMS What is claimed is:
1 . An isolated endonuclease comprising at least one nuclear localization signal, at least one nuclease domain, and at least one domain that interacts with a guide RNA to target the endonuclease to a specific nucleotide sequence for cleavage.
2. The endonuclease of claim 1 , wherein the endonuclease is derived from a Cas9 protein.
3. The isolated endonuclease of either claim 1 or claim 2, wherein the
endonuclease is modified to lack at least one functional nuclease domain.
4. The isolated endonuclease of any one of claims 1 to 3, further comprising a cell-penetrating domain, a marker domain, or both.
5. The isolated endonuclease of any one of claim 1 to 4, which is part of a
protein-RNA complex comprising the guide RNA.
6. The isolated endonuclease of claim 5, wherein the guide RNA is a single molecule comprising a 5' region that is complementary to a target site.
7. An isolated nucleic acid encoding the endonuclease of any one of claims 1 to 4.
8. The isolated nucleic acid of claim 7, wherein the nucleic acid is codon
optimized for translation in mammalian cells.
9. The isolated nucleic acid of claim 7, wherein the nucleic acid is codon
optimized for translation in human cells.
10. The isolated nucleic acid of any one of claims 7 to 9, which is operably linked to a promoter control sequence.
1 1 . A vector comprising the isolated nucleic acid of claim 10.
12. The vector of claim 1 1 , further comprising a sequence encoding a guide RNA that is operably linked to a promoter control sequence.
13. A method for modifying a chromosomal sequence in a eukaryotic cell or embryo, the method comprising:
a) introducing into the eukaryotic cell or embryo (i) at least one RNA- guided endonuclease comprising at least one nuclear localization signal or nucleic acid encoding at least one RNA-guided endonuclease comprising at least one nuclear localization signal, (ii) at least one guide RNA or DNA encoding at least one guide RNA, and, optionally, (iii) at least one donor polynucleotide; and
b) culturing the eukaryotic cell or embryo such that each guide RNA
directs an RNA-guided endonuclease to a targeted site in the chromosomal sequence where the RNA-guided endonuclease introduces a double-stranded break in the targeted site, and the double-stranded break is repaired by a DNA repair process such that the chromosomal sequence is modified.
14. The method of claim 13, wherein the RNA-guided endonuclease is derived from a Cas9 protein.
15. The method of either claim 13 or claim 14, wherein the nucleic acid encoding the RNA-guided endonuclease is mRNA.
16. The method of either claim 13 or claim 14, wherein the nucleic acid encoding the RNA-guided endonuclease is DNA.
17. The method of claim 16, wherein the DNA is part of a vector that further
comprises a sequence encoding the guide RNA.
18. The method of any one of claims 13 to 17, wherein the eukaryotic cell is a human cell, a non-human mammalian cell, a stem cell, a non-mammalian vertebrate cell, an invertebrate cell, a plant cell, or a single cell eukaryotic organism.
19. The method of any one of claims 13 to 17, wherein the embryo is a non- human one cell animal embryo.
20. A fusion protein comprising a CRISP R/Cas-like protein or fragment thereof and an effector domain.
21 . The fusion protein of claim 20, wherein the CRISPR/Cas-like protein is
derived from a Cas9 protein.
22. The fusion protein of claim 21 , wherein the Cas9 protein is modified to lack at least one functional nuclease domain.
23. The fusion protein of claim 21 , wherein the Cas9 protein is modified to lack all nuclease activity.
24. The fusion protein of any one of claims 20 to 23, wherein the effector domain is a cleavage domain.
25. The fusion protein of claim 24, wherein the cleavage domain is a Fokl
endonuclease domain or a modified Fokl cleavage domain.
26. The fusion protein of claim 25, wherein the fusion protein forms a dimer with a second fusion protein.
27. The fusion protein of claim 25, wherein the fusion protein forms a heterodimer with a zinc finger nuclease comprising a Fokl endonuclease domain or a modified Fokl cleavage domain.
28. The fusion protein of claim 20, wherein the CRISPR/Cas-like protein is
derived from a Cas9 protein modified to lack all nuclease activity, and the effector domain is a Fokl endonuclease domain or a modified Fokl cleavage domain.
29. The fusion protein of any one of claims 20 to 23, wherein the effector domain is chosen from a transcriptional activation domain, a transcriptional repressor domain, and an epigenetic modification domain.
30. The fusion protein of any one of claims 20 to 29, wherein the fusion protein further comprises at least one additional domain chosen from a nuclear localization signal, a cell-penetrating domain, and a marker domain.
31 . An isolated nucleic acid encoding the fusion protein of any one claims 20 to 30.
32. A method for modifying a chromosomal sequence or regulating expression of a chromosomal sequence in a cell or embryo, the method comprising introducing into the cell or embryo (a) at least one fusion protein or nucleic acid encoding at least one fusion protein, wherein the fusion protein comprises a CRISPR/Cas-like protein or a fragment thereof and an effector domain, and (b) at least one guide RNA or DNA encoding at least one guide RNA, wherein the one guide RNA guides the CRISPR/Cas-like protein of the fusion protein to a targeted site in the chromosomal sequence and the effector domain of the fusion protein modifies the chromosomal sequence or regulates expression of the chromosomal sequence.
33. The method of claim 32, wherein the CRISPR/Cas-like protein is derived from a Cas9 protein.
34. The method of claim 33, wherein the cas9 protein is modified to lack at least one functional nuclease domain.
35. The method of claim 33, wherein the cas9 protein is modified to lack all
nuclease activity.
36. The method of any one of claims 32 to 35, wherein fusion protein further comprises at least one additional domain chosen from a nuclear localization signal, a cell-penetrating domain, and a marker domain.
37. The method of any one of claims 32 to 36, wherein the effector domain is a cleavage domain.
38. The method of claim 37, wherein one fusion protein or nucleic acid encoding one fusion protein and two guide RNAs or DNA encoding two guide RNAs are introduced into the cell or embryo.
39. The method of claim 38, wherein the fusion protein comprises a Cas9 protein or fragment thereof and a Fokl cleavage domain or a modified Fokl cleavage domain.
40. The method of claim 37, wherein two fusion proteins or nucleic acid encoding two fusion proteins and two guide RNAs or DNA encoding two guide RNAs are introduced into the cell or embryo.
41 . The method of claim 40, wherein each fusion protein comprises a different Cas9 protein or fragment thereof and/or a Fokl cleavage domain or a modified Fokl cleavage domain.
42. The method of claim 37, wherein one fusion protein or nucleic acid encoding one fusion protein, one guide RNA or DNA encoding one guide RNA, and one zinc finger nuclease or nucleic acid encoding one zinc finger nuclease are introduced into the cell or embryo.
43. The method of claim 42, wherein the fusion protein comprises a Cas9 protein or fragment thereof and a Fokl cleavage domain or derivative thereof, and the zinc finger nuclease comprises a Fokl cleavage domain or a modified Fokl cleavage domain.
44. The method of any one of claims 37 to 43, further comprising introducing into the cell or embryo at least one donor polynucleotide.
45. The method of any one of claims 32 to 36, wherein the effector domain is chosen from an epigenetic modification domain, a transcriptional activation domain, or a transcriptional repressor domain.
46. The method of claim 45, wherein one fusion protein or nucleic acid encoding one fusion protein, and one guide RNA or DNA encoding one guide RNA are introduced into the cell or embryo.
47. The method of any one of claims 32 to 46, wherein the cell is a human cell, a non-human mammalian cell, a stem cell, a non-mammalian vertebrate cell, an invertebrate cell, a plant cell, or a single cell eukaryotic organism.
48. The method of any one of claims 32 to 46, wherein the embryo is a non- human one cell animal embryo.
PCT/US2013/073307 2012-12-06 2013-12-05 Crispr-based genome modification and regulation WO2014089290A1 (en)

Priority Applications (57)

Application Number Priority Date Filing Date Title
PL18160519T PL3363902T3 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
EP19201769.7A EP3611263A1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
AU2013355214A AU2013355214B2 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
SG11201503824SA SG11201503824SA (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
KR1020187001934A KR102006880B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
PL13859964T PL2928496T3 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
CN201810574719.4A CN108913676B (en) 2012-12-06 2013-12-05 CRISPR-based genome modification and modulation
DK13859964T DK2928496T3 (en) 2012-12-06 2013-12-05 CRISPR-BASED RE-MODIFICATION AND REGULATION
KR1020217011309A KR102479178B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
EP13859964.2A EP2928496B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
IL300199A IL300199A (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
PL16183724T PL3138911T3 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
KR1020237015558A KR20230070065A (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
KR1020157013843A KR101844123B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
PL18156734T PL3360964T3 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
EP18156734.8A EP3360964B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
EP16183724.0A EP3138911B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
KR1020207023229A KR102243092B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
BR112015012375A BR112015012375A2 (en) 2012-12-06 2013-12-05 modification and regulation of crispr genome
KR1020197022305A KR102145760B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
CA2891347A CA2891347C (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
JP2015545838A JP6620018B2 (en) 2012-12-06 2013-12-05 Genomic modification and control based on CRISPR
KR1020227043854A KR102531576B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
ES13859964T ES2757325T3 (en) 2012-12-06 2013-12-05 Modification and regulation of the genome based on CRISPR
US14/649,777 US20160017366A1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
EP16183725.7A EP3138912B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
CN201380072477.4A CN105142669B (en) 2012-12-06 2013-12-05 Genomic modification and regulation and control based on CRISPR
EP19189913.7A EP3617309A3 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
PL16183725T PL3138912T3 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
EP18160519.7A EP3363902B1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
IL238856A IL238856B (en) 2012-12-06 2015-05-17 Crispr-based genome modification and regulation
HK16106396.9A HK1218389A1 (en) 2012-12-06 2016-06-06 Crispr-based genome modification and regulation crispr
US15/188,899 US20160298125A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,927 US20160298136A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,933 US20160298138A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,911 US10731181B2 (en) 2012-12-06 2016-06-21 CRISPR-based genome modification and regulation
US15/188,909 US20160298133A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,931 US20160298137A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,924 US10745716B2 (en) 2012-12-06 2016-06-21 CRISPR-based genome modification and regulation
US15/188,902 US20160298132A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/342,976 US20170073705A1 (en) 2012-12-06 2016-11-03 Crispr-based genome modification and regulation
US15/456,204 US20170191082A1 (en) 2012-12-06 2017-03-10 Crispr-based genome modification and regulation
AU2017204031A AU2017204031B2 (en) 2012-12-06 2017-06-15 Crispr-based genome modification and regulation
IL257178A IL257178B (en) 2012-12-06 2018-01-28 Crispr-based genome modification and regulation
AU2018229489A AU2018229489B2 (en) 2012-12-06 2018-09-13 Crispr-based genome modification and regulation
AU2019201344A AU2019201344C1 (en) 2012-12-06 2019-02-26 Crispr-based genome modification and regulation
IL267598A IL267598B (en) 2012-12-06 2019-06-24 Crispr-based genome modification and regulation
US16/654,613 US20200140897A1 (en) 2012-12-06 2019-10-16 Crispr-based genome modification and regulation
US16/943,792 US20210079427A1 (en) 2012-12-06 2020-07-30 Crispr-based genome modification and regulation
US16/943,767 US20210207173A1 (en) 2012-12-06 2020-07-30 Crispr-based genome modification and regulation
AU2020230243A AU2020230243B2 (en) 2012-12-06 2020-09-08 Crispr-based genome modification and regulation
AU2020230246A AU2020230246B2 (en) 2012-12-06 2020-09-08 Crispr-based genome modification and regulation
AU2020273316A AU2020273316B2 (en) 2012-12-06 2020-11-19 Crispr-based genome modification and regulation
US17/398,648 US20210388396A1 (en) 2012-12-06 2021-08-10 Crispr-based genome modification and regulation
AU2022200330A AU2022200330B2 (en) 2012-12-06 2022-01-19 Crispr-based genome modification and regulation
IL291129A IL291129B2 (en) 2012-12-06 2022-03-06 Crispr-based genome modification and regulation
AU2023216829A AU2023216829A1 (en) 2012-12-06 2023-08-17 Crispr-based genome modification and regulation

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201261734256P 2012-12-06 2012-12-06
US61/734,256 2012-12-06
US201361758624P 2013-01-30 2013-01-30
US61/758,624 2013-01-30
US201361761046P 2013-02-05 2013-02-05
US61/761,046 2013-02-05
US201361794422P 2013-03-15 2013-03-15
US61/794,422 2013-03-15

Related Child Applications (10)

Application Number Title Priority Date Filing Date
US14/649,777 A-371-Of-International US20160017366A1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation
US15/188,931 Continuation US20160298137A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,909 Division US20160298133A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,924 Continuation US10745716B2 (en) 2012-12-06 2016-06-21 CRISPR-based genome modification and regulation
US15/188,902 Division US20160298132A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,911 Continuation US10731181B2 (en) 2012-12-06 2016-06-21 CRISPR-based genome modification and regulation
US15/188,899 Division US20160298125A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,927 Continuation US20160298136A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/188,933 Continuation US20160298138A1 (en) 2012-12-06 2016-06-21 Crispr-based genome modification and regulation
US15/342,976 Continuation US20170073705A1 (en) 2012-12-06 2016-11-03 Crispr-based genome modification and regulation

Publications (1)

Publication Number Publication Date
WO2014089290A1 true WO2014089290A1 (en) 2014-06-12

Family

ID=50883989

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/073307 WO2014089290A1 (en) 2012-12-06 2013-12-05 Crispr-based genome modification and regulation

Country Status (17)

Country Link
US (15) US20160017366A1 (en)
EP (11) EP3138912B1 (en)
JP (6) JP6620018B2 (en)
KR (7) KR102145760B1 (en)
CN (3) CN105142669B (en)
AU (9) AU2013355214B2 (en)
BR (1) BR112015012375A2 (en)
CA (3) CA3034794A1 (en)
DK (6) DK3363902T3 (en)
ES (6) ES2769310T3 (en)
HK (1) HK1218389A1 (en)
IL (5) IL300199A (en)
LT (4) LT3138912T (en)
PL (6) PL3138911T3 (en)
PT (6) PT3360964T (en)
SG (4) SG10201910987SA (en)
WO (1) WO2014089290A1 (en)

