US20160186152A1 - Modified cascade ribonucleoproteins and uses thereof - Google Patents

Modified cascade ribonucleoproteins and uses thereof Download PDF

Info

Publication number
US20160186152A1
US20160186152A1 US14/997,467 US201614997467A US2016186152A1 US 20160186152 A1 US20160186152 A1 US 20160186152A1 US 201614997467 A US201614997467 A US 201614997467A US 2016186152 A1 US2016186152 A1 US 2016186152A1
Authority
US
United States
Prior art keywords
cascade
sequence
crispr
target
complex
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/997,467
Inventor
Stan Johan Jozef Brouns
John Van Der Oost
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wageningen Universiteit
Caribou Biosciences Inc
Original Assignee
Caribou Biosciences Inc
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
Application filed by Caribou Biosciences Inc filed Critical Caribou Biosciences Inc
Priority to US14/997,467 priority Critical patent/US20160186152A1/en
Publication of US20160186152A1 publication Critical patent/US20160186152A1/en
Assigned to WAGENINGEN UNIVERSITEIT reassignment WAGENINGEN UNIVERSITEIT ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BROUNS, Stan Johan Jozef, VAN DER OOST, JOHN
Assigned to CARIBOU BIOSCIENCES, INC. reassignment CARIBOU BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WAGENINGEN UNIVERSITEIT
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/24Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Enterobacteriaceae (F), e.g. Citrobacter, Serratia, Proteus, Providencia, Morganella, Yersinia
    • C07K14/245Escherichia (G)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • 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
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • C12N15/66General methods for inserting a gene into a vector to form a recombinant vector using cleavage and ligation; Use of non-functional linkers or adaptors, e.g. linkers containing the sequence for a restriction endonuclease
    • 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
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • 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
    • C12N15/74Vectors or expression systems specially adapted for prokaryotic hosts other than E. coli, e.g. Lactobacillus, Micromonospora
    • 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
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • 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
    • C12N15/82Vectors or expression systems specially adapted for eukaryotic hosts for plant cells, e.g. plant artificial chromosomes (PACs)
    • 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
    • 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
    • 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
    • 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)
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/22Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a Strep-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/60Fusion polypeptide containing spectroscopic/fluorescent detection, e.g. green fluorescent protein [GFP]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/71Fusion polypeptide containing domain for protein-protein interaction containing domain for transcriptional activaation, e.g. VP16
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/80Fusion polypeptide containing a DNA binding domain, e.g. Lacl or Tet-repressor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/85Fusion polypeptide containing an RNA binding domain
    • 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
    • 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)

Definitions

  • the additional amino acid sequence with nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity may, as desired, be obtained or derived from the same organism, e.g. E. coli , as the Cas or Cse subunit(s).
  • a Cascade complex of the invention is a Type I CRISPR-Cas system protein complex; more preferably a subtype I-E CRISPR-Cas protein complex or it can be based on a Type I-A or Type I-B complex.
  • a Type I-C, D or F complex is possible.
  • the subunits may have the following stoichiometries: Cse1 1 Cse2 2 Cas7 6 Cas5 1 Cas6 1 or Cse1 1 Cse2 2 Cas7 6 Cas5 1 Cas6e 1 .
  • the additional amino acid sequence having nucleic acid or chromatin modifying activity is fused or linked to the N terminus or the C terminus of a Cse1, a Cse2 or a Cas5 subunit; more preferably the linkage is in the region of the N terminus of a Cse1 subunit, the N terminus of a Cse2 subunit, or the N terminus of a Cas7 subunit.
  • FIG. 10 shows sequence alignments of cas3 genes from organisms containing the Type I-E CRISPR/Cas system. Alignment of cas3-cse1 genes from Streptomyces sp. SPB78 (1 st sequence, Accession Number: ZP_07272643.1) [SEQ ID NO: 43], in Streptomyces griseus (2 nd sequence, Accession Number YP_001825054) [SEQ ID NO: 44], and in Catenulispora acidiphila DSM 44928 (3 rd sequence, Accession Number YP_003114638) [SEQ ID NO: 45] and an artificial E. coli Cas3-Cse1 fusion protein [SEQ ID NO: 46] which includes the polypeptide linker sequence from S. griseus.
  • crRNA is obtained by injecting purified intact Cascade at 75° C.
  • Cascade is very stable as a multi-subunit protein-RNA complex and is easily produced in mg quantities in E. coli .
  • Transfection or micro-injection of the complex in its intact form as purified from E. coli is used as methods of delivery (see FIG. 12 ).
  • Cascade-FokI nucleases are purified from E. coli and encapsulated in protein transfection vesicles. These are then fused with the cell membrane of human HepG2 cells releasing the nucleases in the cytoplasm (step 2). NLS sequences are then be recognized by importin proteins, which facilitate nucleopore passage (step 3).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cell Biology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A clustered regularly interspaced short palindromic repeat (CRISPR)-associated complex for adaptive antiviral defence (Cascade); the Cascade protein complex comprising at least CRISPR-associated protein subunits Cas7, Cas5 and Cas6 which includes at least one subunit with an additional amino acid sequence possessing nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity. The Cascade complex with additional activity is combined with an RNA molecule to produce a ribonucleoprotein complex. The RNA molecule is selected to have substantial complementarity to a target sequence. Targeted ribonucleoproteins can be used as genetic engineering tools for precise cutting of nucleic acids in homologous recombination, non-homologous end joining, gene modification, gene integration, mutation repair or for their visualisation, transcriptional activation or repression. A pair of ribonucleotides fused to FokI dimers may be used to generate double-strand breakages in the DNA to facilitate these applications in a sequence-specific manner.