Cited By (253)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2764103A2 (en) 2012-12-12 2014-08-13 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
US8865406B2 (en) 2012-12-12 2014-10-21 The Broad Institute Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US8871445B2 (en) 2012-12-12 2014-10-28 The Broad Institute Inc. CRISPR-Cas component systems, methods and compositions for sequence manipulation
EP2800811A1 (en) 2012-05-25 2014-11-12 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US8889356B2 (en) 2012-12-12 2014-11-18 The Broad Institute Inc. CRISPR-Cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
US20140356958A1 (en) * 2012-12-17 2014-12-04 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US8906616B2 (en) 2012-12-12 2014-12-09 The Broad Institute Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP2828386A1 (en) 2012-03-20 2015-01-28 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX
US8993233B2 (en) 2012-12-12 2015-03-31 The Broad Institute Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
EP2784162B1 (en) 2012-12-12 2015-04-08 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
WO2015088643A1 (en) 2013-12-11 2015-06-18 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
EP2922393A1 (en) 2013-02-27 2015-09-30 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Gene editing in the oocyte by cas9 nucleases
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
EP2931897A2 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
EP2912175A4 (en) * 2012-10-23 2015-12-09 Toolgen Inc Composition for cleaving a target dna comprising a guide rna specific for the target dna and cas protein-encoding nucleic acid or cas protein, and use thereof
WO2015188109A1 (en) 2014-06-06 2015-12-10 Regeneron Pharmaceuticals, Inc. Methods and compositions for modifying a targeted locus
WO2015195621A1 (en) * 2014-06-16 2015-12-23 The Johns Hopkins University Compositions and methods for the expression of crispr guide rnas using the h1 promoter
WO2015200805A2 (en) 2014-06-26 2015-12-30 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modifications and methods of use
WO2015200334A1 (en) * 2014-06-23 2015-12-30 Regeneron Pharmaceuticals, Inc. Nuclease-mediated dna assembly
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US9260752B1 (en) 2013-03-14 2016-02-16 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
WO2016011080A3 (en) * 2014-07-14 2016-03-03 The Regents Of The University Of California Crispr/cas transcriptional modulation
WO2016036754A1 (en) 2014-09-02 2016-03-10 The Regents Of The University Of California Methods and compositions for rna-directed target dna modification
US20160090607A1 (en) * 2013-05-15 2016-03-31 Sangamo Biosciences, Inc. Methods and compositions for treatment of a genetic condition
EP3004349A1 (en) * 2013-05-29 2016-04-13 Cellectis S.A. A method for producing precise dna cleavage using cas9 nickase activity
WO2016061374A1 (en) 2014-10-15 2016-04-21 Regeneron Pharmaceuticals, Inc. Methods and compositions for generating or maintaining pluripotent cells
JP2016512264A (en) * 2013-03-15 2016-04-25 ザ ジェネラル ホスピタル コーポレイション RNA-induced targeting of genomic and epigenomic regulatory proteins to specific genomic loci
JP2016512048A (en) * 2013-03-15 2016-04-25 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Plant genome manipulation using the CRISPR / Cas system
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
EP3011030A1 (en) * 2013-06-17 2016-04-27 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
WO2016073433A1 (en) * 2014-11-06 2016-05-12 E. I. Du Pont De Nemours And Company Peptide-mediated delivery of rna-guided endonuclease into cells
WO2016081923A2 (en) 2014-11-21 2016-05-26 Regeneron Pharmaceuticals, Inc. METHODS AND COMPOSITIONS FOR TARGETED GENETIC MODIFICATION USING PAIRED GUIDE RNAs
WO2016080795A1 (en) * 2014-11-19 2016-05-26 기초과학연구원 Method for regulating gene expression using cas9 protein expressed from two vectors
WO2016083811A1 (en) 2014-11-27 2016-06-02 Imperial Innovations Limited Genome editing methods
WO2016100857A1 (en) 2014-12-19 2016-06-23 Regeneron Pharmaceuticals, Inc. Stem cells for modeling type 2 diabetes
US20160186213A1 (en) * 2013-06-17 2016-06-30 The Broad Institute Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
US20160186208A1 (en) * 2013-04-16 2016-06-30 Whitehead Institute For Biomedical Research Methods of Mutating, Modifying or Modulating Nucleic Acid in a Cell or Nonhuman Mammal
GB2534074A (en) * 2011-12-30 2016-07-13 Caribou Biosciences Inc Modified cascade ribonucleoproteins and uses thereof
JP2016521554A (en) * 2013-06-04 2016-07-25 プレジデント アンド フェローズ オブ ハーバード カレッジ RNA-induced transcriptional regulation
JP2016521995A (en) * 2013-06-17 2016-07-28 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, use and therapeutic applications of CRISPR-Cas systems and compositions for targeting disorders and diseases using viral components
WO2016120480A1 (en) 2015-01-29 2016-08-04 Meiogenix Method for inducing targeted meiotic recombinations
JP2016523082A (en) * 2013-06-17 2016-08-08 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of mitotic cells
JP2016524472A (en) * 2013-06-17 2016-08-18 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery and use of CRISPR-Cas systems, vectors and compositions for liver targeting and therapy
JP2016528894A (en) * 2013-07-26 2016-09-23 プレジデント アンド フェローズ オブ ハーバード カレッジ Genome engineering
WO2016176617A2 (en) 2015-04-29 2016-11-03 New York University Method for treating high-grade gliomas
WO2016178207A1 (en) 2015-05-04 2016-11-10 Ramot At Tel-Aviv University Ltd. Methods and kits for fragmenting dna
US9512446B1 (en) 2015-08-28 2016-12-06 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
WO2016205554A1 (en) * 2015-06-17 2016-12-22 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
WO2016210271A1 (en) * 2015-06-24 2016-12-29 Sigma-Aldrich Co. Llc Cell cycle dependent genome regulation and modification
US20170007679A1 (en) * 2014-03-25 2017-01-12 Editas Medicine Inc. Crispr/cas-related methods and compositions for treating hiv infection and aids
WO2017027910A1 (en) 2015-08-14 2017-02-23 The University Of Sydney Connexin 45 inhibition for therapy
WO2017040348A1 (en) 2015-08-28 2017-03-09 The General Hospital Corporation Engineered crispr-cas9 nucleases
US20170121693A1 (en) * 2015-10-23 2017-05-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2017075335A1 (en) 2015-10-28 2017-05-04 Voyager Therapeutics, Inc. Regulatable expression using adeno-associated virus (aav)
JP2017518082A (en) * 2014-06-17 2017-07-06 ポセイダ セラピューティクス, インコーポレイテッド Methods and uses for directing proteins to specific loci in the genome
US20170198268A1 (en) * 2014-07-09 2017-07-13 Gen9, Inc. Compositions and Methods for Site-Directed DNA Nicking and Cleaving
JP2017526387A (en) * 2014-08-06 2017-09-14 トゥールジェン インコーポレイテッド Genome editing using RGEN derived from Campylobacter jejuni CRISPR / CAS system
EP3219799A1 (en) 2016-03-17 2017-09-20 IMBA-Institut für Molekulare Biotechnologie GmbH Conditional crispr sgrna expression
EP3138910B1 (en) 2012-12-06 2017-09-20 Sigma-Aldrich Co. LLC Crispr-based genome modification and regulation
WO2017186718A1 (en) * 2016-04-25 2017-11-02 Universität Basel Allele editing and applications thereof
WO2017201476A1 (en) 2016-05-20 2017-11-23 Regeneron Pharmaceuticals, Inc. Methods for breaking immunological tolerance using multiple guide rnas
CN107429246A (en) * 2014-10-31 2017-12-01 麻省理工学院 Large-scale parallel for CRISPR combines science of heredity
US9834786B2 (en) 2012-04-25 2017-12-05 Regeneron Pharmaceuticals, Inc. Nuclease-mediated targeting with large targeting vectors
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
EP3152319A4 (en) * 2014-06-05 2017-12-27 Sangamo BioSciences, Inc. Methods and compositions for nuclease design
EP3272867A1 (en) * 2016-06-02 2018-01-24 Sigma-Aldrich Co. LLC Using programmable dna binding proteins to enhance targeted genome modification
WO2018023014A1 (en) 2016-07-29 2018-02-01 Regeneron Pharmaceuticals, Inc. Mice comprising mutations resulting in expression of c-truncated fibrillin-1
WO2018022967A1 (en) 2016-07-28 2018-02-01 Regeneron Pharmaceuticals, Inc. Gpr156 variants and uses thereof
US9888673B2 (en) 2014-12-10 2018-02-13 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US9926546B2 (en) 2015-08-28 2018-03-27 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
WO2018054911A1 (en) 2016-09-23 2018-03-29 Bayer Cropscience Nv Targeted genome optimization in plants
US9938521B2 (en) 2014-03-10 2018-04-10 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating leber's congenital amaurosis 10 (LCA10)
EP3204399A4 (en) * 2014-10-09 2018-04-25 Seattle Children's Hospital, dba Seattle Children's Research Institute Long poly (a) plasmids and methods for introduction of long poly (a) sequences into the plasmid
WO2018073237A1 (en) 2016-10-17 2018-04-26 The University Court Of The University Of Edinburgh Swine comprising modified cd163 and associated methods
US10011850B2 (en) 2013-06-21 2018-07-03 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to increase specificity for RNA-Guided Genome Editing
WO2018136758A1 (en) 2017-01-23 2018-07-26 Regeneron Pharmaceuticals, Inc. Hsd17b13 variants and uses thereof
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10081807B2 (en) 2012-04-24 2018-09-25 Gen9, Inc. Methods for sorting nucleic acids and multiplexed preparative in vitro cloning
WO2018195545A2 (en) 2017-04-21 2018-10-25 The General Hospital Corporation Variants of cpf1 (cas12a) with altered pam specificity
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
EP3008186B1 (en) 2013-06-14 2018-11-28 Cellectis Methods for non-transgenic genome editing in plants
WO2018218206A1 (en) 2017-05-25 2018-11-29 The General Hospital Corporation Bipartite base editor (bbe) architectures and type-ii-c-cas9 zinc finger editing
WO2018226560A1 (en) 2017-06-05 2018-12-13 Regeneron Pharmaceuticals, Inc. B4galt1 variants and uses thereof
US10166255B2 (en) 2015-07-31 2019-01-01 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
WO2019006034A1 (en) 2017-06-27 2019-01-03 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized asgr1 locus
WO2019028023A2 (en) 2017-07-31 2019-02-07 Regeneron Pharmaceuticals, Inc. Methods and compositions for assessing crispr/cas-mediated disruption or excision and crispr/cas-induced recombination with an exogenous donor nucleic acid in vivo
WO2019028029A1 (en) 2017-07-31 2019-02-07 Regeneron Pharmaceuticals, Inc. Assessment of crispr/cas-induced recombination with an exogenous donor nucleic acid in vivo
WO2019028032A1 (en) 2017-07-31 2019-02-07 Regeneron Pharmaceuticals, Inc. Cas-transgenic mouse embryonic stem cells and mice and uses thereof
US10202608B2 (en) 2006-08-31 2019-02-12 Gen9, Inc. Iterative nucleic acid assembly using activation of vector-encoded traits
WO2019050948A1 (en) * 2017-09-05 2019-03-14 Regeneron Pharmaceuticals, Inc. Delivery of a gene-editing system with a single retroviral particle and methods of generation and use
WO2019051033A1 (en) 2017-09-07 2019-03-14 Regeneron Pharmaceuticals, Inc. Solute carrier family 14 member 1 (slc14a1) variants and uses thereof
WO2019050899A1 (en) 2017-09-06 2019-03-14 Regeneron Pharmaceuticals, Inc. Single immunoglobulin interleukin-1 receptor related (sigirr) variants and uses thereof
US10240145B2 (en) * 2015-11-25 2019-03-26 The Board Of Trustees Of The Leland Stanford Junior University CRISPR/Cas-mediated genome editing to treat EGFR-mutant lung cancer
EP3460063A1 (en) 2013-12-11 2019-03-27 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
WO2019067875A1 (en) 2017-09-29 2019-04-04 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus and methods of use
WO2019079238A1 (en) 2017-10-16 2019-04-25 Regeneron Pharmaceuticals, Inc. Cornulin (crnn) variants and uses thereof
WO2019094735A1 (en) 2017-11-10 2019-05-16 Regeneron Pharmaceuticals, Inc. Non-human animals comprising slc30a8 mutation and methods of use
US10308931B2 (en) 2012-03-21 2019-06-04 Gen9, Inc. Methods for screening proteins using DNA encoded chemical libraries as templates for enzyme catalysis
WO2019108983A1 (en) 2017-11-30 2019-06-06 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized trkb locus
EP3494997A1 (en) * 2012-07-25 2019-06-12 The Broad Institute, Inc. Inducible dna binding proteins and genome perturbation tools and applications thereof
WO2019123014A1 (en) * 2017-12-22 2019-06-27 G+Flas Life Sciences Chimeric genome engineering molecules and methods
WO2019126578A1 (en) * 2017-12-20 2019-06-27 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
US10337001B2 (en) 2014-12-03 2019-07-02 Agilent Technologies, Inc. Guide RNA with chemical modifications
EP3362571A4 (en) * 2015-10-13 2019-07-10 Duke University Genome engineering with type i crispr systems in eukaryotic cells
WO2019138083A1 (en) 2018-01-12 2019-07-18 Basf Se Gene underlying the number of spikelets per spike qtl in wheat on chromosome 7a
US10377998B2 (en) 2013-12-12 2019-08-13 The Broad Institute, Inc. CRISPR-CAS systems and methods for altering expression of gene products, structural information and inducible modular CAS enzymes
US10385359B2 (en) 2013-04-16 2019-08-20 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US10415024B2 (en) 2012-11-16 2019-09-17 Poseida Therapeutics, Inc. Site-specific enzymes and methods of use
WO2019183123A1 (en) 2018-03-19 2019-09-26 Regeneron Pharmaceuticals, Inc. Transcription modulation in animals using crispr/cas systems
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
US10457935B2 (en) 2010-11-12 2019-10-29 Gen9, Inc. Protein arrays and methods of using and making the same
WO2019217803A1 (en) 2018-05-10 2019-11-14 Auxolytic Ltd. Gene therapy methods and compositions using auxotrophic regulatable cells
US10494621B2 (en) 2015-06-18 2019-12-03 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
WO2019234754A1 (en) 2018-06-07 2019-12-12 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Nucleic acid constructs and methods of using same
WO2019234750A1 (en) 2018-06-07 2019-12-12 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Methods of regenerating and transforming cannabis
US10519457B2 (en) 2013-08-22 2019-12-31 E I Du Pont De Nemours And Company Soybean U6 polymerase III promoter and methods of use
US10526589B2 (en) 2013-03-15 2020-01-07 The General Hospital Corporation Multiplex guide RNAs
US10550372B2 (en) 2013-12-12 2020-02-04 The Broad Institute, Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
WO2020030783A2 (en) 2018-08-10 2020-02-13 Vib Vzw Means and methods for drought tolerance in crops
WO2020041313A1 (en) * 2018-08-21 2020-02-27 Sigma-Aldrich Co. Llc Down-regulation of the cytosolic dna sensor pathway
WO2020056122A1 (en) 2018-09-13 2020-03-19 Regeneron Pharmaceuticals, Inc. Complement factor h gene knockout rat as a model of c3 glomerulopathy
US10640789B2 (en) 2013-06-04 2020-05-05 President And Fellows Of Harvard College RNA-guided transcriptional regulation
EP3653048A1 (en) 2014-12-19 2020-05-20 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modification through single-step multiple targeting
WO2020123377A1 (en) 2018-12-10 2020-06-18 Neoimmunetech, Inc. Nrf-2 deficient cells and uses thereof
WO2020128478A1 (en) 2018-12-19 2020-06-25 King's College London Immunotherapeutic methods and compositions
WO2020131632A1 (en) 2018-12-20 2020-06-25 Regeneron Pharmaceuticals, Inc. Nuclease-mediated repeat expansion
US10696986B2 (en) 2014-12-12 2020-06-30 The Board Institute, Inc. Protected guide RNAS (PGRNAS)
WO2020163396A1 (en) 2019-02-04 2020-08-13 The General Hospital Corporation Adenine dna base editor variants with reduced off-target rna editing
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
WO2020169974A1 (en) 2019-02-19 2020-08-27 King's College London Hypoxia-responsive chimeric antigen receptors
US10767193B2 (en) 2018-02-15 2020-09-08 Sigma-Aldrich Co. Llc Engineered CAS9 systems for eukaryotic genome modification
US10767175B2 (en) 2016-06-08 2020-09-08 Agilent Technologies, Inc. High specificity genome editing using chemically modified guide RNAs
US10781444B2 (en) 2013-06-17 2020-09-22 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
WO2020190932A1 (en) 2019-03-18 2020-09-24 Regeneron Pharmaceuticals, Inc. Crispr/cas screening platform to identify genetic modifiers of tau seeding or aggregation
WO2020190927A1 (en) 2019-03-18 2020-09-24 Regeneron Pharmaceuticals, Inc. Crispr/cas dropout screening platform to reveal genetic vulnerabilities associated with tau aggregation
WO2020206162A1 (en) 2019-04-03 2020-10-08 Regeneron Pharmaceuticals, Inc. Methods and compositions for insertion of antibody coding sequences into a safe harbor locus
WO2020206134A1 (en) 2019-04-04 2020-10-08 Regeneron Pharmaceuticals, Inc. Methods for scarless introduction of targeted modifications into targeting vectors
WO2020206139A1 (en) 2019-04-04 2020-10-08 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized coagulation factor 12 locus
WO2020212541A1 (en) 2019-04-16 2020-10-22 University Of Nottingham Fungal strains, production and uses thereof
WO2020229241A1 (en) 2019-05-10 2020-11-19 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
US10851357B2 (en) 2013-12-12 2020-12-01 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
WO2020247812A1 (en) 2019-06-07 2020-12-10 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized albumin locus
WO2020247452A1 (en) 2019-06-04 2020-12-10 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus with a beta-slip mutation and methods of use
WO2020252340A1 (en) 2019-06-14 2020-12-17 Regeneron Pharmaceuticals, Inc. Models of tauopathy
CN112105732A (en) * 2018-05-10 2020-12-18 先正达参股股份有限公司 Methods and compositions for targeted editing of polynucleotides
US10912797B2 (en) 2016-10-18 2021-02-09 Intima Bioscience, Inc. Tumor infiltrating lymphocytes and methods of therapy
US10930367B2 (en) 2012-12-12 2021-02-23 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and conveying results thereof
US10934536B2 (en) 2018-12-14 2021-03-02 Pioneer Hi-Bred International, Inc. CRISPR-CAS systems for genome editing
US10947517B2 (en) 2019-02-15 2021-03-16 Sigma-Aldrich Co. Llc CRISPR/Cas fusion proteins and systems
WO2021050398A1 (en) * 2019-09-10 2021-03-18 The Regents Of The University Of California Synthetic lethality screening platform for cells undergoing alt
WO2021048316A1 (en) 2019-09-12 2021-03-18 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2021050940A1 (en) 2019-09-13 2021-03-18 Regeneron Pharmaceuticals, Inc. Transcription modulation in animals using crispr/cas systems delivered by lipid nanoparticles
WO2021069387A1 (en) 2019-10-07 2021-04-15 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
GB202103131D0 (en) 2021-03-05 2021-04-21 Biosystems Tech Limited Method for preparation of research organisms
WO2021092513A1 (en) 2019-11-08 2021-05-14 Regeneron Pharmaceuticals, Inc. Crispr and aav strategies for x-linked juvenile retinoschisis therapy
WO2021108363A1 (en) 2019-11-25 2021-06-03 Regeneron Pharmaceuticals, Inc. Crispr/cas-mediated upregulation of humanized ttr allele
US11028388B2 (en) 2014-03-05 2021-06-08 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for treating Usher syndrome and retinitis pigmentosa
WO2021110582A1 (en) 2019-12-03 2021-06-10 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
US11033584B2 (en) 2017-10-27 2021-06-15 The Regents Of The University Of California Targeted replacement of endogenous T cell receptors
US11046948B2 (en) 2013-08-22 2021-06-29 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US11072789B2 (en) 2012-06-25 2021-07-27 Gen9, Inc. Methods for nucleic acid assembly and high throughput sequencing
US11078483B1 (en) 2016-09-02 2021-08-03 KSQ Therapeutics, Inc. Methods for measuring and improving CRISPR reagent function
US11078481B1 (en) 2016-08-03 2021-08-03 KSQ Therapeutics, Inc. Methods for screening for cancer targets
WO2021154791A1 (en) 2020-01-28 2021-08-05 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized pnpla3 locus and methods of use
US11084014B2 (en) 2010-11-12 2021-08-10 Gen9, Inc. Methods and devices for nucleic acids synthesis
WO2021158883A1 (en) 2020-02-07 2021-08-12 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized klkb1 locus and methods of use
US11098325B2 (en) 2017-06-30 2021-08-24 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
US11111521B2 (en) 2011-12-22 2021-09-07 President And Fellows Of Harvard College Compositions and methods for analyte detection
WO2021178556A1 (en) 2020-03-04 2021-09-10 Regeneron Pharmaceuticals, Inc. Methods and compositions for sensitization of tumor cells to immune therapy
WO2021195079A1 (en) 2020-03-23 2021-09-30 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus comprising a v30m mutation and methods of use
WO2021202938A1 (en) 2020-04-03 2021-10-07 Creyon Bio, Inc. Oligonucleotide-based machine learning
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11149267B2 (en) 2013-10-28 2021-10-19 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
US11155795B2 (en) 2013-12-12 2021-10-26 The Broad Institute, Inc. CRISPR-Cas systems, crystal structure and uses thereof
US11180793B2 (en) 2015-04-24 2021-11-23 Editas Medicine, Inc. Evaluation of Cas9 molecule/guide RNA molecule complexes
WO2021263146A2 (en) 2020-06-26 2021-12-30 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ace2 locus
GB202118058D0 (en) 2021-12-14 2022-01-26 Univ Warwick Methods to increase yields in crops
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11242525B2 (en) 2014-03-26 2022-02-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating sickle cell disease
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11299767B2 (en) 2013-03-12 2022-04-12 President And Fellows Of Harvard College Method for generating a three-dimensional nucleic acid containing matrix
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
US11306309B2 (en) 2015-04-06 2022-04-19 The Board Of Trustees Of The Leland Stanford Junior University Chemically modified guide RNAs for CRISPR/CAS-mediated gene regulation
EP3988649A1 (en) * 2013-09-18 2022-04-27 Kymab Limited Methods, cells and organisms
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
EP3998344A1 (en) * 2014-10-09 2022-05-18 Life Technologies Corporation Crispr oligonucleotides and gene editing
US11339437B2 (en) 2014-03-10 2022-05-24 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
WO2022120022A1 (en) 2020-12-02 2022-06-09 Regeneron Pharmaceuticals, Inc. Crispr sam biosensor cell lines and methods of use thereof
US11384360B2 (en) 2012-06-19 2022-07-12 Regents Of The University Of Minnesota Gene targeting in plants using DNA viruses
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11407985B2 (en) 2013-12-12 2022-08-09 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for genome editing
US11427817B2 (en) 2015-08-25 2022-08-30 Duke University Compositions and methods of improving specificity in genomic engineering using RNA-guided endonucleases
US11434491B2 (en) 2018-04-19 2022-09-06 The Regents Of The University Of California Compositions and methods for gene editing
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
WO2022240846A1 (en) 2021-05-10 2022-11-17 Sqz Biotechnologies Company Methods for delivering genome editing molecules to the nucleus or cytosol of a cell and uses thereof
US11512311B2 (en) 2016-03-25 2022-11-29 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (A1AT) deficiency
WO2022251644A1 (en) 2021-05-28 2022-12-01 Lyell Immunopharma, Inc. Nr4a3-deficient immune cells and uses thereof
WO2022256437A1 (en) 2021-06-02 2022-12-08 Lyell Immunopharma, Inc. Nr4a3-deficient immune cells and uses thereof
US11542554B2 (en) 2015-11-03 2023-01-03 President And Fellows Of Harvard College Method and apparatus for volumetric imaging
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
US11560568B2 (en) 2014-09-12 2023-01-24 E. I. Du Pont De Nemours And Company Generation of site-specific-integration sites for complex trait loci in corn and soybean, and methods of use
US11566263B2 (en) 2016-08-02 2023-01-31 Editas Medicine, Inc. Compositions and methods for treating CEP290 associated disease
US11578312B2 (en) 2015-06-18 2023-02-14 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of CAS9 orthologs and variants for sequence manipulation
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
WO2023064924A1 (en) 2021-10-14 2023-04-20 Codiak Biosciences, Inc. Modified producer cells for extracellular vesicle production
WO2023077053A2 (en) 2021-10-28 2023-05-04 Regeneron Pharmaceuticals, Inc. Crispr/cas-related methods and compositions for knocking out c5
WO2023081847A1 (en) 2021-11-04 2023-05-11 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a modified cacng1 locus
US20230151345A1 (en) * 2014-10-24 2023-05-18 Life Technologies Corporation Compositions and methods for enhancing homologous recombination
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
WO2023108047A1 (en) 2021-12-08 2023-06-15 Regeneron Pharmaceuticals, Inc. Mutant myocilin disease model and uses thereof
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
EP4198124A1 (en) 2021-12-15 2023-06-21 Versitech Limited Engineered cas9-nucleases and method of use thereof
WO2023122506A1 (en) 2021-12-20 2023-06-29 Regeneron Pharmaceuticals, Inc. Non-human animals comprising humanized ace2 and tmprss loci
WO2023129974A1 (en) 2021-12-29 2023-07-06 Bristol-Myers Squibb Company Generation of landing pad cell lines
US11702662B2 (en) 2011-08-26 2023-07-18 Gen9, Inc. Compositions and methods for high fidelity assembly of nucleic acids
US11713485B2 (en) 2016-04-25 2023-08-01 President And Fellows Of Harvard College Hybridization chain reaction methods for in situ molecular detection
WO2023150798A1 (en) 2022-02-07 2023-08-10 Regeneron Pharmaceuticals, Inc. Compositions and methods for defining optimal treatment timeframes in lysosomal disease
WO2023150181A1 (en) 2022-02-01 2023-08-10 President And Fellows Of Harvard College Methods and compositions for treating cancer
WO2023150620A1 (en) 2022-02-02 2023-08-10 Regeneron Pharmaceuticals, Inc. Crispr-mediated transgene insertion in neonatal cells
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
WO2023212677A2 (en) 2022-04-29 2023-11-02 Regeneron Pharmaceuticals, Inc. Identification of tissue-specific extragenic safe harbors for gene therapy approaches
US11814624B2 (en) 2017-06-15 2023-11-14 The Regents Of The University Of California Targeted non-viral DNA insertions
WO2023220603A1 (en) 2022-05-09 2023-11-16 Regeneron Pharmaceuticals, Inc. Vectors and methods for in vivo antibody production
WO2023225665A1 (en) 2022-05-19 2023-11-23 Lyell Immunopharma, Inc. Polynucleotides targeting nr4a3 and uses thereof
WO2023235726A2 (en) 2022-05-31 2023-12-07 Regeneron Pharmaceuticals, Inc. Crispr interference therapeutics for c9orf72 repeat expansion disease
WO2023235725A2 (en) 2022-05-31 2023-12-07 Regeneron Pharmaceuticals, Inc. Crispr-based therapeutics for c9orf72 repeat expansion disease
US11851690B2 (en) 2017-03-14 2023-12-26 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
US11859219B1 (en) 2016-12-30 2024-01-02 Flagship Pioneering Innovations V, Inc. Methods of altering a target nucleotide sequence with an RNA-guided nuclease and a single guide RNA
WO2024003579A1 (en) 2022-06-30 2024-01-04 University Of Newcastle Upon Tyne Preventing disease recurrence in mitochondrial replacement therapy
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11884915B2 (en) 2021-09-10 2024-01-30 Agilent Technologies, Inc. Guide RNAs with chemical modification for prime editing
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
WO2024026488A2 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a modified transferrin receptor locus
WO2024031053A1 (en) 2022-08-05 2024-02-08 Regeneron Pharmaceuticals, Inc. Aggregation-resistant variants of tdp-43
US11898176B2 (en) 2012-04-27 2024-02-13 Duke University Genetic correction of mutated genes
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
US11913015B2 (en) 2017-04-17 2024-02-27 University Of Maryland, College Park Embryonic cell cultures and methods of using the same
WO2024064952A1 (en) 2022-09-23 2024-03-28 Lyell Immunopharma, Inc. Methods for culturing nr4a-deficient cells overexpressing c-jun
WO2024064958A1 (en) 2022-09-23 2024-03-28 Lyell Immunopharma, Inc. Methods for culturing nr4a-deficient cells
WO2024073606A1 (en) 2022-09-28 2024-04-04 Regeneron Pharmaceuticals, Inc. Antibody resistant modified receptors to enhance cell-based therapies
WO2024077174A1 (en) 2022-10-05 2024-04-11 Lyell Immunopharma, Inc. Methods for culturing nr4a-deficient cells
US11965184B2 (en) 2021-02-10 2024-04-23 Sigma-Aldrich Co. Llc CRISPR/Cas fusion proteins and systems