Description

  • This application is a continuation of U.S. patent application Ser. No. 14/326,099, filed 8 Jul. 2014, now abandoned, which is a continuation of U.S. patent application Ser. No. 14/240,735, filed 24 Feb. 2014, now pending, which is a National Stage Entry of PCT/EP2012/076674, filed 21 Dec. 2012, now expired, which claims the benefit of priority under 35 U.S.C. 119(a)/(b) of United Kingdom Patent Application No. GB 1122458.1, filed 30 Dec. 2011, now expired, the contents of all of which are herein incorporated by reference in their entireties.
  • The present application contains a Sequence Listing that has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. The ASCII copy, created on 15 Jan. 2016, is named CBI010-13 ST25.txt and is 119 kb in size.
  • The invention relates to the field of genetic engineering and more particularly to the area of gene and/or genome modification of organisms, including prokaryotes and eukaryotes. The invention also concerns methods of making site specific tools for use in methods of genome analysis and genetic modification, whether in vivo or in vitro. The invention more particularly relates to the field of ribonucleoproteins which recognise and associate with nucleic acid sequences in a sequence specific way.
  • Bacteria and archaea have a wide variety of defense mechanisms against invasive DNA. So called CRISPR/Cas defense systems provide adaptive immunity by integrating plasmid and viral DNA fragments in loci of clustered regularly interspaced short palindromic repeats (CRISPR) on the host chromosome. The viral or plasmid-derived sequences, known as spacers, are separated from each other by repeating host-derived sequences. These repetitive elements are the genetic memory of this immune system and each CRISPR locus contains a diverse repertoire of unique ‘spacer’ sequences acquired during previous encounters with foreign genetic elements.
  • Acquisition of foreign DNA is the first step of immunization, but protection requires that the CRISPR is transcribed and that these long transcripts are processed into short CRISPR-derived RNAs (crRNAs) that each contains a unique spacer sequence complementary to a foreign nucleic acid challenger.
  • In addition to the crRNA, genetic experiments in several organisms have revealed that a unique set of CRISPR-associated (Cas) proteins is required for the steps of acquiring immunity, for crRNA biogenesis and for targeted interference. Also, a subset of Cas proteins from phylogenetically distinct CRISPR systems have been shown to assemble into large complexes that include a crRNA.
  • A recent re-evaluation of the diversity of CRISPR/Cas systems has resulted in a classification into three distinct types (Makarova K. et al (2011) Nature Reviews Microbiology—AOP 9 May 2011; doi:10.1038/nrmicro2577) that vary in cas gene content, and display major differences throughout the CRISPR defense pathway. (The Makarova classification and nomenclature for CRISPR-associated genes is adopted in the present specification.) RNA transcripts of CRISPR loci (pre-crRNA) are cleaved specifically in the repeat sequences by CRISPR associated (Cas) endoribonucleases in type I and type III systems or by RNase III in type II systems; the generated crRNAs are utilized by a Cas protein complex as a guide RNA to detect complementary sequences of either invading DNA or RNA. Cleavage of target nucleic acids has been demonstrated in vitro for the Pyrococcus furiosus type III-B system, which cleaves RNA in a ruler-anchored mechanism, and, more recently, in vivo for the Streptococcus thermophiles type II system, which cleaves DNA in the complementary target sequence (protospacer). In contrast, for type I systems the mechanism of CRISPR-interference is still largely unknown.
  • The model organism Escherichia coli strain K12 possesses a CRISPR/Cas type I-E (previously known as CRISPR subtype E (Cse)). It contains eight cas genes (cas1, cas2, cas3 and cse1, cse2, cas7, cas5, cas6e) and a downstream CRISPR (type-2 repeats). In Escherichia coli K12 the eight cas genes are encoded upstream of the CRISPR locus. Cas1 and Cas2 do not appear to be needed for target interference, but are likely to participate in new target sequence acquisition. In contrast, six Cas proteins: Cse1, Cse2, Cas3, Cas7, Cas5 and Cas6e (previously also known as CasA, CasB, Cas3, CasC/Cse4, CasD and CasE/Cse3 respectively) are essential for protection against lambda phage challenge. Five of these proteins: Cse1, Cse2, Cas7, Cas5 and Cas6e (previously known as CasA, CasB, CasC/Cse4, CasD and CasE/Cse3 respectively) assemble with a crRNA to form a multi-subunit ribonucleoprotein (RNP) referred to as Cascade.
  • In E. coli, Cascade is a 405 kDa ribonucleoprotein complex composed of an unequal stoichiometry of five functionally essential Cas proteins: Cse11Cse22Cas76Cas51Cas6e1 (i.e. under previous nomenclature CasA1B2C6D1E1) and a 61-nt CRISPR-derived RNA. Cascade is an obligate RNP that relies on the crRNA for complex assembly and stability, and for the identification of invading nucleic acid sequences. Cascade is a surveillance complex that finds and binds foreign nucleic acids that are complementary to the spacer sequence of the crRNA.
  • Jore et al. (2011) entitled “Structural basis for CRISPR RNA-guided DNA recognition by Cascade” Nature Structural & Molecular Biology 18: 529-537 describes how there is a cleavage of the pre-crRNA transcript by the Cas6e subunit of Cascade, resulting in the mature 61 nt crRNA being retained by the CRISPR complex. The crRNA serves as a guide RNA for sequence specific binding of Cascade to double stranded (ds) DNA molecules through base pairing between the crRNA spacer and the complementary protospacer, forming a so-called R-loop. This is known to be an ATP-independent process.
  • Brouns S. J. J., et al (2008) entitled “Small CRISPR RNAs guide antiviral defense in prokaryotes” Science 321: 960-964 teaches that Cascade loaded with a crRNA requires Cas3 for in vivo phage resistance.
  • Marraffini L. & Sontheimer E. (2010) entitled “CRISPR interference: RNA-directed adaptive immunity in bacteria and archaea” Nature Reviews Genetics 11: 181-190 is a review article which summarises the state of knowledge in the art in the field. Some suggestions are made about CRISPR-based applications and technologies, but this is mainly in the area of generating phage resistant strains of domesticated bacteria for the dairy industry. The specific cleavage of RNA molecules in vitro by a crRNP complex in Pyrococcus furiosus is suggested as something which awaits further development. Manipulation of CRISPR systems is also suggested as a possible way of reducing transmission of antibiotic-resistant bacterial strains in hospitals. The authors stress that further research effort will be needed to explore the potential utility of the technology in these areas.
  • US2011236530 A1 (Manoury et al.) entitled “Genetic cluster of strains of Streptococcus thermophilus having unique rheological properties for dairy fermentation” discloses certain S. thermophilus strains which ferment milk so that it is highly viscous and weakly ropy. A specific CRISPR locus of defined sequence is disclosed.
  • US2011217739 A1 (Terns et al.) entitled “Cas6 polypeptides and methods of use” discloses polypeptides which have Cas6 endoribonuclease activity. The polypeptides cleave a target RNA polynucleotide having a Cas6 recognition domain and cleavage site. Cleavage may be carried out in vitro or in vivo. Microbes such as E. coli or Haloferax volcanii are genetically modified so as to express Cas6 endoribonuclease activity.
  • WO2010054154 (Danisco) entitled “Bifidobacteria CRISPR sequences” discloses various CRISPR sequences found in Bifidobacteria and their use in making genetically altered strains of the bacteria which are altered in their phage resistance characteristics.
  • US2011189776 A1 (Terns et al.) entitled “Prokaryotic RNAi-like system and methods of use” describes methods of inactivating target polynucleotides in vitro or in prokaryotic microbes in vivo. The methods use a psiRNA having a 5′ region of 5-10 nucleotides chosen from a repeat from a CRISPR locus immediately upstream of a spacer. The 3′ region is substantially complementary to a portion of the target polynucleotide. Also described are polypeptides having endonuclease activity in the presence of psiRNA and target polynucleotide.
  • EP2341149 A1 (Danisco) entitled “Use of CRISPR associated genes (CAS) describes how one or more Cas genes can be used for modulating resistance of bacterial cells against bacteriophage; particularly bacteria which provide a starter culture or probiotic culture in dairy products.
  • WO2010075424 (The Regents of the University of California) entitled “Compositions and methods for downregulating prokaryotic genes” discloses an isolated polynucleotide comprising a CRISPR array. At least one spacer of the CRISPR is complementary to a gene of a prokaryote so that is can down-regulate expression of the gene; particularly where the gene is associated with biofuel production.
  • WO2008108989 (Danisco) entitled “Cultures with improved phage resistance” discloses selecting bacteriophage resistant strains of bacteria and also selecting the strains which have an additional spacer having 100% identity with a region of phage RNA. Improved strain combinations and starter culture rotations are described for use in the dairy industry. Certain phages are described for use as biocontrol agents.
  • WO2009115861 (Institut Pasteur) entitled “Molecular typing and subtyping of Salmonella by identification of the variable nucleotide sequences of the CRISPR loci” discloses methods for detecting and identifying bacterial of the Salmonella genus by using their variable nucleotide sequences contained in CRISPR loci.
  • WO2006073445 (Danisco) entitled “Detection and typing of bacterial strains” describes detecting and typing of bacterial strains in food products, dietary supplements and environmental samples. Strains of Lactobacillus are identified through specific CRISPR nucleotide sequences.
  • Urnov F et al. (2010) entitled “Genome editing with engineered zinc finger nucleases” Nature 11: 636-646 is a review article about zinc finger nucleases and how they have been instrumental in the field of reverse genetics in a range of model organisms. Zinc finger nucleases have been developed so that precisely targeting genome cleavage is possible followed by gene modification in the subsequent repair process. However, zinc finger nucleases are generated by fusing a number of zinc finger DNA-binding domains to a DNA cleavage domain. DNA sequence specificity is achieved by coupling several zinc fingers in series, each recognising a three nucleotide motif. A significant drawback with the technology is that new zinc fingers need to be developed for each new DNA locus which requires to be cleaved. This requires protein engineering and extensive screening to ensure specificity of DNA binding.
  • In the fields of genetic engineering and genomic research there is an ongoing need for improved agents for sequence/site specific nucleic acid detection and/or cleavage.
  • The inventors have made a surprising discovery in that certain bacteria expressing Cas3, which has helicase-nuclease activity, express Cas3 as a fusion with Cse1. The inventors have also unexpectedly been able to produce artificial fusions of Cse1 with other nuclease enzymes.
  • The inventors have also discovered that Cas3-independent target DNA recognition by Cascade marks DNA for cleavage by Cas3, and that Cascade DNA binding is governed by topological requirements of the target DNA.
  • The inventors have further found that Cascade is unable to bind relaxed target plasmids, but surprisingly Cascade displays high affinity for targets which have a negatively supercoiled (nSC) topology.
  • Accordingly in a first aspect the present invention provides a clustered regularly interspaced short palindromic repeat (CRISPR)-associated complex for antiviral defence (Cascade), the Cascade protein complex, or portion thereof, comprising at least CRISPR-associated protein subunits:
      • Cas7 (or COG 1857) having an amino acid sequence of SEQ ID NO:3 or a sequence of at least 18% identity therewith,
      • Cas5 (or COG1688) having an amino acid sequence of SEQ ID NO:4 or a sequence of at least 17% identity therewith, and
      • Cas6 (or COG 1583) having an amino acid sequence of SEQ ID NO:5 or a sequence of at least 16% identity therewith;
        and wherein at least one of the subunits includes an additional amino acid sequence providing nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity.