Families Citing this family (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3417880A1 (en) * 2013-06-05 2018-12-26 Duke University Rna-guided gene editing and gene regulation
US10584358B2 (en) 2013-10-30 2020-03-10 North Carolina State University Compositions and methods related to a type-II CRISPR-Cas system in Lactobacillus buchneri
US10787654B2 (en) * 2014-01-24 2020-09-29 North Carolina State University Methods and compositions for sequence guiding Cas9 targeting
CA3075047C (en) 2014-02-11 2022-02-01 The Regents Of The University Of Colorado, A Body Corporate Crispr enable method for multiplex genome editing
US10286084B2 (en) 2014-02-18 2019-05-14 Duke University Compositions for the inactivation of virus replication and methods of making and using the same
JP2017512481A (en) 2014-04-08 2017-05-25 ノースカロライナ ステート ユニバーシティーNorth Carolina State University Methods and compositions for RNA-dependent transcriptional repression using CRISPR-related genes
EP3633032A3 (en) 2014-08-28 2020-07-29 North Carolina State University Novel cas9 proteins and guiding features for dna targeting and genome editing
CN107532142A (en) * 2014-11-11 2018-01-02 应用干细胞有限公司 Mescenchymal stem cell is transformed using homologous recombination
EP3303607A4 (en) 2015-05-29 2018-10-10 North Carolina State University Methods for screening bacteria, archaea, algae, and yeast using crispr nucleic acids
EP3303585A4 (en) 2015-06-03 2018-10-31 Board of Regents of the University of Nebraska Dna editing using single-stranded dna
JP7051438B2 (en) 2015-06-15 2022-04-11 ノース カロライナ ステート ユニバーシティ Methods and Compositions for Efficient Delivery of Nucleic Acid and RNA-Based Antibacterial Agents
KR101795999B1 (en) 2015-09-25 2017-11-09 전남대학교산학협력단 Primer for Beta2-Microglobulin gene remove using CRISPR/CAS9 system
KR101745863B1 (en) 2015-09-25 2017-06-12 전남대학교산학협력단 Primer for prohibitin2 gene remove using CRISPR/CAS9 system
EP3356533A1 (en) 2015-09-28 2018-08-08 North Carolina State University Methods and compositions for sequence specific antimicrobials
PL3846375T3 (en) 2015-10-22 2023-04-24 Telefonaktiebolaget Lm Ericsson (Publ) Methods and apparatus relating to selective enhancement of radio signals
US11905521B2 (en) 2015-11-17 2024-02-20 The Chinese University Of Hong Kong Methods and systems for targeted gene manipulation
US11542466B2 (en) 2015-12-22 2023-01-03 North Carolina State University Methods and compositions for delivery of CRISPR based antimicrobials
EP3901258A1 (en) 2016-01-11 2021-10-27 The Board of Trustees of the Leland Stanford Junior University Chimeric proteins and methods of immunotherapy
JP7015239B2 (en) 2016-01-11 2022-03-04 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー How to regulate chimeric protein and gene expression
JP7197363B2 (en) * 2016-04-14 2022-12-27 ボコ シリコン バレー, インコーポレイテッド Genome editing of human neural stem cells using nucleases
CA3025591A1 (en) * 2016-06-03 2017-12-07 Philippe Ravassard Diet controlled expression of a nucleic acid encoding cas9 nuclease and uses thereof
AU2017277810A1 (en) * 2016-06-03 2018-12-06 Temple University - Of The Commonwealth System Of Higher Education Negative feedback regulation of HIV-1 by gene editing strategy
US10337051B2 (en) 2016-06-16 2019-07-02 The Regents Of The University Of California Methods and compositions for detecting a target RNA
US11293021B1 (en) 2016-06-23 2022-04-05 Inscripta, Inc. Automated cell processing methods, modules, instruments, and systems
LT3474669T (en) 2016-06-24 2022-06-10 The Regents Of The University Of Colorado, A Body Corporate Methods for generating barcoded combinatorial libraries
WO2018013720A1 (en) * 2016-07-12 2018-01-18 Washington University Incorporation of internal polya-encoded poly-lysine sequence tags and their variations for the tunable control of protein synthesis in bacterial and eukaryotic cells
EP3500677A4 (en) * 2016-08-20 2020-04-01 Avellino Lab USA, Inc. Single guide rna, crispr/cas9 systems, and methods of use thereof
US20180105806A1 (en) * 2016-09-07 2018-04-19 Massachusetts Institute Of Technology Method for rna-guided endonuclease-based dna assembly
CN106636197B (en) * 2016-09-22 2019-09-03 南京市妇幼保健院 A method of orientation strikes multi-copy gene in drop zebra fish genome
CN110023494A (en) 2016-09-30 2019-07-16 加利福尼亚大学董事会 The nucleic acid modifying enzyme and its application method of RNA guidance
WO2018071572A1 (en) * 2016-10-11 2018-04-19 Stemgenics, Inc. Nanoparticles functionalized with gene editing tools and related methods
IL266362B1 (en) * 2016-11-02 2024-03-01 Univ Basel Immunologically discernible cell surface variants for use in cell therapy
KR102151065B1 (en) * 2016-12-23 2020-09-02 기초과학연구원 Composition and method for base editing in animal embryos
CN106978438B (en) * 2017-02-27 2020-08-28 北京大北农生物技术有限公司 Method for improving homologous recombination efficiency
US11834670B2 (en) 2017-04-19 2023-12-05 Global Life Sciences Solutions Usa Llc Site-specific DNA modification using a donor DNA repair template having tandem repeat sequences
US9982279B1 (en) 2017-06-23 2018-05-29 Inscripta, Inc. Nucleic acid-guided nucleases
US10011849B1 (en) 2017-06-23 2018-07-03 Inscripta, Inc. Nucleic acid-guided nucleases
US20190002874A1 (en) 2017-06-30 2019-01-03 Inscripta, Inc. Cell libraries created using rationally designed nucleic acids
KR20200017479A (en) * 2017-07-31 2020-02-18 시그마-알드리치 컴퍼니., 엘엘씨 Synthetic Induced RNA for CRISPR / CAS Activator Systems
ES2960390T3 (en) * 2017-08-09 2024-03-04 Ricetec Inc Compositions and methods to modify genomes
US10738327B2 (en) 2017-08-28 2020-08-11 Inscripta, Inc. Electroporation cuvettes for automation
US10435713B2 (en) 2017-09-30 2019-10-08 Inscripta, Inc. Flow through electroporation instrumentation
WO2019079195A1 (en) * 2017-10-16 2019-04-25 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Genetically modified mesenchymal stem cells for use in cardiovascular prosthetics
US20210180059A1 (en) * 2017-11-16 2021-06-17 Astrazeneca Ab Compositions and methods for improving the efficacy of cas9-based knock-in strategies
US20210047632A1 (en) * 2018-01-26 2021-02-18 The Children's Medical Center Corporation Targeting bcl11a distal regulatory elements with a cas9-cas9 fusion for fetal hemoglobin reinduction
CN112204131A (en) 2018-03-29 2021-01-08 因思科瑞普特公司 Automated control of cell growth rate for induction and transformation
WO2019200004A1 (en) 2018-04-13 2019-10-17 Inscripta, Inc. Automated cell processing instruments comprising reagent cartridges
US10501738B2 (en) 2018-04-24 2019-12-10 Inscripta, Inc. Automated instrumentation for production of peptide libraries
US10858761B2 (en) 2018-04-24 2020-12-08 Inscripta, Inc. Nucleic acid-guided editing of exogenous polynucleotides in heterologous cells
US10557216B2 (en) 2018-04-24 2020-02-11 Inscripta, Inc. Automated instrumentation for production of T-cell receptor peptide libraries
WO2019209912A2 (en) * 2018-04-27 2019-10-31 Seattle Children's Hospital (dba Seattle Children's Research Institute) Therapeutic genome editing in x-linked hyper igm syndrome
JP2021523745A (en) 2018-05-16 2021-09-09 シンテゴ コーポレイション Methods and systems for guide RNA design and use
CN108624622A (en) * 2018-05-16 2018-10-09 湖南艾佳生物科技股份有限公司 A kind of genetically engineered cell strain that can secrete mouse interleukin -6 based on CRISPR-Cas9 systems structure
EP3575402A1 (en) * 2018-06-01 2019-12-04 Algentech SAS Gene targeting
EP3810801B1 (en) * 2018-06-25 2022-09-28 Bionano Genomics, Inc. Labeling of dna
CN114854720A (en) 2018-06-30 2022-08-05 因思科瑞普特公司 Apparatus, modules and methods for improved detection of editing sequences in living cells
KR20210041008A (en) * 2018-08-03 2021-04-14 빔 테라퓨틱스, 인크. Multi-effector nucleobase editor for modifying nucleic acid target sequences and methods of using the same
US10752874B2 (en) 2018-08-14 2020-08-25 Inscripta, Inc. Instruments, modules, and methods for improved detection of edited sequences in live cells
US11142740B2 (en) 2018-08-14 2021-10-12 Inscripta, Inc. Detection of nuclease edited sequences in automated modules and instruments
US10532324B1 (en) 2018-08-14 2020-01-14 Inscripta, Inc. Instruments, modules, and methods for improved detection of edited sequences in live cells
KR102103104B1 (en) * 2018-08-16 2020-04-22 (주)라트바이오 Genetically modified transgenic animals and transgenic embryos
CN109055379B (en) * 2018-09-10 2022-04-15 石铭 Preparation method of transgenic chicken oviduct bioreactor
KR102121817B1 (en) * 2018-09-12 2020-06-26 한국화학연구원 Vectors for expressing recombinant antigens using CRISPR and method for simultaneously multiplexing thereof
US10711267B2 (en) 2018-10-01 2020-07-14 North Carolina State University Recombinant type I CRISPR-Cas system
WO2020081438A1 (en) 2018-10-16 2020-04-23 Blueallele, Llc Methods for targeted insertion of dna in genes
WO2020086475A1 (en) 2018-10-22 2020-04-30 Inscripta, Inc. Engineered enzymes
US11214781B2 (en) 2018-10-22 2022-01-04 Inscripta, Inc. Engineered enzyme
US20220380740A1 (en) * 2018-10-24 2022-12-01 The Broad Institute, Inc. Constructs for improved hdr-dependent genomic editing
CA3125299A1 (en) * 2019-01-04 2020-07-09 The University Of Chicago Systems and methods for modulating rna
US20220136004A1 (en) * 2019-01-11 2022-05-05 Chan Zuckerberg Biohub, Inc. Targeted In Vivo Genome Modification
EP3924477A4 (en) * 2019-02-14 2023-03-29 Metagenomi, Inc. Enzymes with ruvc domains
CA3130789A1 (en) 2019-03-07 2020-09-10 The Regents Of The University Of California Crispr-cas effector polypeptides and methods of use thereof
US11001831B2 (en) 2019-03-25 2021-05-11 Inscripta, Inc. Simultaneous multiplex genome editing in yeast
US10815467B2 (en) 2019-03-25 2020-10-27 Inscripta, Inc. Simultaneous multiplex genome editing in yeast
WO2020247587A1 (en) 2019-06-06 2020-12-10 Inscripta, Inc. Curing for recursive nucleic acid-guided cell editing
US10907125B2 (en) 2019-06-20 2021-02-02 Inscripta, Inc. Flow through electroporation modules and instrumentation
AU2020297499A1 (en) 2019-06-21 2022-02-03 Inscripta, Inc. Genome-wide rationally-designed mutations leading to enhanced lysine production in E. coli
US10927385B2 (en) 2019-06-25 2021-02-23 Inscripta, Inc. Increased nucleic-acid guided cell editing in yeast
CN110628825A (en) * 2019-10-14 2019-12-31 上海捷易生物科技有限公司 NHEJ-dependent reporter gene knock-in composition and using method thereof
WO2021102059A1 (en) 2019-11-19 2021-05-27 Inscripta, Inc. Methods for increasing observed editing in bacteria
CA3157131A1 (en) 2019-12-10 2021-06-17 Inscripta, Inc. Novel mad nucleases
US10704033B1 (en) 2019-12-13 2020-07-07 Inscripta, Inc. Nucleic acid-guided nucleases
US20230042273A1 (en) 2019-12-16 2023-02-09 BASF Agricultural Solutions Seed US LLC Improved genome editing using paired nickases
AU2020407048A1 (en) 2019-12-18 2022-06-09 Inscripta, Inc. Cascade/dCas3 complementation assays for in vivo detection of nucleic acid-guided nuclease edited cells
CN115243711A (en) * 2020-01-09 2022-10-25 先锋国际良种公司 Two-step gene exchange
US10689669B1 (en) 2020-01-11 2020-06-23 Inscripta, Inc. Automated multi-module cell processing methods, instruments, and systems
KR20220133257A (en) 2020-01-27 2022-10-04 인스크립타 인코포레이티드 Electroporation modules and instruments
EP4127156A4 (en) 2020-03-31 2024-03-27 Metagenomi Inc Class ii, type ii crispr systems
US20210332388A1 (en) 2020-04-24 2021-10-28 Inscripta, Inc. Compositions, methods, modules and instruments for automated nucleic acid-guided nuclease editing in mammalian cells
US11787841B2 (en) 2020-05-19 2023-10-17 Inscripta, Inc. Rationally-designed mutations to the thrA gene for enhanced lysine production in E. coli
CN111849986A (en) * 2020-07-24 2020-10-30 江苏集萃药康生物科技有限公司 Method for reducing tandem connection of double-stranded DNA (deoxyribonucleic acid) fragments in CRISPR-Cas9 gene editing and application thereof
EP4214314A1 (en) 2020-09-15 2023-07-26 Inscripta, Inc. Crispr editing to embed nucleic acid landing pads into genomes of live cells
US11512297B2 (en) 2020-11-09 2022-11-29 Inscripta, Inc. Affinity tag for recombination protein recruitment
WO2022133139A2 (en) * 2020-12-16 2022-06-23 Administrators Of The Tulane Educational Fund Wnt+ adipocytes, exosomes from wnt+ adipocyte, and methods of making and using them
EP4271802A1 (en) 2021-01-04 2023-11-08 Inscripta, Inc. Mad nucleases
EP4274890A1 (en) 2021-01-07 2023-11-15 Inscripta, Inc. Mad nucleases
US11884924B2 (en) 2021-02-16 2024-01-30 Inscripta, Inc. Dual strand nucleic acid-guided nickase editing
WO2023077148A1 (en) 2021-11-01 2023-05-04 Tome Biosciences, Inc. Single construct platform for simultaneous delivery of gene editing machinery and nucleic acid cargo
WO2023122764A1 (en) 2021-12-22 2023-06-29 Tome Biosciences, Inc. Co-delivery of a gene editor construct and a donor template
WO2023205744A1 (en) 2022-04-20 2023-10-26 Tome Biosciences, Inc. Programmable gene insertion compositions
WO2023215831A1 (en) 2022-05-04 2023-11-09 Tome Biosciences, Inc. Guide rna compositions for programmable gene insertion
WO2023225670A2 (en) 2022-05-20 2023-11-23 Tome Biosciences, Inc. Ex vivo programmable gene insertion
WO2023250384A2 (en) * 2022-06-22 2023-12-28 The Regents Of The University Of California Crispr-cas effector polypeptides and methods of use thereof
WO2024020587A2 (en) 2022-07-22 2024-01-25 Tome Biosciences, Inc. Pleiopluripotent stem cell programmable gene insertion

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100055728A1 (en) * 2006-08-04 2010-03-04 Georgia State University Research Foundation, Inc. Enzyme sensors, methods for preparing and using such sensors, and methods of detecting protease activity
US20110217739A1 (en) * 2008-11-06 2011-09-08 University Of Georgia Research Foundation, Inc. Cas6 polypeptides and methods of use
WO2012012738A1 (en) * 2010-07-23 2012-01-26 Sigma-Aldrich Co., Llc Genome editing using targeting endonucleases and single-stranded nucleic acids
US20120192298A1 (en) * 2009-07-24 2012-07-26 Sigma Aldrich Co. Llc Method for genome editing
WO2013142578A1 (en) * 2012-03-20 2013-09-26 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX

Family Cites Families (163)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4952496A (en) 1984-03-30 1990-08-28 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
WO1988008450A1 (en) 1987-05-01 1988-11-03 Birdwell Finlayson Gene therapy for metabolite disorders
US5350689A (en) 1987-05-20 1994-09-27 Ciba-Geigy Corporation Zea mays plants and transgenic Zea mays plants regenerated from protoplasts or protoplast-derived cells
US5767367A (en) 1990-06-23 1998-06-16 Hoechst Aktiengesellschaft Zea mays (L.) with capability of long term, highly efficient plant regeneration including fertile transgenic maize plants having a heterologous gene, and their preparation
US7150982B2 (en) 1991-09-09 2006-12-19 Third Wave Technologies, Inc. RNA detection assays
FR2763797B1 (en) * 1997-05-30 1999-07-16 Tabacs & Allumettes Ind VERY LOW TAR RATE CIGARETTE WITH A TOBACCO TASTE COMPARABLE TO THAT OF A CLASSIC CIGARETTE WITH HIGHER TAR RATE
US20040186071A1 (en) 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
US20020182673A1 (en) 1998-05-15 2002-12-05 Genentech, Inc. IL-17 homologous polypedies and therapeutic uses thereof
WO2000046386A2 (en) 1999-02-03 2000-08-10 The Children's Medical Center Corporation Gene repair involving the induction of double-stranded dna cleavage at a chromosomal target site
US8183339B1 (en) * 1999-10-12 2012-05-22 Xigen S.A. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
WO2002026967A2 (en) 2000-09-25 2002-04-04 Thomas Jefferson University Targeted gene correction by single-stranded oligodeoxynucleotides
CA2881568C (en) 2000-10-27 2019-09-24 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups a & b
JP3454818B1 (en) * 2001-03-16 2003-10-06 直哉 小林 Liver cell proliferation method, liver cell obtained by the method, and use thereof
CA2453183C (en) 2001-07-12 2016-05-10 University Of Massachusetts In vivo production of small interfering rnas that mediate gene silencing
US20060253913A1 (en) 2001-12-21 2006-11-09 Yue-Jin Huang Production of hSA-linked butyrylcholinesterases in transgenic mammals
US8106255B2 (en) * 2002-01-23 2012-01-31 Dana Carroll Targeted chromosomal mutagenasis using zinc finger nucleases
EP2368982A3 (en) 2002-03-21 2011-10-12 Sangamo BioSciences, Inc. Methods and compositions for using zinc finger endonucleases to enhance homologous recombination
AU2003224897A1 (en) 2002-04-09 2003-10-27 Kenneth L. Beattie Oligonucleotide probes for genosensor chips
AU2003233719A1 (en) * 2002-06-06 2003-12-22 Her Majesty The Queen In Right Of Canada As Represented By The Minister Of Agriculture And Agri-Food Modifying the dna recombination potential in eukaryotes
CA2497913C (en) 2002-09-05 2014-06-03 California Institute Of Technology Use of chimeric nucleases to stimulate gene targeting
DE10260805A1 (en) * 2002-12-23 2004-07-22 Geneart Gmbh Method and device for optimizing a nucleotide sequence for expression of a protein
EP1651660B1 (en) * 2003-08-08 2018-01-24 Sangamo Therapeutics, Inc. Methods and compositions for targeted cleavage and recombination
WO2005070948A1 (en) 2004-01-23 2005-08-04 Intronn, Inc. Correction of alpha-1-antitrypsin genetic defects using spliceosome mediated rna trans splicing
US7972854B2 (en) 2004-02-05 2011-07-05 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
US20050220796A1 (en) 2004-03-31 2005-10-06 Dynan William S Compositions and methods for modulating DNA repair
US7919277B2 (en) 2004-04-28 2011-04-05 Danisco A/S Detection and typing of bacterial strains
CA2592099A1 (en) * 2004-12-22 2006-06-29 Nucleonics, Inc. Conserved hbv and hcv sequences useful for gene silencing
US7892224B2 (en) 2005-06-01 2011-02-22 Brainlab Ag Inverse catheter planning
US7534819B2 (en) 2005-06-10 2009-05-19 University Of Washington Compositions and methods for intracellular delivery of biotinylated cargo
US20060282289A1 (en) 2005-06-14 2006-12-14 Healthmatch Solutions, Llc System and method for health care financing
WO2007014181A2 (en) * 2005-07-25 2007-02-01 Johns Hopkins University Site-specific modification of the human genome using custom-designed zinc finger nucleases
KR20080033455A (en) * 2005-07-26 2008-04-16 상가모 바이오사이언스 인코포레이티드 Targeted integration and expression of exogenous nucleic acid sequences
US10022457B2 (en) 2005-08-05 2018-07-17 Gholam A. Peyman Methods to regulate polarization and enhance function of cells
NZ597299A (en) 2005-08-26 2013-03-28 Dupont Nutrition Biosci Aps Use of a cas gene in combination with CRISPR repeats for modulating resistance in a cell
KR100877824B1 (en) * 2005-11-11 2009-01-12 한국생명공학연구원 E2EPF ubiquitin carrier protein-von Hippel-Lindau interaction and uses of thereof
EP1991706B1 (en) * 2006-03-02 2014-01-15 The Ohio State University Research Foundation Microrna expression profile associated with pancreatic cancer
US9677123B2 (en) 2006-03-15 2017-06-13 Siemens Healthcare Diagnostics Inc. Degenerate nucleobase analogs
JP2009531444A (en) * 2006-03-28 2009-09-03 ノバルティス アーゲー Covalently linked complex of HIVTAT protein and Env protein
DK2016183T3 (en) 2006-05-10 2011-09-19 Deinove Sa Method for chromosomal modification using a novel DNA repair system
ES2373586T3 (en) 2006-05-19 2012-02-06 Danisco A/S MARKED MICROORGANISMS AND METHODS TO MARK.
EP2019839B1 (en) 2006-05-25 2011-12-07 Sangamo BioSciences, Inc. Methods and compositions for gene inactivation
DK2034848T3 (en) 2006-06-16 2017-02-06 Dupont Nutrition Biosci Aps STREPTOCOCCUS THERMOPHILUS BACTERIA
CN104531672B (en) 2007-03-02 2020-01-10 杜邦营养生物科学有限公司 Cultures with improved phage resistance
GB0806086D0 (en) 2008-04-04 2008-05-14 Ulive Entpr Ltd Dendrimer polymer hybrids
US8546553B2 (en) 2008-07-25 2013-10-01 University Of Georgia Research Foundation, Inc. Prokaryotic RNAi-like system and methods of use
KR20160015400A (en) 2008-08-22 2016-02-12 상가모 바이오사이언스 인코포레이티드 Methods and compositions for targeted single-stranded cleavage and targeted integration
WO2010030963A2 (en) 2008-09-15 2010-03-18 Children's Medical Center Corporation Modulation of bcl11a for treatment of hemoglobinopathies
US20100076057A1 (en) * 2008-09-23 2010-03-25 Northwestern University TARGET DNA INTERFERENCE WITH crRNA
WO2010054154A2 (en) 2008-11-07 2010-05-14 Danisco A/S Bifidobacteria crispr sequences
DK2367938T3 (en) 2008-12-12 2014-08-25 Dupont Nutrition Biosci Aps GENETIC CLUSTER OF TREASURES OF STREPTOCOCCUS THERMOPHILUS WITH UNIQUE RHEOLOGICAL PROPERTIES FOR MILK Fermentation
WO2010075424A2 (en) 2008-12-22 2010-07-01 The Regents Of University Of California Compositions and methods for downregulating prokaryotic genes
GB0823658D0 (en) 2008-12-30 2009-02-04 Angiomed Ag Stent delivery device
US8392349B2 (en) 2009-02-23 2013-03-05 Shalini Vajjhala Global adaptation atlas and method of creating same
AU2010235161B2 (en) 2009-04-09 2015-01-22 Sangamo Therapeutics, Inc. Targeted integration into stem cells
WO2010125471A2 (en) 2009-04-30 2010-11-04 Fondazione Centro San Raffaele Del Monte Tabor Gene vector
EP2456877A1 (en) 2009-07-24 2012-05-30 Sigma-Aldrich Co. LLC Method for genome editing
EP2461819A4 (en) 2009-07-28 2013-07-31 Sangamo Biosciences Inc Methods and compositions for treating trinucleotide repeat disorders
KR101418355B1 (en) 2009-10-23 2014-07-11 (주)바이오니아 High throughput oligonucleotide synthesizer
DE102009052674B4 (en) 2009-11-12 2012-10-18 Karl Weinhold Method and device for connecting double-walled pipes
US20110294114A1 (en) 2009-12-04 2011-12-01 Cincinnati Children's Hospital Medical Center Optimization of determinants for successful genetic correction of diseases, mediated by hematopoietic stem cells
BR112012014080A2 (en) 2009-12-10 2015-10-27 Univ Iowa State Res Found method for modifying genetic material, method for generating a nucleic acid, effector endonuclease monomer such, method for generating an aninal, method for generating a plant, method for directed genetic recombination, nucleic acid, expression cassette, and host cell
US9255259B2 (en) 2010-02-09 2016-02-09 Sangamo Biosciences, Inc. Targeted genomic modification with partially single-stranded donor molecules
US10087431B2 (en) 2010-03-10 2018-10-02 The Regents Of The University Of California Methods of generating nucleic acid fragments
CN103038338B (en) 2010-05-10 2017-03-08 加利福尼亚大学董事会 Endoribonuclease compositionss and its using method
WO2011146121A1 (en) * 2010-05-17 2011-11-24 Sangamo Biosciences, Inc. Novel dna-binding proteins and uses thereof
EP2392208B1 (en) * 2010-06-07 2016-05-04 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Fusion proteins comprising a DNA-binding domain of a Tal effector protein and a non-specific cleavage domain of a restriction nuclease and their use
WO2011156430A2 (en) 2010-06-07 2011-12-15 Fred Hutchinson Cancer Research Center Generation and expression of engineered i-onui endonuclease and its homologues and uses thereof
WO2011159369A1 (en) 2010-06-14 2011-12-22 Iowa State University Research Foundation, Inc. Nuclease activity of tal effector and foki fusion protein
EP2601611B1 (en) 2010-08-02 2020-12-09 Integrated Dna Technologies, Inc. Methods for predicting stability and melting temperatures of nucleic acid duplexes
WO2012054726A1 (en) 2010-10-20 2012-04-26 Danisco A/S Lactococcus crispr-cas sequences
US9403880B2 (en) * 2010-11-26 2016-08-02 Institut Pasteur Identification of a human gyrovirus and applications
US20120214241A1 (en) 2010-12-22 2012-08-23 Josee Laganiere Zinc finger nuclease modification of leucine rich repeat kinase 2 (lrrk2) mutant fibroblasts and ipscs
KR20120096395A (en) 2011-02-22 2012-08-30 주식회사 툴젠 Methods for enriching cells containing nuclease-induced gene disruptions
WO2012164565A1 (en) 2011-06-01 2012-12-06 Yeda Research And Development Co. Ltd. Compositions and methods for downregulating prokaryotic genes
US9150847B2 (en) 2011-09-21 2015-10-06 Sangamo Biosciences, Inc. Methods and compositions for regulation of transgene expression
CA2854819C (en) * 2011-11-16 2022-07-19 Sangamo Biosciences, Inc. Modified dna-binding proteins and uses thereof
US8450107B1 (en) 2011-11-30 2013-05-28 The Broad Institute Inc. Nucleotide-specific recognition sequences for designer TAL effectors
GB201122458D0 (en) 2011-12-30 2012-02-08 Univ Wageningen Modified cascade ribonucleoproteins and uses thereof
KR101833589B1 (en) 2012-02-24 2018-03-02 프레드 헛친슨 켄서 리서치 센터 Compositions and methods for the treatment of hemoglobinopathies
MX359327B (en) 2012-02-29 2018-09-25 Sangamo Biosciences Inc Methods and compositions for treating huntington's disease.
WO2013141680A1 (en) 2012-03-20 2013-09-26 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX
AU2013204327B2 (en) 2012-04-20 2016-09-01 Aviagen Cell transfection method
WO2013160230A1 (en) 2012-04-23 2013-10-31 Bayer Cropscience Nv Targeted genome engineering in plants
US9834786B2 (en) 2012-04-25 2017-12-05 Regeneron Pharmaceuticals, Inc. Nuclease-mediated targeting with large targeting vectors
KR102091298B1 (en) 2012-05-02 2020-03-19 다우 아그로사이언시즈 엘엘씨 Targeted modification of malate dehydrogenase
EP2847338B1 (en) 2012-05-07 2018-09-19 Sangamo Therapeutics, Inc. Methods and compositions for nuclease-mediated targeted integration of transgenes
US11120889B2 (en) 2012-05-09 2021-09-14 Georgia Tech Research Corporation Method for synthesizing a nuclease with reduced off-site cleavage
PE20190844A1 (en) * 2012-05-25 2019-06-17 Emmanuelle Charpentier MODULATION OF TRANSCRIPTION WITH ADDRESSING RNA TO GENERIC DNA
MX2014014650A (en) 2012-05-30 2015-10-14 Univ Washington Supercoiled minivectors as a tool for dna repair, alteration and replacement.
US9102936B2 (en) 2012-06-11 2015-08-11 Agilent Technologies, Inc. Method of adaptor-dimer subtraction using a CRISPR CAS6 protein
EP2858486A4 (en) 2012-06-12 2016-04-13 Hoffmann La Roche Methods and compositions for generating conditional knock-out alleles
EP2674501A1 (en) 2012-06-14 2013-12-18 Agence nationale de sécurité sanitaire de l'alimentation,de l'environnement et du travail Method for detecting and identifying enterohemorrhagic Escherichia coli
US9688971B2 (en) 2012-06-15 2017-06-27 The Regents Of The University Of California Endoribonuclease and methods of use thereof
WO2013192278A1 (en) 2012-06-19 2013-12-27 Regents Of The University Of Minnesota Gene targeting in plants using dna viruses
EP2872154B1 (en) 2012-07-11 2017-05-31 Sangamo BioSciences, Inc. Methods and compositions for delivery of biologics
HUE051612T2 (en) 2012-07-11 2021-03-01 Sangamo Therapeutics Inc Methods and compositions for the treatment of lysosomal storage diseases
KR20230065381A (en) 2012-07-25 2023-05-11 더 브로드 인스티튜트, 인코퍼레이티드 Inducible dna binding proteins and genome perturbation tools and applications thereof
WO2014022702A2 (en) 2012-08-03 2014-02-06 The Regents Of The University Of California Methods and compositions for controlling gene expression by rna processing
IN2015DN01480A (en) 2012-08-29 2015-07-03 Sangamo Biosciences Inc
UA118090C2 (en) 2012-09-07 2018-11-26 ДАУ АГРОСАЙЄНСІЗ ЕлЕлСі Fad2 performance loci and corresponding target site specific binding proteins capable of inducing targeted breaks
UA119135C2 (en) 2012-09-07 2019-05-10 ДАУ АГРОСАЙЄНСІЗ ЕлЕлСі Engineered transgene integration platform (etip) for gene targeting and trait stacking
CN105264067B (en) 2012-09-07 2020-11-10 美国陶氏益农公司 FAD3 performance loci and corresponding target site specific binding proteins capable of inducing targeted breaks
WO2014059255A1 (en) 2012-10-12 2014-04-17 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (lsd1) fusion proteins
KR101706085B1 (en) * 2012-10-23 2017-02-14 주식회사 툴젠 Composition for cleaving a target DNA comprising a guide RNA specific for the target DNA and Cas protein-encoding nucleic acid or Cas protein, and use thereof
BR112015009589A2 (en) 2012-10-30 2017-11-14 Recombinetics Inc Production processes of a non-human production animal, non-human genetically modified production animal, non-human in vitro organism chosen from the group consisting of a cell or a blastocyst animal, the in vitro organism, production process of an animal cell non-human genetically modified production animal, non-human production animal and method of production of a non-human genetically modified production animal
CA2890160A1 (en) 2012-10-31 2014-05-08 Cellectis Coupling herbicide resistance with targeted insertion of transgenes in plants
US20140127752A1 (en) 2012-11-07 2014-05-08 Zhaohui Zhou Method, composition, and reagent kit for targeted genomic enrichment
KR102145760B1 (en) 2012-12-06 2020-08-19 시그마-알드리치 컴퍼니., 엘엘씨 Crispr-based genome modification and regulation
WO2014093479A1 (en) 2012-12-11 2014-06-19 Montana State University Crispr (clustered regularly interspaced short palindromic repeats) rna-guided control of gene regulation
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
ES2701749T3 (en) 2012-12-12 2019-02-25 Broad Inst Inc Methods, models, systems and apparatus to identify target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and transmit results thereof
EP3045537A1 (en) 2012-12-12 2016-07-20 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
EP2931899A1 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
WO2014093635A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
EP4299741A3 (en) 2012-12-12 2024-02-28 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
EP2931898B1 (en) 2012-12-12 2016-03-09 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
WO2014093694A1 (en) 2012-12-12 2014-06-19 The Broad Institute, Inc. Crispr-cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
CN113528577A (en) 2012-12-12 2021-10-22 布罗德研究所有限公司 Engineering of systems, methods and optimized guide compositions for sequence manipulation
CN114634950A (en) 2012-12-12 2022-06-17 布罗德研究所有限公司 CRISPR-CAS component systems, methods, and compositions for sequence manipulation
JP6473419B2 (en) 2012-12-13 2019-02-20 ダウ アグロサイエンシィズ エルエルシー DNA detection method for site-specific nuclease activity
KR20150095861A (en) * 2012-12-17 2015-08-21 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 Rna-guided human genome engineering
DK2938184T3 (en) 2012-12-27 2018-12-17 Keygene Nv Method of removing a genetic linkage in a plant
CA2898184A1 (en) 2013-01-16 2014-07-24 Emory University Cas9-nucleic acid complexes and uses related thereto
CN103233028B (en) 2013-01-25 2015-05-13 南京徇齐生物技术有限公司 Specie limitation-free eucaryote gene targeting method having no bio-safety influence and helical-structure DNA sequence
WO2014127287A1 (en) 2013-02-14 2014-08-21 Massachusetts Institute Of Technology Method for in vivo tergated mutagenesis
CA2900992C (en) 2013-02-20 2023-02-28 Regeneron Pharmaceuticals, Inc. Genetic modification of rats
JP6491113B2 (en) 2013-02-25 2019-03-27 サンガモ セラピューティクス, インコーポレイテッド Methods and compositions for enhancing nuclease-mediated gene disruption
DK3620534T3 (en) 2013-03-14 2021-12-06 Caribou Biosciences Inc CRISPR-CAS NUCLEIC ACID COMPOSITIONS-TARGETING NUCLEIC ACIDS
US9234213B2 (en) 2013-03-15 2016-01-12 System Biosciences, Llc Compositions and methods directed to CRISPR/Cas genomic engineering systems
EP3741868A1 (en) 2013-03-15 2020-11-25 The General Hospital Corporation Rna-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US20140273230A1 (en) 2013-03-15 2014-09-18 Sigma-Aldrich Co., Llc Crispr-based genome modification and regulation
US10760064B2 (en) 2013-03-15 2020-09-01 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US11332719B2 (en) 2013-03-15 2022-05-17 The Broad Institute, Inc. Recombinant virus and preparations thereof
US20140364333A1 (en) 2013-03-15 2014-12-11 President And Fellows Of Harvard College Methods for Live Imaging of Cells
EP2970997A1 (en) 2013-03-15 2016-01-20 Regents of the University of Minnesota Engineering plant genomes using crispr/cas systems
JP2016522679A (en) 2013-04-04 2016-08-04 プレジデント アンド フェローズ オブ ハーバード カレッジ Therapeutic use of genome editing with the CRISPR / Cas system
RU2723130C2 (en) 2013-04-05 2020-06-08 ДАУ АГРОСАЙЕНСИЗ ЭлЭлСи Methods and compositions for embedding an exogenous sequence into a plant genome
SG11201508028QA (en) 2013-04-16 2015-10-29 Regeneron Pharma Targeted modification of rat genome
CN103224947B (en) 2013-04-28 2015-06-10 陕西师范大学 Gene targeting system
US10604771B2 (en) 2013-05-10 2020-03-31 Sangamo Therapeutics, Inc. Delivery methods and compositions for nuclease-mediated genome engineering
EP3778899A1 (en) 2013-05-22 2021-02-17 Northwestern University Rna-directed dna cleavage and gene editing by cas9 enzyme from neisseria meningitidis
US9873907B2 (en) 2013-05-29 2018-01-23 Agilent Technologies, Inc. Method for fragmenting genomic DNA using CAS9
US9267135B2 (en) 2013-06-04 2016-02-23 President And Fellows Of Harvard College RNA-guided transcriptional regulation
WO2014204727A1 (en) 2013-06-17 2014-12-24 The Broad Institute Inc. Functional genomics using crispr-cas systems, compositions methods, screens and applications thereof
EP4245853A3 (en) 2013-06-17 2023-10-18 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
BR112015031611A2 (en) 2013-06-17 2017-12-12 Massachusetts Inst Technology application, manipulation and optimization of systems, methods and compositions for targeting and modeling post-mitotic cell diseases and disorders
MX2015017313A (en) 2013-06-17 2016-11-25 Broad Inst Inc Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components.
EP3011031B1 (en) 2013-06-17 2020-09-30 The Broad Institute Inc. Delivery and use of the crispr-cas systems, vectors and compositions for hepatic targeting and therapy
CN103343120B (en) 2013-07-04 2015-03-04 中国科学院遗传与发育生物学研究所 Wheat genome site-specific modification method
CN103382468B (en) * 2013-07-04 2015-04-29 中国科学院遗传与发育生物学研究所 Site-directed modification method of rice genome
CA2917639C (en) 2013-07-10 2024-01-02 President And Fellows Of Harvard College Orthogonal cas9 proteins for rna-guided gene regulation and editing
CN103388006B (en) 2013-07-26 2015-10-28 华东师范大学 A kind of construction process of site-directed point mutation
US10421957B2 (en) 2013-07-29 2019-09-24 Agilent Technologies, Inc. DNA assembly using an RNA-programmable nickase
AU2014341934B2 (en) 2013-11-04 2017-12-07 Corteva Agriscience Llc Optimal soybean loci
RU2709734C2 (en) 2013-11-04 2019-12-19 ДАУ АГРОСАЙЕНСИЗ ЭлЭлСи Optimum maize loci
UY35814A (en) 2013-11-04 2015-05-29 Dow Agrosciences Llc ? OPTIMAL PLACES FOR SOYBEAN ?.
LT3066201T (en) 2013-11-07 2018-08-10 Editas Medicine, Inc. Crispr-related methods and compositions with governing grnas
JP6174811B2 (en) 2013-12-11 2017-08-02 リジェネロン・ファーマシューティカルズ・インコーポレイテッドRegeneron Pharmaceuticals, Inc. Methods and compositions for targeted genomic modification
WO2015116686A1 (en) 2014-01-29 2015-08-06 Agilent Technologies, Inc. Cas9-based isothermal method of detection of specific dna sequence
US20150291969A1 (en) 2014-01-30 2015-10-15 Chromatin, Inc. Compositions for reduced lignin content in sorghum and improving cell wall digestibility, and methods of making the same
US20150225801A1 (en) 2014-02-11 2015-08-13 California Institute Of Technology Recording and mapping lineage information and molecular events in individual cells
WO2015127439A1 (en) 2014-02-24 2015-08-27 Sangamo Biosciences, Inc. Methods and compositions for nuclease-mediated targeted integration
JP6594891B2 (en) 2014-03-18 2019-10-23 サンガモ セラピューティクス, インコーポレイテッド Methods and compositions for modulating zinc finger protein expression

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100055728A1 (en) * 2006-08-04 2010-03-04 Georgia State University Research Foundation, Inc. Enzyme sensors, methods for preparing and using such sensors, and methods of detecting protease activity
US20110217739A1 (en) * 2008-11-06 2011-09-08 University Of Georgia Research Foundation, Inc. Cas6 polypeptides and methods of use
US20120192298A1 (en) * 2009-07-24 2012-07-26 Sigma Aldrich Co. Llc Method for genome editing
WO2012012738A1 (en) * 2010-07-23 2012-01-26 Sigma-Aldrich Co., Llc Genome editing using targeting endonucleases and single-stranded nucleic acids
WO2013142578A1 (en) * 2012-03-20 2013-09-26 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
GASIUNAS ET AL.: "Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria.", PNAS, vol. 109, no. 39, 4 September 2012 (2012-09-04), pages E2579 - E2586, XP055068588 *
JINEK ET AL.: "A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity", SCIENCE, vol. 337, 17 August 2012 (2012-08-17), pages 816 - 820, XP055229606 *
JINEK ET AL.: "RNA-programmed genome editing in human cells", ELIFE, vol. 2, no. E00471, 29 January 2013 (2013-01-29), pages 1 - 9, XP055245475 *

Cited By (623)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10202608B2 (en) 2006-08-31 2019-02-12 Gen9, Inc. Iterative nucleic acid assembly using activation of vector-encoded traits
US10457935B2 (en) 2010-11-12 2019-10-29 Gen9, Inc. Protein arrays and methods of using and making the same
US11084014B2 (en) 2010-11-12 2021-08-10 Gen9, Inc. Methods and devices for nucleic acids synthesis
US10982208B2 (en) 2010-11-12 2021-04-20 Gen9, Inc. Protein arrays and methods of using and making the same
US11845054B2 (en) 2010-11-12 2023-12-19 Gen9, Inc. Methods and devices for nucleic acids synthesis
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US9322006B2 (en) 2011-07-22 2016-04-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
US11702662B2 (en) 2011-08-26 2023-07-18 Gen9, Inc. Compositions and methods for high fidelity assembly of nucleic acids
US11293052B2 (en) 2011-12-22 2022-04-05 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11549136B2 (en) 2011-12-22 2023-01-10 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11639518B2 (en) 2011-12-22 2023-05-02 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11111521B2 (en) 2011-12-22 2021-09-07 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11566277B2 (en) 2011-12-22 2023-01-31 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11566276B2 (en) 2011-12-22 2023-01-31 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11293051B2 (en) 2011-12-22 2022-04-05 President And Fellows Of Harvard College Compositions and methods for analyte detection
US11939604B2 (en) 2011-12-30 2024-03-26 Caribou Biosciences, Inc. Modified cascade ribonucleoproteins and uses thereof
US10435678B2 (en) 2011-12-30 2019-10-08 Caribou Biosciences, Inc. Modified cascade ribonucleoproteins and uses thereof
EP3091072A1 (en) * 2011-12-30 2016-11-09 Caribou Biosciences, Inc. Modified cascade ribonucleoproteins and uses thereof
US10954498B2 (en) 2011-12-30 2021-03-23 Caribou Biosciences, Inc. Modified cascade ribonucleoproteins and uses thereof
US9885026B2 (en) 2011-12-30 2018-02-06 Caribou Biosciences, Inc. Modified cascade ribonucleoproteins and uses thereof
GB2512246B (en) * 2011-12-30 2016-07-20 Caribou Biosciences Inc Modified cascade ribonucleoproteins and uses thereof
GB2534074A (en) * 2011-12-30 2016-07-13 Caribou Biosciences Inc Modified cascade ribonucleoproteins and uses thereof
US10711257B2 (en) 2011-12-30 2020-07-14 Caribou Biosciences, Inc. Modified cascade ribonucleoproteins and uses thereof
US11555187B2 (en) 2012-03-20 2023-01-17 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
US10844378B2 (en) 2012-03-20 2020-11-24 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
EP2828386A1 (en) 2012-03-20 2015-01-28 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX
EP2828386B1 (en) * 2012-03-20 2019-07-10 Vilnius University RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX
EP3594341A1 (en) * 2012-03-20 2020-01-15 Vilnius University Rna-directed dna cleavage by the cas9-crrna complex
US9637739B2 (en) 2012-03-20 2017-05-02 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
US10308931B2 (en) 2012-03-21 2019-06-04 Gen9, Inc. Methods for screening proteins using DNA encoded chemical libraries as templates for enzyme catalysis
US10081807B2 (en) 2012-04-24 2018-09-25 Gen9, Inc. Methods for sorting nucleic acids and multiplexed preparative in vitro cloning
US10927369B2 (en) 2012-04-24 2021-02-23 Gen9, Inc. Methods for sorting nucleic acids and multiplexed preparative in vitro cloning
US10301646B2 (en) 2012-04-25 2019-05-28 Regeneron Pharmaceuticals, Inc. Nuclease-mediated targeting with large targeting vectors
US9834786B2 (en) 2012-04-25 2017-12-05 Regeneron Pharmaceuticals, Inc. Nuclease-mediated targeting with large targeting vectors
US11898176B2 (en) 2012-04-27 2024-02-13 Duke University Genetic correction of mutated genes
US11814645B2 (en) 2012-05-25 2023-11-14 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11401532B2 (en) 2012-05-25 2022-08-02 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10415061B2 (en) 2012-05-25 2019-09-17 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10570419B2 (en) 2012-05-25 2020-02-25 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10407697B2 (en) 2012-05-25 2019-09-10 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11293034B2 (en) 2012-05-25 2022-04-05 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP3241902A1 (en) * 2012-05-25 2017-11-08 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US10400253B2 (en) 2012-05-25 2019-09-03 The Regents Of The University Of California Methods and compositions or RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10385360B2 (en) 2012-05-25 2019-08-20 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10563227B2 (en) 2012-05-25 2020-02-18 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10752920B2 (en) 2012-05-25 2020-08-25 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10358658B2 (en) 2012-05-25 2019-07-23 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10358659B2 (en) 2012-05-25 2019-07-23 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10351878B2 (en) 2012-05-25 2019-07-16 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10421980B2 (en) 2012-05-25 2019-09-24 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10774344B1 (en) 2012-05-25 2020-09-15 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10337029B2 (en) 2012-05-25 2019-07-02 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11274318B2 (en) 2012-05-25 2022-03-15 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10428352B2 (en) 2012-05-25 2019-10-01 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10793878B1 (en) 2012-05-25 2020-10-06 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10577631B2 (en) 2012-05-25 2020-03-03 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11674159B2 (en) 2012-05-25 2023-06-13 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11242543B2 (en) 2012-05-25 2022-02-08 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP2800811A1 (en) 2012-05-25 2014-11-12 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
EP3241902B1 (en) 2012-05-25 2018-02-28 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US11634730B2 (en) 2012-05-25 2023-04-25 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11186849B2 (en) 2012-05-25 2021-11-30 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10443076B2 (en) 2012-05-25 2019-10-15 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10308961B2 (en) 2012-05-25 2019-06-04 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10301651B2 (en) 2012-05-25 2019-05-28 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10676759B2 (en) 2012-05-25 2020-06-09 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10550407B2 (en) 2012-05-25 2020-02-04 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11332761B2 (en) 2012-05-25 2022-05-17 The Regenis of Wie University of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10266850B2 (en) 2012-05-25 2019-04-23 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP2800811A4 (en) * 2012-05-25 2015-09-23 Univ California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US10900054B2 (en) 2012-05-25 2021-01-26 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10487341B2 (en) 2012-05-25 2019-11-26 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10000772B2 (en) 2012-05-25 2018-06-19 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP3401400B1 (en) 2012-05-25 2019-04-03 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US10669560B2 (en) 2012-05-25 2020-06-02 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10227611B2 (en) 2012-05-25 2019-03-12 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10597680B2 (en) 2012-05-25 2020-03-24 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10640791B2 (en) 2012-05-25 2020-05-05 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10982230B2 (en) 2012-05-25 2021-04-20 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10513712B2 (en) 2012-05-25 2019-12-24 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10982231B2 (en) 2012-05-25 2021-04-20 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP3597749A1 (en) * 2012-05-25 2020-01-22 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
GB2537000A (en) * 2012-05-25 2016-10-05 Univ California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10519467B2 (en) 2012-05-25 2019-12-31 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10526619B2 (en) 2012-05-25 2020-01-07 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10626419B2 (en) 2012-05-25 2020-04-21 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10988780B2 (en) 2012-05-25 2021-04-27 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10533190B2 (en) 2012-05-25 2020-01-14 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11549127B2 (en) 2012-05-25 2023-01-10 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10988782B2 (en) 2012-05-25 2021-04-27 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11001863B2 (en) 2012-05-25 2021-05-11 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11008590B2 (en) 2012-05-25 2021-05-18 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11479794B2 (en) 2012-05-25 2022-10-25 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP2800811B1 (en) 2012-05-25 2017-05-10 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US11028412B2 (en) 2012-05-25 2021-06-08 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11473108B2 (en) 2012-05-25 2022-10-18 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11008589B2 (en) 2012-05-25 2021-05-18 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
GB2537000B (en) * 2012-05-25 2017-03-08 Univ California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
EP3401400A1 (en) * 2012-05-25 2018-11-14 The Regents of The University of California Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US10113167B2 (en) 2012-05-25 2018-10-30 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US10612045B2 (en) 2012-05-25 2020-04-07 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification and for RNA-directed modulation of transcription
US11384360B2 (en) 2012-06-19 2022-07-12 Regents Of The University Of Minnesota Gene targeting in plants using DNA viruses
US11072789B2 (en) 2012-06-25 2021-07-27 Gen9, Inc. Methods for nucleic acid assembly and high throughput sequencing
EP3494997B1 (en) 2012-07-25 2019-09-18 The Broad Institute, Inc. Inducible dna binding proteins and genome perturbation tools and applications thereof
EP3494997A1 (en) * 2012-07-25 2019-06-12 The Broad Institute, Inc. Inducible dna binding proteins and genome perturbation tools and applications thereof
AU2018247306B2 (en) * 2012-07-25 2021-08-19 Massachusetts Institute Of Technology Inducible dna binding proteins and genome perturbation tools and applications thereof
EP3808844A1 (en) * 2012-07-25 2021-04-21 The Broad Institute, Inc. Inducible dna binding proteins and genome perturbation tools and applications thereof
US10851380B2 (en) 2012-10-23 2020-12-01 Toolgen Incorporated Methods for cleaving a target DNA using a guide RNA specific for the target DNA and Cas protein-encoding nucleic acid or Cas protein
JP2016500003A (en) * 2012-10-23 2016-01-07 ツールゲン インコーポレイテッド Composition for cleaving target DNA comprising guide RNA specific for target DNA and CAS protein-encoding nucleic acid or CAS protein, and use thereof
AU2017254854B2 (en) * 2012-10-23 2019-12-05 Toolgen Incorporated Composition For Cleaving A Target DNA Comprising A Guide RNA Specific For The Target DNA And Cas Protein-Encoding Nucleic Acid Or Cas Protein, And Use Thereof
JP2016027807A (en) * 2012-10-23 2016-02-25 ツールゲン インコーポレイテッド Composition for cleaving target dna comprising guide rna specific for target dna and cas protein-encoding nucleic acid or cas protein, and use thereof
EP3346003B1 (en) 2012-10-23 2021-06-09 Toolgen Incorporated Composition for cleaving a target dna comprising a guide rna specific for the target dna and cas protein-encoding nucleic acid or cas protein, and use thereof
EP3733847B1 (en) 2012-10-23 2022-06-01 Toolgen Incorporated Composition for cleaving a target dna comprising a guide rna specific for the target dna and cas protein-encoding nucleic acid or cas protein, and use thereof
EP3346003A1 (en) * 2012-10-23 2018-07-11 Toolgen Incorporated Composition for cleaving a target dna comprising a guide rna specific for the target dna and cas protein-encoding nucleic acid or cas protein, and use thereof
EP2912175A4 (en) * 2012-10-23 2015-12-09 Toolgen Inc Composition for cleaving a target dna comprising a guide rna specific for the target dna and cas protein-encoding nucleic acid or cas protein, and use thereof
US10844361B2 (en) 2012-11-16 2020-11-24 Poseida Therapeutics, Inc. Site-specific enzymes and methods of use
US10415024B2 (en) 2012-11-16 2019-09-17 Poseida Therapeutics, Inc. Site-specific enzymes and methods of use
EP3138910B1 (en) 2012-12-06 2017-09-20 Sigma-Aldrich Co. LLC Crispr-based genome modification and regulation
EP2928496B1 (en) 2012-12-06 2019-10-09 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
EP3138912B1 (en) 2012-12-06 2018-12-05 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
EP3360964B1 (en) 2012-12-06 2019-10-02 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
EP3138911B1 (en) 2012-12-06 2018-12-05 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
US10745716B2 (en) 2012-12-06 2020-08-18 Sigma-Aldrich Co. Llc CRISPR-based genome modification and regulation
EP3363902B1 (en) 2012-12-06 2019-11-27 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
US10731181B2 (en) 2012-12-06 2020-08-04 Sigma, Aldrich Co. LLC CRISPR-based genome modification and regulation
JP2019103511A (en) * 2012-12-12 2019-06-27 ザ・ブロード・インスティテュート・インコーポレイテッド Engineering of systems, methods and optimized guide compositions for sequence manipulation
US10930367B2 (en) 2012-12-12 2021-02-23 The Broad Institute, Inc. Methods, models, systems, and apparatus for identifying target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and conveying results thereof
EP3252160B1 (en) 2012-12-12 2020-10-28 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
JP2019103513A (en) * 2012-12-12 2019-06-27 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US8865406B2 (en) 2012-12-12 2014-10-21 The Broad Institute Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US9840713B2 (en) 2012-12-12 2017-12-12 The Broad Institute Inc. CRISPR-Cas component systems, methods and compositions for sequence manipulation
CN106170549B (en) * 2012-12-12 2020-02-21 布罗德研究所有限公司 CRISPR-CAS systems and methods for altering expression of gene products
JP2022040308A (en) * 2012-12-12 2022-03-10 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
AU2019236591B2 (en) * 2012-12-12 2022-02-24 Massachusetts Institute Of Technology Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
JP7013406B2 (en) 2012-12-12 2022-02-15 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
EP2931897B1 (en) 2012-12-12 2017-11-01 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
AU2017202542B2 (en) * 2012-12-12 2019-07-11 Massachusetts Institute Of Technology Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US8871445B2 (en) 2012-12-12 2014-10-28 The Broad Institute Inc. CRISPR-Cas component systems, methods and compositions for sequence manipulation
JP2019115348A (en) * 2012-12-12 2019-07-18 ザ・ブロード・インスティテュート・インコーポレイテッ Crispr-cas systems and methods for altering expression of gene products
US8889356B2 (en) 2012-12-12 2014-11-18 The Broad Institute Inc. CRISPR-Cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
EP2998400A1 (en) * 2012-12-12 2016-03-23 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
AU2019201608B2 (en) * 2012-12-12 2021-12-09 Massachusetts Institute Of Technology CRISPR-Cas systems and methods for altering expression of gene products
JP7228059B2 (en) 2012-12-12 2023-02-22 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for sequence engineering and therapeutic applications
US8889418B2 (en) 2012-12-12 2014-11-18 The Broad Institute Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
US8895308B1 (en) 2012-12-12 2014-11-25 The Broad Institute Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
EP2931898B1 (en) 2012-12-12 2016-03-09 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
JP2021166513A (en) * 2012-12-12 2021-10-21 ザ・ブロード・インスティテュート・インコーポレイテッド CRISPR-Cas COMPONENT SYSTEM, METHOD AND COMPOSITION FOR SEQUENCE MANIPULATION
EP2921557B1 (en) 2012-12-12 2016-07-13 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
US8906616B2 (en) 2012-12-12 2014-12-09 The Broad Institute Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
AU2019229420B2 (en) * 2012-12-12 2022-06-02 Massachusetts Institute Of Technology Engineering of systems, methods and optimized guide compositions for sequence manipulation
US8932814B2 (en) 2012-12-12 2015-01-13 The Broad Institute Inc. CRISPR-Cas nickase systems, methods and compositions for sequence manipulation in eukaryotes
JP2016129516A (en) * 2012-12-12 2016-07-21 ザ・ブロード・インスティテュート・インコーポレイテッド Engineering of systems, methods and optimized guide compositions for sequence manipulation
CN106170549A (en) * 2012-12-12 2016-11-30 布罗德研究所有限公司 For changing the CRISPR cas system of expression and the method for gene outcome
AU2019283878B2 (en) * 2012-12-12 2022-06-09 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
JP2018099131A (en) * 2012-12-12 2018-06-28 ザ・ブロード・インスティテュート・インコーポレイテッド Engineering of systems, methods and optimized guide compositions for sequence manipulation
US8945839B2 (en) 2012-12-12 2015-02-03 The Broad Institute Inc. CRISPR-Cas systems and methods for altering expression of gene products
AU2019280394B2 (en) * 2012-12-12 2022-06-09 Massachusetts Institute Of Technology CRISPR-Cas component systems, methods and compositions for sequence manipulation
JP2016504026A (en) * 2012-12-12 2016-02-12 ザ・ブロード・インスティテュート・インコーポレイテッド Engineering systems, methods and optimization guide compositions for sequence manipulation
JP2021104055A (en) * 2012-12-12 2021-07-26 ザ・ブロード・インスティテュート・インコーポレイテッド Crispr-cas systems and methods for altering expression of gene products
US8993233B2 (en) 2012-12-12 2015-03-31 The Broad Institute Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US11041173B2 (en) 2012-12-12 2021-06-22 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
JP2016501531A (en) * 2012-12-12 2016-01-21 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
AU2013359199C1 (en) * 2012-12-12 2021-06-17 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
JP7090775B2 (en) 2012-12-12 2022-06-24 ザ・ブロード・インスティテュート・インコーポレイテッド CRISPR-Cas systems and methods for altering expression of gene products
US8999641B2 (en) 2012-12-12 2015-04-07 The Broad Institute Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
EP2825654B1 (en) * 2012-12-12 2017-04-26 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
US9822372B2 (en) 2012-12-12 2017-11-21 The Broad Institute Inc. CRISPR-Cas component systems, methods and compositions for sequence manipulation
JP2016500262A (en) * 2012-12-12 2016-01-12 ザ・ブロード・インスティテュート・インコーポレイテッ CRISPR-Cas system and method for altering expression of gene products
EP2784162B1 (en) 2012-12-12 2015-04-08 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
JP7269990B2 (en) 2012-12-12 2023-05-09 ザ・ブロード・インスティテュート・インコーポレイテッド CRISPR-Cas Component Systems, Methods and Compositions for Sequence Engineering
EP2898075A1 (en) 2012-12-12 2015-07-29 The Broad Institute, Inc. Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
EP2764103B1 (en) 2012-12-12 2015-08-19 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
EP2896697B1 (en) 2012-12-12 2015-09-02 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
EP2931897A2 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
EP2764103A2 (en) 2012-12-12 2014-08-13 The Broad Institute, Inc. Crispr-cas systems and methods for altering expression of gene products
AU2013359262B2 (en) * 2012-12-12 2019-10-31 Massachusetts Institute Of Technology CRISPR-Cas component systems, methods and compositions for sequence manipulation
AU2013359199B2 (en) * 2012-12-12 2019-10-31 Massachusetts Institute Of Technology Delivery, engineering and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
AU2013359262C1 (en) * 2012-12-12 2021-05-13 Massachusetts Institute Of Technology CRISPR-Cas component systems, methods and compositions for sequence manipulation
EP2932421A1 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Methods, systems, and apparatus for identifying target sequences for cas enzymes or crispr-cas systems for target sequences and conveying results thereof
JP2016171817A (en) * 2012-12-12 2016-09-29 ザ・ブロード・インスティテュート・インコーポレイテッ Crispr-cas systems and methods for altering expression of gene products
EP2931898A2 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
US9023649B2 (en) * 2012-12-17 2015-05-05 President And Fellows Of Harvard College RNA-guided human genome engineering
EP2931891A4 (en) * 2012-12-17 2016-07-13 Harvard College Rna-guided human genome engineering
US11535863B2 (en) 2012-12-17 2022-12-27 President And Fellows Of Harvard College RNA-guided human genome engineering
US11512325B2 (en) 2012-12-17 2022-11-29 President And Fellows Of Harvard College RNA-guided human genome engineering
US11236359B2 (en) 2012-12-17 2022-02-01 President And Fellows Of Harvard College RNA-guided human genome engineering
AU2013363194B2 (en) * 2012-12-17 2019-05-16 President And Fellows Of Harvard College RNA-guided human genome engineering
US10435708B2 (en) 2012-12-17 2019-10-08 President And Fellows Of Harvard College RNA-guided human genome engineering
EP3553174A1 (en) * 2012-12-17 2019-10-16 President and Fellows of Harvard College Rna-guided human genome engineering
EP4282970A3 (en) * 2012-12-17 2024-01-17 President and Fellows of Harvard College Rna-guided human genome engineering
US10717990B2 (en) 2012-12-17 2020-07-21 President And Fellows Of Harvard College RNA-guided human genome engineering
JP2021048882A (en) * 2012-12-17 2021-04-01 プレジデント アンド フェローズ オブ ハーバード カレッジ Rna-guided human genome engineering
US10273501B2 (en) 2012-12-17 2019-04-30 President And Fellows Of Harvard College RNA-guided human genome engineering
JP2016501036A (en) * 2012-12-17 2016-01-18 プレジデント アンド フェローズ オブ ハーバード カレッジ RNA-induced human genome modification
US11365429B2 (en) 2012-12-17 2022-06-21 President And Fellows Of Harvard College RNA-guided human genome engineering
US11359211B2 (en) 2012-12-17 2022-06-14 President And Fellows Of Harvard College RNA-guided human genome engineering
US20140356958A1 (en) * 2012-12-17 2014-12-04 President And Fellows Of Harvard College RNA-Guided Human Genome Engineering
US9260723B2 (en) 2012-12-17 2016-02-16 President And Fellows Of Harvard College RNA-guided human genome engineering
US9970024B2 (en) 2012-12-17 2018-05-15 President And Fellows Of Harvard College RNA-guided human genome engineering
JP2016507244A (en) * 2013-02-27 2016-03-10 ヘルムホルツ・ツェントルム・ミュンヒェン・ドイチェス・フォルシュンクスツェントルム・フューア・ゲズントハイト・ウント・ウムベルト(ゲーエムベーハー)Helmholtz Zentrum MuenchenDeutsches Forschungszentrum fuer Gesundheit und Umwelt (GmbH) Gene editing in oocytes by Cas9 nuclease
EP2922393A1 (en) 2013-02-27 2015-09-30 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Gene editing in the oocyte by cas9 nucleases
US9783780B2 (en) 2013-02-27 2017-10-10 Helmholtz Zentrum München Deutsches Forschungszentrum Für Gesundheit Und Umwelt Gene editing in the oocyte by CAS9 nucleases
US10214723B2 (en) 2013-02-27 2019-02-26 Helmholtz Zentrum München Deutsches Forschungszentrum Für Gesundheit Und Umwelt Gene editing in the oocyte by Cas9 nucleases
US11299767B2 (en) 2013-03-12 2022-04-12 President And Fellows Of Harvard College Method for generating a three-dimensional nucleic acid containing matrix