  • A subunit which includes an additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity is an example of what may be termed “a subunit linked to at least one functional moiety”; a functional moiety being the polypeptide or protein made up of the additional amino acid sequence. The transcription activating activity may be that leading to activation or upregulation of a desired genes; the transcription repressing activity leading to repressing or downregulation of a desired genes. The selection of the gene being due to the targeting of the cascade complex of the invention with an RNA molecule, as described further below.
  • The additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity is preferably formed of contiguous amino acid residues. These additional amino acids may be viewed as a polypeptide or protein which is contiguous and forms part of the Cas or Cse subunit(s) concerned. Such a polypeptide or protein sequence is preferably not normally part of any Cas or Cse subunit amino acid sequence. In other words, the additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity may be other than a Cas or Cse subunit amino acid sequence, or portion thereof, i.e. may be other than a Cas3 submit amino acid sequence or portion thereof.
  • The additional amino acid sequence with nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity may, as desired, be obtained or derived from the same organism, e.g. E. coli, as the Cas or Cse subunit(s).
  • Additionally and/or alternatively to the above, the additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity may be “heterologous” to the amino acid sequence of the Cas or Cse subunit(s). Therefore, the additional amino acid sequence may be obtained or derived from an organism different from the organism from which the Cas and/or Cse subunit(s) are derived or originate.
  • Throughout, sequence identity may be determined by way of BLAST and subsequent Cobalt multiple sequence alignment at the National Center for Biotechnology Information webserver, where the sequence in question is compared to a reference sequence (e.g. SEQ ID NO: 3, 4 or 5). The amino acid sequences may be defined in terms of percentage sequence similarity based on a BLOSUM62 matrix or percentage identity with a given reference sequence (e.g. SEQ ID NO:3, 4 or 5). The similarity or identity of a sequence involves an initial step of making the best alignment before calculating the percentage conservation with the reference and reflects a measure of evolutionary relationship of sequences.
  • Cas7 may have a sequence similarity of at least 31% with SEQ ID NO:3; Cas5 may have a sequence similarity of at least 26% with SEQ ID NO:4. Cas6 may have a sequence similarity of at least 27% with SEQ ID NO:5.
  • For Cse1/CasA(502 AA):
    >gi|16130667|ref|NP_417240.1| CRISP RNA (crRNA)
    containing Cascade antiviral complex protein
    [Escherichia coli str. K-12 sub str. MG1655]
    [SEQ ID NO: 1]
    MNLLIDNWIPVRPRNGGKVQIINLQSLYCSRDQWRLSLPRDDMELAALA
    LLVCIGQIIAPAKDDVEFRHRIMNPLTEDEFQQLIAPWIDMFYLNHAEH
    PFMQTKGVKANDVTPMEKLLAGVSGATNCAFVNQPGQGEALCGGCTAIA
    LFNQANQAPGFGGGFKSGLRGGTPVTTFVRGIDLRSTVLLNVLTLPRLQ
    KQFPNESHTENQPTWIKPIKSNESIPASSIGFVRGLFWQPAHIELCDPI
    GIGKCSCCGQESNLRYTGFLKEKFTFTVNGLWPHPHSPCLVTVKKGEVE
    EKFLAFTTSAPSWTQISRVVVDKIIQNENGNRVAAVVNQFRNIAPQSPL
    ELIMGGYRNNQASILERRHDVLMFNQGWQQYGNVINEIVTVGLGYKTAL
    RKALYTFAEGFKNKDFKGAGVSVHETAERHFYRQSELLIPDVLANVNFS
    QADEVIADLRDKLHQLCEMLFNQSVAPYAHHPKLISTLALARATLYKHL
    RELKPQGGPSNG
    For Cse2/CasB (160 AA):
    >gi|16130666|ref|NP_417239.1| CRISP RNA (crRNA)
    containing Cascade antiviral complex protein
    [Escherichia coli str. K-12 substr. MG1655]
    [SEQ ID NO: 2]
    MADEIDAMALYRAWQQLDNGSCAQIRRVSEPDELRDIPAFYRLVQPFGW
    ENPRHQQALLRMVFCLSAGKNVIRHQDKKSEQTTGISLGRALANSGRIN
    ERRIFQLIRADRTADMVQLRRLLTHAEPVLDWPLMARMLTWWGKRERQQ
    LLEDFVLTTNKNA
    For Cas7/CasC/Cse4 (363 AA):
    >gi|16130665|ref|NP_417238.11 CRISP RNA (crRNA)
    containing Cascade antiviral complex protein
    [Escherichia coli str. K-12 substr. MG1655]
    [SEQ ID NO: 3]
    MSNFINIHVLISHSPSCLNRDDMNMQKDAIFGGKRRVRISSQSLKRAMR
    KSGYYAQNIGESSLRTIHLAQLRDVLRQKLGERFDQKIIDKTLALLSGK
    SVDEAEKISADAVTPWVVGEIAWFCEQVAKAEADNLDDKKLLKVLKEDI
    AAIRVNLQQGVDIALSGRMATSGMMTELGKVDGAMSIAHAITTHQVDSD
    IDWFTAVDDLQEQGSAHLGTQEFSSGVFYRYANINLAQLQENLGGASRE
    QALEIATHVVHMLATEVPGAKQRTYAAFNPADMVMVNFSDMPLSMANAF
    EKAVKAKDGFLQPSIQAFNQYWDRVANGYGLNGAAAQFSLSDVDPITAQ
    VKQMPTLEQLKSWVRNNGEA
    For Cas5/CasD(224 AA):
    >gi|90111483|ref|NP_417237.2| CRISP RNA (crRNA)
    containing Cascade antiviral complex protein
    [Escherichia coli str. K-12 substr. MG1655]
    [SEQ ID NO: 4]
    MRSYLILRLAGPMQAWGQPTFEGTRPTGRFPTRSGLLGLLGACLGIQRD
    DTSSLQALSESVQFAVRCDELILDDRRVSVTGLRDYHTVLGAREDYRGL
    KSHETIQTWREYLCDASFTVALWLTPHATMVISELEKAVLKPRYTPYLG
    RRSCPLTHPLFLGTCQASDPQKALLNYEPVGGDIYSEESVTGHHLKFTA
    RDEPMITLPRQFASREWYVIKGGMDVSQ
    For Cas6e/CasE(199 AA):
    >gi|16130663|ref|NP_417236.1| CRISPR RNA
    precursor cleavage enzyme; CRISP RNA (crRNA)
    containing Cascade antiviral complex protein
    [Escherichia coli str. K-12 substr. MG1655]
    [SEQ ID NO: 5]
    MYLSKVIIARAWSRDLYQLHQGLWHLFPNRPDAARDFLFHVEKRNTPEG
    CHVLLQSAQMPVSTAVATVIKTKQVEFQLQVGVPLYFRLRANPIKTILD
    NQKRLDSKGNIKRCRVPLIKEAEQIAWLQRKLGNAARVEDVHPISERPQ
    YFSGDGKSGKIQTVCFEGVLTINDAPALIDLVQQGIGPAKSMGCGLLSL
    APL
  • In defining the range of sequence variants which fall within the scope of the invention, for the avoidance of doubt, the following are each optional limits on the extent of variation, to be applied for each of SEQ ID NO:1, 2, 3, 4 or 5 starting from the respect broadest range of variants as specified in terms of the respective percentage identity above. The range of variants therefore may therefore include: at least 16%, or at least 17%, or at least 18%, or at least 19%, or at least 20%, or at least 21%, or at least 22%, or at least 23%, or at least 24%, or at least 25%, or at least 26%, or at least 27%, or at least 28%, or at least 29%, or at least 30%, or at least 31%, or at least 32%, or at least 33%, or at least 34%, or at least 35%, or at least 36%, or at least 37%, or at least 38%, or at least 39%, or at least 40%, or at least 41%, or at least 42%, or at least 43%, at least 44%, or at least 45%, or at least 46%, or at least 47%, or at least 48%, or at least 49%, or at least 50%, or at least 51%, or at least 52%, or at least 53%, or at least 54%, or at least 55%, or at least 56%, or at least 57%, or at least 58%, or at least 59%, or at least 60%, or at least 61%, or at least 62%, or at least 63%, or at least 64%, or at least 65%, or at least 66%, or at least 67%, or at least 68%, or at least 69%, or at least 70%, or at least 71%, at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least 79%, or at least 80%, or at least 81%, or at least 82%, or at least 83%, or at least 84%, or at least 85%, or at least 86%, or at least 87%, or at least 88%, or at least 89%, or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99%, or 100% amino acid sequence identity.
  • Throughout, the Makarova et al. (2011) nomenclature is being used in the definition of the Cas protein subunits. Table 2 on page 5 of the Makarova et al. article lists the Cas genes and the names of the families and superfamilies to which they belong. Throughout, reference to a Cas protein or Cse protein subunit includes cross reference to the family or superfamily of which these subunits form part.
  • Throughout, the reference sequences of the Cas and Cse subunits of the invention may be defined as a nucleotide sequence encoding the amino acid sequence. For example, the amino acid sequence of SEQ ID NO:3 for Cas7 also includes all nucleic acid sequences which encode that amino acid sequence. The variants of Cas7 included within the scope of the invention therefore include nucleotide sequences of at least the defined amino acid percentage identities or similarities with the reference nucleic acid sequence; as well as all possible percentage identities or similarities between that lower limit and 100%.
  • The Cascade complexes of the invention may be made up of subunits derived or modified from more than one different bacterial or archaeal prokaryote. Also, the subunits from different Cas subtypes may be mixed.
  • In a preferred aspect, the Cas6 subunit is a Cas6e subunit of SEQ ID NO: 17 below, or a sequence of at least 16% identity therewith.
  • The sequence of a preferred Cas6e subunit is
    >gi|16130663|ref|NP_417236.1| CRISPR RNA
    precursor cleavage enzyme; CRISP RNA (crRNA)
    containing Cascade antiviral complex protein
    [Escherichia coli str. K-12 substr. MG1655]:
    [SEQ ID NO: 17]
    MYLSKVIIARAWSRDLYQLHQGLWHLFPNRPDAARDFLFHVEKRNTPEG
    CHVLLQSAQMPVSTAVATVIKTKQVEFQLQVGVPLYFRLRANPIKTILD
    NQKRLDSKGNIKRCRVPLIKEAEQIAWLQRKLGNAARVEDVHPISERPQ
    YFSGDGKSGKIQTVCFEGVLTINDAPALIDLVQQGIGPAKSMGCGLLSL
    APL
  • The Cascade complexes, or portions thereof, of the invention—which comprise at least one subunit which includes an additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity—may further comprise a Cse2 (or YgcK-like) subunit having an amino acid sequence of SEQ ID NO:2 or a sequence of at least 20% identity therewith, or a portion thereof. Alternatively, the Cse subunit is defined as having at least 38% similarity with SEQ ID NO:2. Optionally, within the protein complex of the invention it is the Cse2 subunit which includes the additional amino acid sequence having nucleic acid or chromatin modifying activity.