US10125361B2 (en) 2013-03-14 2018-11-13 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US9410198B2 (en) 2013-03-14 2016-08-09 Caribou Biosciences, Inc. Compostions and methods of nucleic acid-targeting nucleic acids
US9909122B2 (en) 2013-03-14 2018-03-06 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US9725714B2 (en) 2013-03-14 2017-08-08 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US9260752B1 (en) 2013-03-14 2016-02-16 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US9803194B2 (en) 2013-03-14 2017-10-31 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US9809814B1 (en) 2013-03-14 2017-11-07 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US11312953B2 (en) 2013-03-14 2022-04-26 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
US10119133B2 (en) 2013-03-15 2018-11-06 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US9567604B2 (en) 2013-03-15 2017-02-14 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US10138476B2 (en) 2013-03-15 2018-11-27 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
JP2016512691A (en) * 2013-03-15 2016-05-09 ザ ジェネラル ホスピタル コーポレイション Increased specificity of RNA-guided genome editing using truncated guide RNA (tru-gRNA)
JP2016517276A (en) * 2013-03-15 2016-06-16 ザ ジェネラル ホスピタル コーポレイション Increased specificity of RNA-guided genome editing using RNA-induced FokI nuclease (RFN)
JP2020031637A (en) * 2013-03-15 2020-03-05 ザ ジェネラル ホスピタル コーポレイション Rna-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
JP2016512048A (en) * 2013-03-15 2016-04-25 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Plant genome manipulation using the CRISPR / Cas system
JP7053706B2 (en) 2013-03-15 2022-04-12 ザ ジェネラル ホスピタル コーポレイション Increased specificity of RNA-induced genome editing with shortened guide RNA (tru-gRNA)
JP2016512264A (en) * 2013-03-15 2016-04-25 ザ ジェネラル ホスピタル コーポレイション RNA-induced targeting of genomic and epigenomic regulatory proteins to specific genomic loci
JP2019205470A (en) * 2013-03-15 2019-12-05 リージェンツ オブ ザ ユニバーシティ オブ ミネソタ Engineering plant genomes using crispr/cas systems
US9885033B2 (en) 2013-03-15 2018-02-06 The General Hospital Corporation Increasing specificity for RNA-guided genome editing
JP7126588B2 (en) 2013-03-15 2022-08-26 ザ ジェネラル ホスピタル コーポレイション Increased specificity of RNA-guided genome editing using RNA-guided FokI nuclease (RFN)
JP2020039350A (en) * 2013-03-15 2020-03-19 ザ ジェネラル ホスピタル コーポレイション USING RNA-GUIDED FokI NUCLEASES (RFNS) TO INCREASE SPECIFICITY FOR RNA-GUIDED GENOME EDITING
JP7379572B2 (en) 2013-03-15 2023-11-14 ザ ジェネラル ホスピタル コーポレイション Increasing specificity of RNA-guided genome editing using truncated guide RNA (tru-gRNA)
US9567603B2 (en) 2013-03-15 2017-02-14 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US10544433B2 (en) 2013-03-15 2020-01-28 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US10526589B2 (en) 2013-03-15 2020-01-07 The General Hospital Corporation Multiplex guide RNAs
US10760064B2 (en) 2013-03-15 2020-09-01 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US10844403B2 (en) 2013-03-15 2020-11-24 The General Hospital Corporation Increasing specificity for RNA-guided genome editing
US10378027B2 (en) 2013-03-15 2019-08-13 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
JP2021118726A (en) * 2013-03-15 2021-08-12 ザ ジェネラル ホスピタル コーポレイション Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing
US11098326B2 (en) 2013-03-15 2021-08-24 The General Hospital Corporation Using RNA-guided FokI nucleases (RFNs) to increase specificity for RNA-guided genome editing
US10415059B2 (en) 2013-03-15 2019-09-17 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
US11634731B2 (en) 2013-03-15 2023-04-25 The General Hospital Corporation Using truncated guide RNAs (tru-gRNAs) to increase specificity for RNA-guided genome editing
JP2020120661A (en) * 2013-03-15 2020-08-13 ザ ジェネラル ホスピタル コーポレイション TO INCREASE SPECIFICITY FOR RNA-GUIDED GENOME EDITING USING TRUNCATED GUIDE RNAS (tru-gRNAs)
US11920152B2 (en) 2013-03-15 2024-03-05 The General Hospital Corporation Increasing specificity for RNA-guided genome editing
US11168338B2 (en) 2013-03-15 2021-11-09 The General Hospital Corporation RNA-guided targeting of genetic and epigenomic regulatory proteins to specific genomic loci
US20160186208A1 (en) * 2013-04-16 2016-06-30 Whitehead Institute For Biomedical Research Methods of Mutating, Modifying or Modulating Nucleic Acid in a Cell or Nonhuman Mammal
US10975390B2 (en) 2013-04-16 2021-04-13 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US10385359B2 (en) 2013-04-16 2019-08-20 Regeneron Pharmaceuticals, Inc. Targeted modification of rat genome
US10196652B2 (en) 2013-05-15 2019-02-05 Sangamo Therapeutics, Inc. Methods and compositions for treatment of a genetic condition
US9902974B2 (en) * 2013-05-15 2018-02-27 Sangamo Therapeutics, Inc. Methods and compositions for treatment of a genetic condition
JP2016521975A (en) * 2013-05-15 2016-07-28 サンガモ バイオサイエンシーズ, インコーポレイテッド Methods and compositions for the treatment of genetic conditions
US20160090607A1 (en) * 2013-05-15 2016-03-31 Sangamo Biosciences, Inc. Methods and compositions for treatment of a genetic condition
US10196651B2 (en) 2013-05-15 2019-02-05 Sangamo Therapeutics, Inc. Methods and compositions for treatment of a genetic condition
JP2021003107A (en) * 2013-05-29 2021-01-14 セレクティスCellectis Method for producing precise dna cleavage using cas9 nickase activity
EP3004349B1 (en) * 2013-05-29 2018-03-28 Cellectis S.A. A method for producing precise dna cleavage using cas9 nickase activity
EP3004349A1 (en) * 2013-05-29 2016-04-13 Cellectis S.A. A method for producing precise dna cleavage using cas9 nickase activity
JP2016520317A (en) * 2013-05-29 2016-07-14 セレクティスCellectis Method for producing precise DNA cleavage using CAS9 nickase activity
AU2014274939B2 (en) * 2013-06-04 2020-03-19 President And Fellows Of Harvard College RNA-guideded transcriptional regulation
US10767194B2 (en) 2013-06-04 2020-09-08 President And Fellows Of Harvard College RNA-guided transcriptional regulation
JP7376650B2 (en) 2013-06-04 2023-11-08 プレジデント アンド フェローズ オブ ハーバード カレッジ RNA-induced transcriptional regulation
EP3603679A1 (en) * 2013-06-04 2020-02-05 President and Fellows of Harvard College Rna-guideded transcriptional regulation
JP2019122384A (en) * 2013-06-04 2019-07-25 プレジデント アンド フェローズ オブ ハーバード カレッジ Rna guided transcriptional regulation
JP2022166077A (en) * 2013-06-04 2022-11-01 プレジデント アンド フェローズ オブ ハーバード カレッジ Rna-guided transcriptional regulation
JP2021045153A (en) * 2013-06-04 2021-03-25 プレジデント アンド フェローズ オブ ハーバード カレッジ Rna-guided transcriptional regulation
JP7119055B2 (en) 2013-06-04 2022-08-16 プレジデント アンド フェローズ オブ ハーバード カレッジ RNA-induced transcriptional regulation
JP2016521554A (en) * 2013-06-04 2016-07-25 プレジデント アンド フェローズ オブ ハーバード カレッジ RNA-induced transcriptional regulation
AU2020203977B2 (en) * 2013-06-04 2021-05-27 President And Fellows Of Harvard College RNA-guided transcriptional regulation
US10640789B2 (en) 2013-06-04 2020-05-05 President And Fellows Of Harvard College RNA-guided transcriptional regulation
EP3003392A4 (en) * 2013-06-04 2017-05-03 President and Fellows of Harvard College Rna-guideded transcriptional regulation
JP7036511B2 (en) 2013-06-04 2022-03-15 プレジデント アンド フェローズ オブ ハーバード カレッジ RNA-induced transcriptional regulation
EP4159243A1 (en) * 2013-06-04 2023-04-05 President and Fellows of Harvard College Rna-guided transcriptional regulation
EP3008186B1 (en) 2013-06-14 2018-11-28 Cellectis Methods for non-transgenic genome editing in plants
JP2016524472A (en) * 2013-06-17 2016-08-18 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery and use of CRISPR-Cas systems, vectors and compositions for liver targeting and therapy
US11597949B2 (en) 2013-06-17 2023-03-07 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
US10577630B2 (en) 2013-06-17 2020-03-03 The Broad Institute, Inc. Delivery and use of the CRISPR-Cas systems, vectors and compositions for hepatic targeting and therapy
JP2020063238A (en) * 2013-06-17 2020-04-23 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of postmitotic cells
EP3011030A1 (en) * 2013-06-17 2016-04-27 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
US10781444B2 (en) 2013-06-17 2020-09-22 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
US11008588B2 (en) 2013-06-17 2021-05-18 The Broad Institute, Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
US20160186213A1 (en) * 2013-06-17 2016-06-30 The Broad Institute Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
EP3011030B1 (en) * 2013-06-17 2023-11-08 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
US10946108B2 (en) 2013-06-17 2021-03-16 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for targeting disorders and diseases using viral components
JP2016523082A (en) * 2013-06-17 2016-08-08 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of mitotic cells
US10711285B2 (en) 2013-06-17 2020-07-14 The Broad Institute, Inc. Optimized CRISPR-Cas double nickase systems, methods and compositions for sequence manipulation
EP3597755A1 (en) * 2013-06-17 2020-01-22 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components
JP2016521995A (en) * 2013-06-17 2016-07-28 ザ・ブロード・インスティテュート・インコーポレイテッド Delivery, use and therapeutic applications of CRISPR-Cas systems and compositions for targeting disorders and diseases using viral components
US10011850B2 (en) 2013-06-21 2018-07-03 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to increase specificity for RNA-Guided Genome Editing
US11306328B2 (en) 2013-07-26 2022-04-19 President And Fellows Of Harvard College Genome engineering
JP2016528894A (en) * 2013-07-26 2016-09-23 プレジデント アンド フェローズ オブ ハーバード カレッジ Genome engineering
US10563225B2 (en) 2013-07-26 2020-02-18 President And Fellows Of Harvard College Genome engineering
US10954548B2 (en) 2013-08-09 2021-03-23 President And Fellows Of Harvard College Nuclease profiling system
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
US10508298B2 (en) 2013-08-09 2019-12-17 President And Fellows Of Harvard College Methods for identifying a target site of a CAS9 nuclease
US11920181B2 (en) 2013-08-09 2024-03-05 President And Fellows Of Harvard College Nuclease profiling system
US11773400B2 (en) 2013-08-22 2023-10-03 E.I. Du Pont De Nemours And Company Methods for producing genetic modifications in a plant genome without incorporating a selectable transgene marker, and compositions thereof
US11046948B2 (en) 2013-08-22 2021-06-29 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US10519457B2 (en) 2013-08-22 2019-12-31 E I Du Pont De Nemours And Company Soybean U6 polymerase III promoter and methods of use
US10682410B2 (en) 2013-09-06 2020-06-16 President And Fellows Of Harvard College Delivery system for functional nucleases
US10912833B2 (en) 2013-09-06 2021-02-09 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US10858639B2 (en) 2013-09-06 2020-12-08 President And Fellows Of Harvard College CAS9 variants and uses thereof
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US10597679B2 (en) 2013-09-06 2020-03-24 President And Fellows Of Harvard College Switchable Cas9 nucleases and uses thereof
US11299755B2 (en) 2013-09-06 2022-04-12 President And Fellows Of Harvard College Switchable CAS9 nucleases and uses thereof
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US9999671B2 (en) 2013-09-06 2018-06-19 President And Fellows Of Harvard College Delivery of negatively charged proteins using cationic lipids
US9737604B2 (en) 2013-09-06 2017-08-22 President And Fellows Of Harvard College Use of cationic lipids to deliver CAS9
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9340800B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College Extended DNA-sensing GRNAS
US11920128B2 (en) 2013-09-18 2024-03-05 Kymab Limited Methods, cells and organisms
EP3988649A1 (en) * 2013-09-18 2022-04-27 Kymab Limited Methods, cells and organisms
US11149267B2 (en) 2013-10-28 2021-10-19 The Broad Institute, Inc. Functional genomics using CRISPR-Cas systems, compositions, methods, screens and applications thereof
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US9834791B2 (en) 2013-11-07 2017-12-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US11390887B2 (en) 2013-11-07 2022-07-19 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10640788B2 (en) 2013-11-07 2020-05-05 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAs
US11820997B2 (en) 2013-12-11 2023-11-21 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US9228208B2 (en) 2013-12-11 2016-01-05 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
EP4349980A2 (en) 2013-12-11 2024-04-10 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US10208317B2 (en) 2013-12-11 2019-02-19 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse embryonic stem cell genome
WO2015088643A1 (en) 2013-12-11 2015-06-18 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US9546384B2 (en) 2013-12-11 2017-01-17 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse genome
EP3460063A1 (en) 2013-12-11 2019-03-27 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a genome
US10711280B2 (en) 2013-12-11 2020-07-14 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse ES cell genome
US11155795B2 (en) 2013-12-12 2021-10-26 The Broad Institute, Inc. CRISPR-Cas systems, crystal structure and uses thereof
US10550372B2 (en) 2013-12-12 2020-02-04 The Broad Institute, Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
US9068179B1 (en) 2013-12-12 2015-06-30 President And Fellows Of Harvard College Methods for correcting presenilin point mutations
US10465176B2 (en) 2013-12-12 2019-11-05 President And Fellows Of Harvard College Cas variants for gene editing
US9840699B2 (en) 2013-12-12 2017-12-12 President And Fellows Of Harvard College Methods for nucleic acid editing
US10377998B2 (en) 2013-12-12 2019-08-13 The Broad Institute, Inc. CRISPR-CAS systems and methods for altering expression of gene products, structural information and inducible modular CAS enzymes
AU2014362208B2 (en) * 2013-12-12 2021-02-11 President And Fellows Of Harvard College Cas variants for gene editing
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
US11124782B2 (en) 2013-12-12 2021-09-21 President And Fellows Of Harvard College Cas variants for gene editing
US11591581B2 (en) 2013-12-12 2023-02-28 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
US11407985B2 (en) 2013-12-12 2022-08-09 The Broad Institute, Inc. Delivery, use and therapeutic applications of the CRISPR-Cas systems and compositions for genome editing
US10851357B2 (en) 2013-12-12 2020-12-01 The Broad Institute, Inc. Compositions and methods of use of CRISPR-Cas systems in nucleotide repeat disorders
AU2021200375B2 (en) * 2013-12-12 2023-08-17 President And Fellows Of Harvard College Cas variants for gene editing
US11149259B2 (en) 2013-12-12 2021-10-19 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products, structural information and inducible modular Cas enzymes
US11597919B2 (en) 2013-12-12 2023-03-07 The Broad Institute Inc. Systems, methods and compositions for sequence manipulation with optimized functional CRISPR-Cas systems
US11028388B2 (en) 2014-03-05 2021-06-08 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for treating Usher syndrome and retinitis pigmentosa
US10253312B2 (en) 2014-03-10 2019-04-09 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating Leber's Congenital Amaurosis 10 (LCA10)
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US9938521B2 (en) 2014-03-10 2018-04-10 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating leber's congenital amaurosis 10 (LCA10)
US11339437B2 (en) 2014-03-10 2022-05-24 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
US11268086B2 (en) 2014-03-10 2022-03-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating Leber's Congenital Amaurosis 10 (LCA10)
US20170007679A1 (en) * 2014-03-25 2017-01-12 Editas Medicine Inc. Crispr/cas-related methods and compositions for treating hiv infection and aids
US11242525B2 (en) 2014-03-26 2022-02-08 Editas Medicine, Inc. CRISPR/CAS-related methods and compositions for treating sickle cell disease
EP3152319A4 (en) * 2014-06-05 2017-12-27 Sangamo BioSciences, Inc. Methods and compositions for nuclease design
US9970001B2 (en) 2014-06-05 2018-05-15 Sangamo Therapeutics, Inc. Methods and compositions for nuclease design
EP3708671A1 (en) 2014-06-06 2020-09-16 Regeneron Pharmaceuticals, Inc. Methods and compositions for modifying a targeted locus
WO2015188109A1 (en) 2014-06-06 2015-12-10 Regeneron Pharmaceuticals, Inc. Methods and compositions for modifying a targeted locus
AU2015277369B2 (en) * 2014-06-16 2021-08-19 The Johns Hopkins University Compositions and methods for the expression of CRISPR guide RNAs using the H1 promoter
EA039693B1 (en) * 2014-06-16 2022-02-28 Дзе Джонс Хопкинс Юниверсити COMPOSITIONS AND METHODS FOR THE EXPRESSION OF CRISPR GUIDE RNAs USING THE H1 PROMOTER
US10668173B2 (en) * 2014-06-16 2020-06-02 The Johns Hopkins University Compositions and methods for the expression of CRISPR guide RNAS using the H1 promoter
US10406245B2 (en) 2014-06-16 2019-09-10 The Johns Hopkins University Compositions and methods for the expression of CRISPR guide RNAs using the H1 promoter
WO2015195621A1 (en) * 2014-06-16 2015-12-23 The Johns Hopkins University Compositions and methods for the expression of crispr guide rnas using the h1 promoter
JP2017519500A (en) * 2014-06-16 2017-07-20 ザ・ジョンズ・ホプキンス・ユニバーシティー Compositions and methods for expression of CRISPR guide RNA using the H1 promoter
US10369234B2 (en) 2014-06-16 2019-08-06 The Johns Hopkins University Compositions and methods for the expression of CRISPR guide RNAs using the H1 promoter
US11896679B2 (en) 2014-06-16 2024-02-13 The Johns Hopkins University Compositions and methods for the expression of CRISPR guide RNAs using the H1 promoter
CN106852157A (en) * 2014-06-16 2017-06-13 约翰斯·霍普金斯大学 Composition and method for expressing guide CRISPR RNA using H1 promoters
JP2021052809A (en) * 2014-06-16 2021-04-08 ザ・ジョンズ・ホプキンス・ユニバーシティー Compositions and methods for expression of crispr guide rna using h1 promoter
US9907863B2 (en) 2014-06-16 2018-03-06 The Johns Hopkins University Compositions and methods for the expression of CRISPR guide RNAs using the H1 promoter
JP2017518082A (en) * 2014-06-17 2017-07-06 ポセイダ セラピューティクス, インコーポレイテッド Methods and uses for directing proteins to specific loci in the genome
EP3354732A1 (en) 2014-06-23 2018-08-01 Regeneron Pharmaceuticals, Inc. Nuclease-mediated dna assembly
US10626402B2 (en) 2014-06-23 2020-04-21 Regeneron Pharmaceuticals, Inc. Nuclease-mediated DNA assembly
US10273488B2 (en) 2014-06-23 2019-04-30 Regeneron Pharmaceuticals, Inc. Nuclease-mediated DNA assembly
US9738897B2 (en) 2014-06-23 2017-08-22 Regeneron Pharmaceuticals, Inc. Nuclease-mediated DNA assembly
CN106715694A (en) * 2014-06-23 2017-05-24 瑞泽恩制药公司 Nuclease-mediated DNA assembly
WO2015200334A1 (en) * 2014-06-23 2015-12-30 Regeneron Pharmaceuticals, Inc. Nuclease-mediated dna assembly
US11932859B2 (en) 2014-06-23 2024-03-19 Regeneron Pharmaceuticals, Inc. Nuclease-mediated DNA assembly
EP3708663A1 (en) 2014-06-23 2020-09-16 Regeneron Pharmaceuticals, Inc. Nuclease-mediated dna assembly
US9580715B2 (en) 2014-06-23 2017-02-28 Regeneron Pharmaceuticals, Inc. Nuclease-mediated DNA assembly
WO2015200805A2 (en) 2014-06-26 2015-12-30 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modifications and methods of use
EP3461885A1 (en) 2014-06-26 2019-04-03 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modifications and methods of use
US20170198268A1 (en) * 2014-07-09 2017-07-13 Gen9, Inc. Compositions and Methods for Site-Directed DNA Nicking and Cleaving
WO2016011080A3 (en) * 2014-07-14 2016-03-03 The Regents Of The University Of California Crispr/cas transcriptional modulation
US11578343B2 (en) 2014-07-30 2023-02-14 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10704062B2 (en) 2014-07-30 2020-07-07 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10077453B2 (en) 2014-07-30 2018-09-18 President And Fellows Of Harvard College CAS9 proteins including ligand-dependent inteins
US10519454B2 (en) 2014-08-06 2019-12-31 Toolgen Incorporated Genome editing using Campylobacter jejuni CRISPR/CAS system-derived RGEN
JP2017526387A (en) * 2014-08-06 2017-09-14 トゥールジェン インコーポレイテッド Genome editing using RGEN derived from Campylobacter jejuni CRISPR / CAS system
WO2016036754A1 (en) 2014-09-02 2016-03-10 The Regents Of The University Of California Methods and compositions for rna-directed target dna modification
US11560568B2 (en) 2014-09-12 2023-01-24 E. I. Du Pont De Nemours And Company Generation of site-specific-integration sites for complex trait loci in corn and soybean, and methods of use
US11400169B2 (en) 2014-10-09 2022-08-02 Seattle Children's Hospital Long poly(A) plasmids and methods for introduction of long poly(A) sequences into the plasmid
EP3204399A4 (en) * 2014-10-09 2018-04-25 Seattle Children's Hospital, dba Seattle Children's Research Institute Long poly (a) plasmids and methods for introduction of long poly (a) sequences into the plasmid
EP3998344A1 (en) * 2014-10-09 2022-05-18 Life Technologies Corporation Crispr oligonucleotides and gene editing
WO2016061374A1 (en) 2014-10-15 2016-04-21 Regeneron Pharmaceuticals, Inc. Methods and compositions for generating or maintaining pluripotent cells
EP3561052A1 (en) 2014-10-15 2019-10-30 Regeneron Pharmaceuticals, Inc. Methods and compositions for generating or maintaining pluripotent cells
US20230151345A1 (en) * 2014-10-24 2023-05-18 Life Technologies Corporation Compositions and methods for enhancing homologous recombination
CN107429246A (en) * 2014-10-31 2017-12-01 麻省理工学院 Large-scale parallel for CRISPR combines science of heredity
US10584322B2 (en) 2014-11-06 2020-03-10 Dupont Us Holding, Llc Peptide-mediated delivery of RNA-guided endonuclease into cells
JP2017532979A (en) * 2014-11-06 2017-11-09 イー・アイ・デュポン・ドウ・ヌムール・アンド・カンパニーE.I.Du Pont De Nemours And Company Peptide-mediated transport of RNA-induced endonuclease into cells
US10208298B2 (en) 2014-11-06 2019-02-19 E.I. Du Pont De Nemours And Company Peptide-mediated delivery of RNA-guided endonuclease into cells
WO2016073433A1 (en) * 2014-11-06 2016-05-12 E. I. Du Pont De Nemours And Company Peptide-mediated delivery of rna-guided endonuclease into cells
EP3597740A1 (en) * 2014-11-06 2020-01-22 E. I. du Pont de Nemours and Company Peptide-mediated delivery of rna-guided endonuclease into cells
EA038321B1 (en) * 2014-11-06 2021-08-09 Е.И. Дюпон Де Немур Энд Компани Peptide-mediated delivery of rna-guided endonuclease into cells
CN107406838A (en) * 2014-11-06 2017-11-28 纳幕尔杜邦公司 Peptide-mediated delivering of the endonuclease of RNA guiding into cell
AU2015343307B2 (en) * 2014-11-06 2021-05-20 Iff Us Holding, Llc Peptide-mediated delivery of RNA-guided endonuclease into cells
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US10858662B2 (en) 2014-11-19 2020-12-08 Institute For Basic Science Genome editing with split Cas9 expressed from two vectors
WO2016080795A1 (en) * 2014-11-19 2016-05-26 기초과학연구원 Method for regulating gene expression using cas9 protein expressed from two vectors
WO2016081923A2 (en) 2014-11-21 2016-05-26 Regeneron Pharmaceuticals, Inc. METHODS AND COMPOSITIONS FOR TARGETED GENETIC MODIFICATION USING PAIRED GUIDE RNAs
EP3521437A1 (en) 2014-11-21 2019-08-07 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modification using paired guide rnas
WO2016083811A1 (en) 2014-11-27 2016-06-02 Imperial Innovations Limited Genome editing methods
US10900034B2 (en) 2014-12-03 2021-01-26 Agilent Technologies, Inc. Guide RNA with chemical modifications
US10337001B2 (en) 2014-12-03 2019-07-02 Agilent Technologies, Inc. Guide RNA with chemical modifications
US10278372B2 (en) 2014-12-10 2019-05-07 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US11234418B2 (en) 2014-12-10 2022-02-01 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US10993419B2 (en) 2014-12-10 2021-05-04 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US9888673B2 (en) 2014-12-10 2018-02-13 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
US11624078B2 (en) 2014-12-12 2023-04-11 The Broad Institute, Inc. Protected guide RNAS (pgRNAS)
US10696986B2 (en) 2014-12-12 2020-06-30 The Board Institute, Inc. Protected guide RNAS (PGRNAS)
WO2016100857A1 (en) 2014-12-19 2016-06-23 Regeneron Pharmaceuticals, Inc. Stem cells for modeling type 2 diabetes
EP3653048A1 (en) 2014-12-19 2020-05-20 Regeneron Pharmaceuticals, Inc. Methods and compositions for targeted genetic modification through single-step multiple targeting
WO2016120480A1 (en) 2015-01-29 2016-08-04 Meiogenix Method for inducing targeted meiotic recombinations
EP3798302A1 (en) 2015-01-29 2021-03-31 Meiogenix Method for inducing targeted meiotic recombinations