  • Additionally or alternatively, the Cascade complexes of the invention may further comprise a Cse1 (or YgcL-like) subunit having an amino acid sequence of SEQ ID NO: 1 or a sequence of at least 9% identity therewith, or a portion thereof. Optionally within the protein complex of the invention it is the Cse1 subunit which includes the additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity.
  • In preferred embodiments, a Cascade complex of the invention is a Type I CRISPR-Cas system protein complex; more preferably a subtype I-E CRISPR-Cas protein complex or it can be based on a Type I-A or Type I-B complex. A Type I-C, D or F complex is possible. In particularly preferred embodiments based on the E. coli system, the subunits may have the following stoichiometries: Cse11Cse22Cas76Cas51 Cas61 or Cse11Cse22Cas76Cas51Cas6e1.
  • The additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity may be translationally fused through expression in natural or artificial protein expression systems, or covalently linked by a chemical synthesis step to the at least one subunit; preferably the at least one functional moiety is fused or linked to at least the region of the N terminus and/or the region of the C terminus of at least one of a Cse1, Cse2, Cas7, Cas5, Cas6 or Cas6e subunit. In particularly preferred embodiments, the additional amino acid sequence having nucleic acid or chromatin modifying activity is fused or linked to the N terminus or the C terminus of a Cse1, a Cse2 or a Cas5 subunit; more preferably the linkage is in the region of the N terminus of a Cse1 subunit, the N terminus of a Cse2 subunit, or the N terminus of a Cas7 subunit.
  • The additional amino acid sequence having nucleic acid or chromatin modifying, activating, repressing or visualising activity may be a protein; optionally selected from a helicase, a nuclease, a nuclease-helicase, a DNA methyltransferase (e.g. Dam), or DNA demethylase, a histone methyltransferase, a histone demethylase, an acetylase, a deacetylase, a phosphatase, a kinase, a transcription (co-)activator, an RNA polymerase submit, a transcription repressor, a DNA binding protein, a DNA structuring protein, a marker protein, a reporter protein, a fluorescent protein, a ligand binding protein (e.g. mCherry or a heavy metal binding protein), a signal peptide (e.g. Tat-signal sequence), a subcellular localisation sequence (e.g. nuclear localisation sequence) or an antibody epitope.
  • The protein concerned may be a heterologous protein from a species other than the bacterial species from which the Cascade protein subunits have their sequence origin.
  • When the protein is a nuclease, it may be one selected from a type II restriction endonuclease such as FokI, or a mutant or an active portion thereof. Other type II restriction endonucleases which may be used include EcoR1, EcoRV, BgII, BamHI, BsgI and BspMI. Preferably, one protein complex of the invention may be fused to the N terminal domain of FokI and another protein complex of the invention may be fused to the C terminal domain of FokI. These two protein complexes may then be used together to achieve an advantageous locus specific double stranded cut in a nucleic acid, whereby the location of the cut in the genetic material is at the design and choice of the user, as guided by the RNA component (defined and described below) and due to presence of a so-called “protospacer adjacent motif” (PAM) sequence in the target nucleic acid strand (also described in more detail below).
  • In a preferred embodiment, a protein complex of the invention has an additional amino acid sequence which is a modified restriction endonuclease, e.g. FokI. The modification is preferably in the catalytic domain. In preferred embodiments, the modified FokI is KKR Sharkey or ELD Sharkey which is fused to the Cse1 protein of the protein complex. In a preferred application of these complexes of the invention, two of these complexes (KKR Sharkey and ELD Sharkey) may be together in combination. A heterodimer pair of protein complexes employing differently modified FokI is has particular advantage in targeted double stranded cutting of nucleic acid. If homodimers are used then it is possible that there is more cleavage at non-target sites due to non-specific activity. A heterodimer approach advantageously increases the fidelity of the cleavage in a sample of material.
  • The Cascade complex with additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity defined and described above is a component part of an overall system of the invention which advantageously permits the user to select in a predetermined matter a precise genetic locus which is desired to be cleaved, tagged or otherwise altered in some way, e.g methylation, using any of the nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing entities defined herein. The other component part of the system is an RNA molecule which acts as a guide for directing the Cascade complex of the invention to the correct locus on DNA or RNA intending to be modified, cut or tagged.
  • The Cascade complex of the invention preferably also comprises an RNA molecule which comprises a ribonucleotide sequence of at least 50% identity to a desired target nucleic acid sequence, and wherein the protein complex and the RNA molecule form a ribonucleoprotein complex. Preferably the ribonucleoprotein complex forms when the RNA molecule is hybridized to its intended target nucleic acid sequence. The ribonucleoprotein complex forms when the necessary components of Cascade-functional moiety combination and RNA molecule and nucleic acid (DNA or RNA) are present together in suitable physiological conditions, whether in vivo or in vitro. Without wishing to be bound by any particular theory, the inventors believe that in the context of dsDNA, particularly negatively supercoiled DNA, the Cascade complex associating with the dsDNA causes a partial unwinding of the duplex strands which then allows the RNA to associate with one strand; the whole ribonucleoprotein complex then migrates along the DNA strand until a target sequence substantially complementary to at least a portion of the RNA sequence is reached, at which point a stable interaction between RNA and DNA strand occurs, and the function of the functional moiety takes effect, whether by modifying, nuclease cutting or tagging of the DNA at that locus.