US11306309B2 (en) 2015-04-06 2022-04-19 The Board Of Trustees Of The Leland Stanford Junior University Chemically modified guide RNAs for CRISPR/CAS-mediated gene regulation
US11535846B2 (en) 2015-04-06 2022-12-27 The Board Of Trustees Of The Leland Stanford Junior University Chemically modified guide RNAS for CRISPR/Cas-mediated gene regulation
US11851652B2 (en) 2015-04-06 2023-12-26 The Board Of Trustees Of The Leland Stanford Junior Compositions comprising chemically modified guide RNAs for CRISPR/Cas-mediated editing of HBB
US11180793B2 (en) 2015-04-24 2021-11-23 Editas Medicine, Inc. Evaluation of Cas9 molecule/guide RNA molecule complexes
WO2016176617A2 (en) 2015-04-29 2016-11-03 New York University Method for treating high-grade gliomas
WO2016178207A1 (en) 2015-05-04 2016-11-10 Ramot At Tel-Aviv University Ltd. Methods and kits for fragmenting dna
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
JP2021112211A (en) * 2015-06-17 2021-08-05 ポセイダ セラピューティクス, インコーポレイテッド Compositions and methods for directing proteins to specific loci in genome
CN108026519A (en) * 2015-06-17 2018-05-11 波赛达治疗公司 The composition and method for particular locus protein being directed in genome
AU2021250992B2 (en) * 2015-06-17 2023-09-28 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
EP3929286A1 (en) * 2015-06-17 2021-12-29 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
JP2018521689A (en) * 2015-06-17 2018-08-09 ポセイダ セラピューティクス, インコーポレイテッド Compositions and methods for directing proteins to specific loci in the genome
JP7170090B2 (en) 2015-06-17 2022-11-11 ポセイダ セラピューティクス, インコーポレイテッド Compositions and methods for directing proteins to specific loci within the genome
US11473082B2 (en) 2015-06-17 2022-10-18 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
WO2016205554A1 (en) * 2015-06-17 2016-12-22 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
AU2016278226B2 (en) * 2015-06-17 2021-08-12 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
US10876100B2 (en) 2015-06-18 2020-12-29 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
US11578312B2 (en) 2015-06-18 2023-02-14 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of CAS9 orthologs and variants for sequence manipulation
US10494621B2 (en) 2015-06-18 2019-12-03 The Broad Institute, Inc. Crispr enzyme mutations reducing off-target effects
WO2016210271A1 (en) * 2015-06-24 2016-12-29 Sigma-Aldrich Co. Llc Cell cycle dependent genome regulation and modification
US11266692B2 (en) 2015-07-31 2022-03-08 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US11583556B2 (en) 2015-07-31 2023-02-21 Regents Of The University Of Minnesota Modified cells and methods of therapy
US10166255B2 (en) 2015-07-31 2019-01-01 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US11903966B2 (en) 2015-07-31 2024-02-20 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US10406177B2 (en) 2015-07-31 2019-09-10 Regents Of The University Of Minnesota Modified cells and methods of therapy
US11925664B2 (en) 2015-07-31 2024-03-12 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11147837B2 (en) 2015-07-31 2021-10-19 Regents Of The University Of Minnesota Modified cells and methods of therapy
US11642374B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11642375B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
WO2017027910A1 (en) 2015-08-14 2017-02-23 The University Of Sydney Connexin 45 inhibition for therapy
EP4043074A1 (en) 2015-08-14 2022-08-17 The University of Sydney Connexin 45 inhibition for therapy
US11427817B2 (en) 2015-08-25 2022-08-30 Duke University Compositions and methods of improving specificity in genomic engineering using RNA-guided endonucleases
EP4036236A1 (en) 2015-08-28 2022-08-03 The General Hospital Corporation Engineered crispr-cas9 nucleases
US10093910B2 (en) 2015-08-28 2018-10-09 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US11060078B2 (en) 2015-08-28 2021-07-13 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
WO2017040348A1 (en) 2015-08-28 2017-03-09 The General Hospital Corporation Engineered crispr-cas9 nucleases
US10526591B2 (en) 2015-08-28 2020-01-07 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US9926546B2 (en) 2015-08-28 2018-03-27 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US10633642B2 (en) 2015-08-28 2020-04-28 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US9512446B1 (en) 2015-08-28 2016-12-06 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
EP3362571A4 (en) * 2015-10-13 2019-07-10 Duke University Genome engineering with type i crispr systems in eukaryotic cells
EP4089175A1 (en) * 2015-10-13 2022-11-16 Duke University Genome engineering with type i crispr systems in eukaryotic cells
US11421251B2 (en) 2015-10-13 2022-08-23 Duke University Genome engineering with type I CRISPR systems in eukaryotic cells
US20170121693A1 (en) * 2015-10-23 2017-05-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
US11214780B2 (en) 2015-10-23 2022-01-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2017075335A1 (en) 2015-10-28 2017-05-04 Voyager Therapeutics, Inc. Regulatable expression using adeno-associated virus (aav)
US11542554B2 (en) 2015-11-03 2023-01-03 President And Fellows Of Harvard College Method and apparatus for volumetric imaging
US10240145B2 (en) * 2015-11-25 2019-03-26 The Board Of Trustees Of The Leland Stanford Junior University CRISPR/Cas-mediated genome editing to treat EGFR-mutant lung cancer
WO2017158153A1 (en) 2016-03-17 2017-09-21 Imba - Institut Für Molekulare Biotechnologie Gmbh Conditional crispr sgrna expression
EP3219799A1 (en) 2016-03-17 2017-09-20 IMBA-Institut für Molekulare Biotechnologie GmbH Conditional crispr sgrna expression
US11512311B2 (en) 2016-03-25 2022-11-29 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (A1AT) deficiency
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11713485B2 (en) 2016-04-25 2023-08-01 President And Fellows Of Harvard College Hybridization chain reaction methods for in situ molecular detection
WO2017186718A1 (en) * 2016-04-25 2017-11-02 Universität Basel Allele editing and applications thereof
WO2017201476A1 (en) 2016-05-20 2017-11-23 Regeneron Pharmaceuticals, Inc. Methods for breaking immunological tolerance using multiple guide rnas
EP3604527A1 (en) * 2016-06-02 2020-02-05 Sigma Aldrich Co. LLC Using programmable dna binding proteins to enhance targeted genome modification
EP3272867A1 (en) * 2016-06-02 2018-01-24 Sigma-Aldrich Co. LLC Using programmable dna binding proteins to enhance targeted genome modification
US10266851B2 (en) 2016-06-02 2019-04-23 Sigma-Aldrich Co. Llc Using programmable DNA binding proteins to enhance targeted genome modification
EP3907286A1 (en) * 2016-06-02 2021-11-10 Sigma-Aldrich Co., LLC Using programmable dna binding proteins to enhance targeted genome modification
US10767175B2 (en) 2016-06-08 2020-09-08 Agilent Technologies, Inc. High specificity genome editing using chemically modified guide RNAs
WO2018022967A1 (en) 2016-07-28 2018-02-01 Regeneron Pharmaceuticals, Inc. Gpr156 variants and uses thereof
EP4230648A2 (en) 2016-07-28 2023-08-23 Regeneron Pharmaceuticals, Inc. Gpr156 variants and uses thereof
WO2018023014A1 (en) 2016-07-29 2018-02-01 Regeneron Pharmaceuticals, Inc. Mice comprising mutations resulting in expression of c-truncated fibrillin-1
US11566263B2 (en) 2016-08-02 2023-01-31 Editas Medicine, Inc. Compositions and methods for treating CEP290 associated disease
US10947530B2 (en) 2016-08-03 2021-03-16 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11912987B2 (en) 2016-08-03 2024-02-27 KSQ Therapeutics, Inc. Methods for screening for cancer targets
US11702651B2 (en) 2016-08-03 2023-07-18 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11078481B1 (en) 2016-08-03 2021-08-03 KSQ Therapeutics, Inc. Methods for screening for cancer targets
US10113163B2 (en) 2016-08-03 2018-10-30 President And Fellows Of Harvard College Adenosine nucleobase editors and uses thereof
US11661590B2 (en) 2016-08-09 2023-05-30 President And Fellows Of Harvard College Programmable CAS9-recombinase fusion proteins and uses thereof
US11542509B2 (en) 2016-08-24 2023-01-03 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
US11078483B1 (en) 2016-09-02 2021-08-03 KSQ Therapeutics, Inc. Methods for measuring and improving CRISPR reagent function
US11946163B2 (en) 2016-09-02 2024-04-02 KSQ Therapeutics, Inc. Methods for measuring and improving CRISPR reagent function
WO2018054911A1 (en) 2016-09-23 2018-03-29 Bayer Cropscience Nv Targeted genome optimization in plants
US11306324B2 (en) 2016-10-14 2022-04-19 President And Fellows Of Harvard College AAV delivery of nucleobase editors
WO2018073237A1 (en) 2016-10-17 2018-04-26 The University Court Of The University Of Edinburgh Swine comprising modified cd163 and associated methods
US11154574B2 (en) 2016-10-18 2021-10-26 Regents Of The University Of Minnesota Tumor infiltrating lymphocytes and methods of therapy
US10912797B2 (en) 2016-10-18 2021-02-09 Intima Bioscience, Inc. Tumor infiltrating lymphocytes and methods of therapy
US10745677B2 (en) 2016-12-23 2020-08-18 President And Fellows Of Harvard College Editing of CCR5 receptor gene to protect against HIV infection
US11820969B2 (en) 2016-12-23 2023-11-21 President And Fellows Of Harvard College Editing of CCR2 receptor gene to protect against HIV infection
US11859219B1 (en) 2016-12-30 2024-01-02 Flagship Pioneering Innovations V, Inc. Methods of altering a target nucleotide sequence with an RNA-guided nuclease and a single guide RNA
WO2018136758A1 (en) 2017-01-23 2018-07-26 Regeneron Pharmaceuticals, Inc. Hsd17b13 variants and uses thereof
US11466271B2 (en) 2017-02-06 2022-10-11 Novartis Ag Compositions and methods for the treatment of hemoglobinopathies
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
US11851690B2 (en) 2017-03-14 2023-12-26 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
US11268082B2 (en) 2017-03-23 2022-03-08 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable DNA binding proteins
US11913015B2 (en) 2017-04-17 2024-02-27 University Of Maryland, College Park Embryonic cell cultures and methods of using the same
WO2018195545A2 (en) 2017-04-21 2018-10-25 The General Hospital Corporation Variants of cpf1 (cas12a) with altered pam specificity
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
WO2018218166A1 (en) 2017-05-25 2018-11-29 The General Hospital Corporation Using split deaminases to limit unwanted off-target base editor deamination
WO2018218206A1 (en) 2017-05-25 2018-11-29 The General Hospital Corporation Bipartite base editor (bbe) architectures and type-ii-c-cas9 zinc finger editing
WO2018226560A1 (en) 2017-06-05 2018-12-13 Regeneron Pharmaceuticals, Inc. B4galt1 variants and uses thereof
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
US11098297B2 (en) 2017-06-09 2021-08-24 Editas Medicine, Inc. Engineered Cas9 nucleases
US11814624B2 (en) 2017-06-15 2023-11-14 The Regents Of The University Of California Targeted non-viral DNA insertions
WO2019006034A1 (en) 2017-06-27 2019-01-03 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized asgr1 locus
US11696572B2 (en) 2017-06-27 2023-07-11 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ASGR1 locus
US11098325B2 (en) 2017-06-30 2021-08-24 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
US11732274B2 (en) 2017-07-28 2023-08-22 President And Fellows Of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (PACE)
WO2019028029A1 (en) 2017-07-31 2019-02-07 Regeneron Pharmaceuticals, Inc. Assessment of crispr/cas-induced recombination with an exogenous donor nucleic acid in vivo
WO2019028023A2 (en) 2017-07-31 2019-02-07 Regeneron Pharmaceuticals, Inc. Methods and compositions for assessing crispr/cas-mediated disruption or excision and crispr/cas-induced recombination with an exogenous donor nucleic acid in vivo
WO2019028032A1 (en) 2017-07-31 2019-02-07 Regeneron Pharmaceuticals, Inc. Cas-transgenic mouse embryonic stem cells and mice and uses thereof
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
US11932884B2 (en) 2017-08-30 2024-03-19 President And Fellows Of Harvard College High efficiency base editors comprising Gam
WO2019050948A1 (en) * 2017-09-05 2019-03-14 Regeneron Pharmaceuticals, Inc. Delivery of a gene-editing system with a single retroviral particle and methods of generation and use
WO2019050899A1 (en) 2017-09-06 2019-03-14 Regeneron Pharmaceuticals, Inc. Single immunoglobulin interleukin-1 receptor related (sigirr) variants and uses thereof
WO2019051033A1 (en) 2017-09-07 2019-03-14 Regeneron Pharmaceuticals, Inc. Solute carrier family 14 member 1 (slc14a1) variants and uses thereof
WO2019067875A1 (en) 2017-09-29 2019-04-04 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus and methods of use
EP4276185A2 (en) 2017-09-29 2023-11-15 Regeneron Pharmaceuticals, Inc. Rodents comprising a humanized ttr locus and methods of use
WO2019079238A1 (en) 2017-10-16 2019-04-25 Regeneron Pharmaceuticals, Inc. Cornulin (crnn) variants and uses thereof
US11795443B2 (en) 2017-10-16 2023-10-24 The Broad Institute, Inc. Uses of adenosine base editors
US11033584B2 (en) 2017-10-27 2021-06-15 The Regents Of The University Of California Targeted replacement of endogenous T cell receptors
US11590171B2 (en) 2017-10-27 2023-02-28 The Regents Of The University Of California Targeted replacement of endogenous T cell receptors
US11331346B2 (en) 2017-10-27 2022-05-17 The Regents Of The University Of California Targeted replacement of endogenous T cell receptors
US11083753B1 (en) 2017-10-27 2021-08-10 The Regents Of The University Of California Targeted replacement of endogenous T cell receptors
WO2019094735A1 (en) 2017-11-10 2019-05-16 Regeneron Pharmaceuticals, Inc. Non-human animals comprising slc30a8 mutation and methods of use
EP4299732A2 (en) 2017-11-30 2024-01-03 Regeneron Pharmaceuticals, Inc. Rats comprising a humanized trkb locus
WO2019108983A1 (en) 2017-11-30 2019-06-06 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized trkb locus
WO2019126578A1 (en) * 2017-12-20 2019-06-27 Poseida Therapeutics, Inc. Compositions and methods for directing proteins to specific loci in the genome
WO2019123014A1 (en) * 2017-12-22 2019-06-27 G+Flas Life Sciences Chimeric genome engineering molecules and methods
US11293019B2 (en) 2017-12-22 2022-04-05 Gflas Life Sciences, Inc. Chimeric genome engineering molecules and methods
WO2019138083A1 (en) 2018-01-12 2019-07-18 Basf Se Gene underlying the number of spikelets per spike qtl in wheat on chromosome 7a
US10767193B2 (en) 2018-02-15 2020-09-08 Sigma-Aldrich Co. Llc Engineered CAS9 systems for eukaryotic genome modification
WO2019183123A1 (en) 2018-03-19 2019-09-26 Regeneron Pharmaceuticals, Inc. Transcription modulation in animals using crispr/cas systems
US11434491B2 (en) 2018-04-19 2022-09-06 The Regents Of The University Of California Compositions and methods for gene editing
WO2019217803A1 (en) 2018-05-10 2019-11-14 Auxolytic Ltd. Gene therapy methods and compositions using auxotrophic regulatable cells
EP3790976A4 (en) * 2018-05-10 2022-08-10 Syngenta Participations Ag Methods and compositions for targeted editing of polynucleotides
CN112105732A (en) * 2018-05-10 2020-12-18 先正达参股股份有限公司 Methods and compositions for targeted editing of polynucleotides
WO2019234750A1 (en) 2018-06-07 2019-12-12 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Methods of regenerating and transforming cannabis
WO2019234754A1 (en) 2018-06-07 2019-12-12 The State Of Israel, Ministry Of Agriculture & Rural Development, Agricultural Research Organization (Aro) (Volcani Center) Nucleic acid constructs and methods of using same
WO2020030783A2 (en) 2018-08-10 2020-02-13 Vib Vzw Means and methods for drought tolerance in crops
CN113166753A (en) * 2018-08-21 2021-07-23 西格马-奥尔德里奇有限责任公司 Down-regulation of cytoplasmic DNA sensor pathway
WO2020041313A1 (en) * 2018-08-21 2020-02-27 Sigma-Aldrich Co. Llc Down-regulation of the cytosolic dna sensor pathway
WO2020056122A1 (en) 2018-09-13 2020-03-19 Regeneron Pharmaceuticals, Inc. Complement factor h gene knockout rat as a model of c3 glomerulopathy
WO2020123377A1 (en) 2018-12-10 2020-06-18 Neoimmunetech, Inc. Nrf-2 deficient cells and uses thereof
US11807878B2 (en) 2018-12-14 2023-11-07 Pioneer Hi-Bred International, Inc. CRISPR-Cas systems for genome editing
US10934536B2 (en) 2018-12-14 2021-03-02 Pioneer Hi-Bred International, Inc. CRISPR-CAS systems for genome editing
WO2020128478A1 (en) 2018-12-19 2020-06-25 King's College London Immunotherapeutic methods and compositions
WO2020131632A1 (en) 2018-12-20 2020-06-25 Regeneron Pharmaceuticals, Inc. Nuclease-mediated repeat expansion
WO2020163396A1 (en) 2019-02-04 2020-08-13 The General Hospital Corporation Adenine dna base editor variants with reduced off-target rna editing
US10947517B2 (en) 2019-02-15 2021-03-16 Sigma-Aldrich Co. Llc CRISPR/Cas fusion proteins and systems
WO2020169974A1 (en) 2019-02-19 2020-08-27 King's College London Hypoxia-responsive chimeric antigen receptors
WO2020190932A1 (en) 2019-03-18 2020-09-24 Regeneron Pharmaceuticals, Inc. Crispr/cas screening platform to identify genetic modifiers of tau seeding or aggregation
EP4317950A2 (en) 2019-03-18 2024-02-07 Regeneron Pharmaceuticals, Inc. Crispr/cas screening platform to identify genetic modifiers of tau seeding or aggregation
WO2020190927A1 (en) 2019-03-18 2020-09-24 Regeneron Pharmaceuticals, Inc. Crispr/cas dropout screening platform to reveal genetic vulnerabilities associated with tau aggregation
US11643652B2 (en) 2019-03-19 2023-05-09 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11447770B1 (en) 2019-03-19 2022-09-20 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
US11795452B2 (en) 2019-03-19 2023-10-24 The Broad Institute, Inc. Methods and compositions for prime editing nucleotide sequences
WO2020206162A1 (en) 2019-04-03 2020-10-08 Regeneron Pharmaceuticals, Inc. Methods and compositions for insertion of antibody coding sequences into a safe harbor locus
WO2020206134A1 (en) 2019-04-04 2020-10-08 Regeneron Pharmaceuticals, Inc. Methods for scarless introduction of targeted modifications into targeting vectors
US11111504B2 (en) 2019-04-04 2021-09-07 Regeneron Pharmaceuticals, Inc. Methods for scarless introduction of targeted modifications into targeting vectors
US11499164B2 (en) 2019-04-04 2022-11-15 Regeneran Pharmaceuticals, Inc. Methods for scarless introduction of targeted modifications into targeting vectors
WO2020206139A1 (en) 2019-04-04 2020-10-08 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized coagulation factor 12 locus
WO2020212541A1 (en) 2019-04-16 2020-10-22 University Of Nottingham Fungal strains, production and uses thereof
WO2020229241A1 (en) 2019-05-10 2020-11-19 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2020247452A1 (en) 2019-06-04 2020-12-10 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus with a beta-slip mutation and methods of use
WO2020247812A1 (en) 2019-06-07 2020-12-10 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized albumin locus
WO2020252340A1 (en) 2019-06-14 2020-12-17 Regeneron Pharmaceuticals, Inc. Models of tauopathy
WO2021050398A1 (en) * 2019-09-10 2021-03-18 The Regents Of The University Of California Synthetic lethality screening platform for cells undergoing alt
WO2021048316A1 (en) 2019-09-12 2021-03-18 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2021050940A1 (en) 2019-09-13 2021-03-18 Regeneron Pharmaceuticals, Inc. Transcription modulation in animals using crispr/cas systems delivered by lipid nanoparticles
WO2021069387A1 (en) 2019-10-07 2021-04-15 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2021092513A1 (en) 2019-11-08 2021-05-14 Regeneron Pharmaceuticals, Inc. Crispr and aav strategies for x-linked juvenile retinoschisis therapy
US11963982B2 (en) 2019-11-08 2024-04-23 Editas Medicine, Inc. CRISPR/RNA-guided nuclease systems and methods
WO2021108363A1 (en) 2019-11-25 2021-06-03 Regeneron Pharmaceuticals, Inc. Crispr/cas-mediated upregulation of humanized ttr allele
WO2021110582A1 (en) 2019-12-03 2021-06-10 Basf Se Regulatory nucleic acid molecules for enhancing gene expression in plants
WO2021154791A1 (en) 2020-01-28 2021-08-05 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized pnpla3 locus and methods of use
WO2021158883A1 (en) 2020-02-07 2021-08-12 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized klkb1 locus and methods of use
WO2021178556A1 (en) 2020-03-04 2021-09-10 Regeneron Pharmaceuticals, Inc. Methods and compositions for sensitization of tumor cells to immune therapy
WO2021195079A1 (en) 2020-03-23 2021-09-30 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus comprising a v30m mutation and methods of use
WO2021202938A1 (en) 2020-04-03 2021-10-07 Creyon Bio, Inc. Oligonucleotide-based machine learning
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
WO2021263146A2 (en) 2020-06-26 2021-12-30 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ace2 locus
WO2022120022A1 (en) 2020-12-02 2022-06-09 Regeneron Pharmaceuticals, Inc. Crispr sam biosensor cell lines and methods of use thereof
US11965184B2 (en) 2021-02-10 2024-04-23 Sigma-Aldrich Co. Llc CRISPR/Cas fusion proteins and systems
GB202103131D0 (en) 2021-03-05 2021-04-21 Biosystems Tech Limited Method for preparation of research organisms
WO2022240846A1 (en) 2021-05-10 2022-11-17 Sqz Biotechnologies Company Methods for delivering genome editing molecules to the nucleus or cytosol of a cell and uses thereof
WO2022251644A1 (en) 2021-05-28 2022-12-01 Lyell Immunopharma, Inc. Nr4a3-deficient immune cells and uses thereof
WO2022256437A1 (en) 2021-06-02 2022-12-08 Lyell Immunopharma, Inc. Nr4a3-deficient immune cells and uses thereof
US11884915B2 (en) 2021-09-10 2024-01-30 Agilent Technologies, Inc. Guide RNAs with chemical modification for prime editing
WO2023064924A1 (en) 2021-10-14 2023-04-20 Codiak Biosciences, Inc. Modified producer cells for extracellular vesicle production
WO2023077053A2 (en) 2021-10-28 2023-05-04 Regeneron Pharmaceuticals, Inc. Crispr/cas-related methods and compositions for knocking out c5
WO2023081847A1 (en) 2021-11-04 2023-05-11 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a modified cacng1 locus
WO2023108047A1 (en) 2021-12-08 2023-06-15 Regeneron Pharmaceuticals, Inc. Mutant myocilin disease model and uses thereof
WO2023111541A1 (en) 2021-12-14 2023-06-22 The University Of Warwick Methods to increase yields in crops
GB202118058D0 (en) 2021-12-14 2022-01-26 Univ Warwick Methods to increase yields in crops
EP4198124A1 (en) 2021-12-15 2023-06-21 Versitech Limited Engineered cas9-nucleases and method of use thereof
WO2023122506A1 (en) 2021-12-20 2023-06-29 Regeneron Pharmaceuticals, Inc. Non-human animals comprising humanized ace2 and tmprss loci
WO2023129974A1 (en) 2021-12-29 2023-07-06 Bristol-Myers Squibb Company Generation of landing pad cell lines
WO2023150181A1 (en) 2022-02-01 2023-08-10 President And Fellows Of Harvard College Methods and compositions for treating cancer
WO2023150620A1 (en) 2022-02-02 2023-08-10 Regeneron Pharmaceuticals, Inc. Crispr-mediated transgene insertion in neonatal cells
WO2023150798A1 (en) 2022-02-07 2023-08-10 Regeneron Pharmaceuticals, Inc. Compositions and methods for defining optimal treatment timeframes in lysosomal disease
WO2023212677A2 (en) 2022-04-29 2023-11-02 Regeneron Pharmaceuticals, Inc. Identification of tissue-specific extragenic safe harbors for gene therapy approaches
WO2023220603A1 (en) 2022-05-09 2023-11-16 Regeneron Pharmaceuticals, Inc. Vectors and methods for in vivo antibody production
WO2023225665A1 (en) 2022-05-19 2023-11-23 Lyell Immunopharma, Inc. Polynucleotides targeting nr4a3 and uses thereof
WO2023235726A2 (en) 2022-05-31 2023-12-07 Regeneron Pharmaceuticals, Inc. Crispr interference therapeutics for c9orf72 repeat expansion disease
WO2023235725A2 (en) 2022-05-31 2023-12-07 Regeneron Pharmaceuticals, Inc. Crispr-based therapeutics for c9orf72 repeat expansion disease
WO2024003579A1 (en) 2022-06-30 2024-01-04 University Of Newcastle Upon Tyne Preventing disease recurrence in mitochondrial replacement therapy
WO2024026488A2 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a modified transferrin receptor locus
WO2024026474A1 (en) 2022-07-29 2024-02-01 Regeneron Pharmaceuticals, Inc. Compositions and methods for transferrin receptor (tfr)-mediated delivery to the brain and muscle
WO2024031053A1 (en) 2022-08-05 2024-02-08 Regeneron Pharmaceuticals, Inc. Aggregation-resistant variants of tdp-43
WO2024064958A1 (en) 2022-09-23 2024-03-28 Lyell Immunopharma, Inc. Methods for culturing nr4a-deficient cells
WO2024064952A1 (en) 2022-09-23 2024-03-28 Lyell Immunopharma, Inc. Methods for culturing nr4a-deficient cells overexpressing c-jun
WO2024073606A1 (en) 2022-09-28 2024-04-04 Regeneron Pharmaceuticals, Inc. Antibody resistant modified receptors to enhance cell-based therapies
WO2024077174A1 (en) 2022-10-05 2024-04-11 Lyell Immunopharma, Inc. Methods for culturing nr4a-deficient cells