  • In preferred embodiments, a portion of the RNA molecule has at least 50% identity to the target nucleic acid sequence; more preferably at least 95% identity to the target sequence. In more preferred embodiments, the portion of the RNA molecule is substantially complementary along its length to the target DNA sequence; i.e. there is only one, two, three, four or five mismatches which may be contiguous or non-contiguous. The RNA molecule (or portion thereof) may have at least 51%, or at least 52%, or at least 53%, or at least 54%, or at least 55%, or at least 56%, or at least 57%, or at least 58%, or at least 59%, or at least 60%, or at least 61%, or at least 62%, or at least 63%, or at least 64%, or least 65%, or at least 66%, or at least 67%, or at least 68%, or at least 69%, or at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least 79%, or at least 80%, or at least 81%, or at least 82%, or at least 83%, or at least 84%, or least 85%, or at least 86%, or at least 87%, or at least 88%, or at least 89%, or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99%, or 100% identity to the target sequence.
  • The target nucleic acid may be DNA (ss or ds) or RNA.
  • In other preferred embodiments, the RNA molecule or portion thereof has at least 70% identity with the target nucleic acid. At such levels of identity, the target nucleic acid is preferably dsDNA.
  • The RNA molecule will preferably require a high specificity and affinity for the target nucleic acid sequence. A dissociation constant (Kd) in the range 1 pM to 1 μM, preferably 1-100 nM is desirable as determined by preferably native gel electrophoresis, or alternatively isothermal titration calorimetry, surface plasmon resonance, or fluorescence based titration methods. Affinity may be determined using an electrophoretic mobility shift assay (EMSA), also called gel retardation assay (see Semenova E et al. (2011) Proc. Natl. Acad. Sci. USA 108: 10098-10103).
  • The RNA molecule is preferably modelled on what are known from nature in prokaryotes as CRISPR RNA (crRNA) molecules. The structure of crRNA molecules is already established and explained in more detail in Jore et al. (2011) Nature Structural & Molecular Biology 18: 529-537. In brief, a mature crRNA of type I-E is often 61 nucleotides long and consists of a 5′ “handle” region of 8 nucleotides, the “spacer” sequence of 32 nucleotides, and a 3′ sequence of 21 nucleotides which form a hairpin with a tetranucleotide loop. However, the RNA used in the invention does not have to be designed strictly to the design of naturally occurring crRNA, whether in length, regions or specific RNA sequences. What is clear though, is that RNA molecules for use in the invention may be designed based on gene sequence information in the public databases or newly discovered, and then made artificially, e.g. by chemical synthesis in whole or in part. The RNA molecules of the invention may also be designed and produced by way of expression in genetically modified cells or cell free expression systems and this option may include synthesis of some or all of the RNA sequence.
  • The structure and requirements of crRNA has also been described in Semenova E et al. (2011) Proc. Natl. Acad. Sci. USA 108: 10098-10103. There is a so-called “SEED” portion forming the 5′ end of the spacer sequence and which is flanked 5′ thereto by the 5′ handle of 8 nucleotides. Semenova et al. (2011) have found that all residues of the SEED sequence should be complementary to the target sequence, although for the residue at position 6, a mismatch may be tolerated. Similarly, when designing and making an RNA component of a ribonucleoprotein complex of the invention directed at a target locus (i.e. sequence), the necessary match and mismatch rules for the SEED sequence can be applied.
  • The invention therefore includes a method of detecting and/or locating a single base change in a target nucleic acid molecule comprising contacting a nucleic acid sample with a ribonucleoprotein complex of the invention as hereinbefore described, or with a Cascade complex and separate RNA component of the invention as hereinbefore described, and wherein the sequence of the RNA component (including when in the ribonucleoprotein complex) is such that it discriminates between a normal allele and a mutant allele by virtue of a single base change at position 6 of a contiguous sequence of 8 nucleotide residues.
  • In embodiments of the invention, the RNA molecule may have a length in the range of 35-75 residues. In preferred embodiments, the portion of the RNA which is complementary to and used for targeting a desired nucleic acid sequence is 32 or 33 residues long. (In the context of a naturally occurring crRNA, this would correspond to the spacer portion; as shown in FIG. 1 of Semenova et al. (2011)).
  • A ribonucleoprotein complex of the invention may additionally have an RNA component comprising 8 residues 5′ to the RNA sequence which has at least substantial complementarity to the nucleic acid target sequence. (The RNA sequence having at least substantial complementarity to the nucleic acid target sequence would be understood to correspond in the context of a crRNA as being the spacer sequence. The 5′ flanking sequence of the RNA would be considered to correspond to the 5′ handle of a crRNA. This is shown in FIG. 1 of Semenova et al. (2011)).
  • A ribonucleoprotein complex of the invention may have a hairpin and tetranucleotide loop forming sequence 3′ to the RNA sequence which has at least substantial complementarity to the DNA target sequence. (In the context of crRNA, this would correspond to a 3′ handle flanking the spacer sequence as shown in FIG. 1 of Semenova et al. (2011)).
  • In some embodiments, the RNA may be a CRISPR RNA (crRNA).
  • The Cascade proteins and complexes of the invention may be characterised in vitro in terms of its activity of association with the RNA guiding component to form a ribonucleoprotein complex in the presence of the target nucleic acid (which may be DNA or RNA). An electrophoretic mobility shift assay (EMSA) may be used as a functional assay for interaction of complexes of the invention with their nucleic acid targets. Basically, Cascade-functional moiety complex of the invention is mixed with nucleic acid targets and the stable interaction of the Cascade-functional moiety complex is monitored by EMSA or by specific readout out the functional moiety, for example endonucleolytic cleavage of target DNA at the desired site. This can be determined by further restriction fragment length analysis using commercially available enzymes with known specificities and cleavage sites in a target DNA molecule.
  • Visualisation of binding of Cascade proteins or complexes of the invention to DNA or RNA in the presence of guiding RNA may be achieved using scanning/atomic force microscopy (SFM/AFM) imaging and this may provide an assay for the presence of functional complexes of the invention.