Also Published As

Publication number Publication date
KR102243092B1 (en) 2021-04-22
EP3138911B1 (en) 2018-12-05
EP3138909A1 (en) 2017-03-08
ES2769310T3 (en) 2020-06-25
EP3138910B1 (en) 2017-09-20
AU2020273316B2 (en) 2023-05-18
US20170073705A1 (en) 2017-03-16
SG10202107423UA (en) 2021-08-30
EP3138911A1 (en) 2017-03-08
JP2020120674A (en) 2020-08-13
DK3138910T3 (en) 2017-10-16
IL238856B (en) 2018-03-29
US20210079427A1 (en) 2021-03-18
LT3138911T (en) 2019-02-25
PL3138912T3 (en) 2019-04-30
CA2977152C (en) 2019-04-09
IL291129A (en) 2022-05-01
SG10201910987SA (en) 2020-01-30
EP3141604A1 (en) 2017-03-15
KR20230070065A (en) 2023-05-19
IL291129B1 (en) 2023-03-01
AU2019201344A1 (en) 2019-03-21
EP3138910A1 (en) 2017-03-08
US20210207173A1 (en) 2021-07-08
US20160298133A1 (en) 2016-10-13
PT3363902T (en) 2019-12-19
PT3360964T (en) 2019-10-29
IL257178A (en) 2018-03-29
CN105142669B (en) 2018-07-03
PL3360964T3 (en) 2020-03-31
IL238856A0 (en) 2015-06-30
JP2021101706A (en) 2021-07-15
US20160298138A1 (en) 2016-10-13
KR102531576B1 (en) 2023-05-11
IL257178B (en) 2019-07-31
LT3363902T (en) 2020-02-10
PL3138911T3 (en) 2019-04-30
PL3363902T3 (en) 2020-05-18
EP3138912A1 (en) 2017-03-08
BR112015012375A2 (en) 2017-09-26
JP2022115994A (en) 2022-08-09
CA2891347C (en) 2018-02-27
AU2017204031B2 (en) 2018-06-14
PL2928496T3 (en) 2020-04-30
AU2020230243B2 (en) 2021-10-21
JP6620018B2 (en) 2019-12-11
CN108913676A (en) 2018-11-30
US20160298135A1 (en) 2016-10-13
PT3138911T (en) 2018-12-28
US20160298132A1 (en) 2016-10-13
KR20230003624A (en) 2023-01-06
EP3611263A1 (en) 2020-02-19
IL267598B (en) 2022-05-01
PT3138912T (en) 2018-12-28
EP3617309A2 (en) 2020-03-04
US20160298137A1 (en) 2016-10-13
AU2020230246B2 (en) 2020-11-05
ES2757325T3 (en) 2020-04-28
KR20180011351A (en) 2018-01-31
US20160298125A1 (en) 2016-10-13
AU2018229489B2 (en) 2018-12-06
US20160017366A1 (en) 2016-01-21
EP3360964B1 (en) 2019-10-02
KR20150091052A (en) 2015-08-07
AU2023216829A1 (en) 2023-09-14
AU2019201344B2 (en) 2020-09-03
DK3138911T3 (en) 2019-01-21
AU2022200330B2 (en) 2023-11-09
DK3360964T3 (en) 2019-10-28
EP2928496A1 (en) 2015-10-14
AU2018229489A1 (en) 2018-10-04
US20210388396A1 (en) 2021-12-16
AU2022200330A1 (en) 2022-02-17
KR20210045515A (en) 2021-04-26
KR102145760B1 (en) 2020-08-19
EP3360964A1 (en) 2018-08-15
DK2928496T3 (en) 2019-11-11
SG10201800585VA (en) 2018-02-27
KR102479178B1 (en) 2022-12-19
IL300199A (en) 2023-03-01
ES2714154T3 (en) 2019-05-27
SG11201503824SA (en) 2015-06-29
CA2891347A1 (en) 2014-06-12
JP2019037231A (en) 2019-03-14
US10731181B2 (en) 2020-08-04
PT2928496T (en) 2019-11-11
AU2019201344C1 (en) 2020-12-24
AU2020273316A1 (en) 2020-12-17
AU2020230243A1 (en) 2020-10-01
US20170191082A1 (en) 2017-07-06
JP2017192392A (en) 2017-10-26
CA2977152A1 (en) 2014-06-12
DK3138912T3 (en) 2019-01-21
EP3363902B1 (en) 2019-11-27
ES2713243T3 (en) 2019-05-20
EP3138912B1 (en) 2018-12-05
KR101844123B1 (en) 2018-04-02
EP3363902A1 (en) 2018-08-22
IL267598A (en) 2019-08-29
LT3138910T (en) 2017-11-10
ES2757808T3 (en) 2020-04-30
PL3138910T3 (en) 2018-01-31
CN108715602A (en) 2018-10-30
EP2928496B1 (en) 2019-10-09
CN105142669A (en) 2015-12-09
US10745716B2 (en) 2020-08-18
KR20200098727A (en) 2020-08-20
JP2016502840A (en) 2016-02-01
CA3034794A1 (en) 2014-06-12
LT3138912T (en) 2019-02-25
AU2013355214B2 (en) 2017-06-15
US20200140897A1 (en) 2020-05-07
EP3617309A3 (en) 2020-05-06
ES2653212T3 (en) 2018-02-06
AU2017204031A1 (en) 2017-07-06
US20160298134A1 (en) 2016-10-13
IL291129B2 (en) 2023-07-01
PT3138910T (en) 2017-10-18
US20160298136A1 (en) 2016-10-13
DK3363902T3 (en) 2020-01-02
HK1218389A1 (en) 2017-02-17
KR102006880B1 (en) 2019-08-02
KR20190093680A (en) 2019-08-09
AU2013355214A1 (en) 2015-06-04
EP2928496A4 (en) 2017-03-01
EP3135765A1 (en) 2017-03-01
AU2020230246A1 (en) 2020-10-01
CN108913676B (en) 2023-11-03

Similar Documents

Publication Publication Date Title
AU2019201344B2 (en) Crispr-based genome modification and regulation

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201380072477.4

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13859964

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2891347

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 238856

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 20157013843

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2013355214

Country of ref document: AU

Date of ref document: 20131205

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015545838

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015012375

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 2013859964

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 112015012375

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20150528