  • The invention also provides a nucleic acid molecule encoding at least one clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein subunit selected from:
      • a. a Cse1 subunit having an amino acid sequence of SEQ ID NO: 1 or a sequence of at least 9% identity therewith;
      • b. a Cse2 subunit having an amino acid sequence of SEQ ID NO:2 or a sequence of at least 20% identity therewith;
      • c. a Cas7 subunit having an amino acid sequence of SEQ ID NO:3 or a sequence of at least 18% identity therewith;
      • d. a Cas5 subunit having an amino acid sequence of SEQ ID NO:4 or a sequence of at least 17% identity therewith;
      • e. a Cas6 subunit having an amino acid sequence of SEQ ID NO:5 or a sequence of at least 16% identity therewith; and
        wherein at least a, b, c, d or e includes an additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity.
  • The additional amino acid sequence having nucleic acid or chromatin modifying, visualising, transcription activating or transcription repressing activity is preferably fused to the CRISPR-associated protein subunit.
  • In the nucleic acids of the invention defined above, the nucleotide sequence may be that which encodes the respective SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4 or SEQ ID NO:5, or in defining the range of variant sequences thereto, it may be a sequence hybridisable to that nucleotide sequence, preferably under stringent conditions, more preferably very high stringency conditions. A variety of stringent hybridisation conditions will be familiar to the skilled reader in the field. Hybridization of a nucleic acid molecule occurs when two complementary nucleic acid molecules undergo an amount of hydrogen bonding to each other known as Watson-Crick base pairing. The stringency of hybridization can vary according to the environmental (i.e. chemical/physical/biological) conditions surrounding the nucleic acids, temperature, the nature of the hybridization method, and the composition and length of the nucleic acid molecules used. Calculations regarding hybridization conditions required for attaining particular degrees of stringency are discussed in Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y., 2001); and Tijssen, Laboratory Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Acid Probes Part I, Chapter 2 (Elsevier, New York, 1993). The Tm is the temperature at which 50% of a given strand of a nucleic acid molecule is hybridized to its complementary strand. The following is an exemplary set of hybridization conditions and is not limiting:
  • Very High Stringency (Allows Sequences that Share at Least 90% Identity to Hybridize)
    Hybridization: 5×SSC at 65° C. for 16 hours
    Wash twice: 2×SSC at room temperature (RT) for 15 minutes each
    Wash twice: 0.5×SSC at 65° C. for 20 minutes each
    High Stringency (Allows Sequences that Share at Least 80% Identity to Hybridize)
    Hybridization: 5×-6×SSC at 65° C.-70° C. for 16-20 hours
    Wash twice: 2×SSC at RT for 5-20 minutes each
    Wash twice: 1×SSC at 55° C.-70° C. for 30 minutes each
    Low Stringency (Allows Sequences that Share at Least 50% Identity to Hybridize)
    Hybridization: 6×SSC at RT to 55° C. for 16-20 hours
    Wash at least twice: 2×-3×SSC at RT to 55° C. for 20-30 minutes each.
  • The nucleic acid molecule may be an isolated nucleic acid molecule and may be an RNA or a DNA molecule.
  • The additional amino acid sequence may be selected from a helicase, a nuclease, a nuclease-helicase (e.g. Cas3), a DNA methyltransferase (e.g. Dam), a DNA demethylase, a histone methyltransferase, a histone demethylase, an acetylase, a deacetylase, a phosphatase, a kinase, a transcription (co-)activator, an RNA polymerase subunit, a transcription repressor, a DNA binding protein, a DNA structuring protein, a marker protein, a reporter protein, a fluorescent protein, a ligand binding protein (e.g. mCherry or a heavy metal binding protein), a signal peptide (e.g. Tat-signal sequence), a subcellular localisation sequence (e.g. nuclear localisation sequence), or an antibody epitope. The additional amino acid sequence may be, or from a different protein from the organism from which the relevant Cascade protein subunit(s) are derived.
  • The invention includes an expression vector comprising a nucleic acid molecule as hereinbefore defined. One expression vector may contain the nucleotide sequence encoding a single Cascade protein subunit and also the nucleotide sequence encoding the additional amino acid sequence, whereby on expression the subunit and additional sequence are fused. Other expression vectors may comprise nucleotide sequences encoding just one or more Cascade protein subunits which are not fused to any additional amino acid sequence.
  • The additional amino acid sequence with nucleic acid or chromatin modifying activity may be fused to any of the Cascade subunits via a linker polypeptide. The linker may be of any length up to about 60 or up to about 100 amino acid residues. Preferably the linker has a number of amino acids in the range 10 to 60, more preferably 10-20. The amino acids are preferably polar and/or small and/or charged amino acids (e.g. Gln, Ser, Thr, Pro, Ala, Glu, Asp, Lys, Arg, His, Asn, Cys, Tyr). The linker peptide is preferably designed to obtain the correct spacing and positioning of the fused functional moiety and the subunit of Cascade to which the moiety is fused to allow proper interaction with the target nucleotide.
  • An expression vector of the invention (with or without nucleotide sequence encoding amino acid residues which on expression will be fused to a Cascade protein subunit) may further comprise a sequence encoding an RNA molecule as hereinbefore defined. Consequently, such expression vectors can be used in an appropriate host to generate a ribonucleoprotein of the invention which can target a desired nucleotide sequence.
  • Accordingly, the invention also provides a method of modifying, visualising, or activating or repressing transcription of a target nucleic acid comprising contacting the nucleic acid with a ribonucleoprotein complex as hereinbefore defined. The modifying may be by cleaving the nucleic acid or binding to it.
  • The invention also includes a method of modifying, visualising, or activating or repressing transcription of a target nucleic acid comprising contacting the nucleic acid with a Cascade protein complex as hereinbefore defined, plus an RNA molecule as hereinbefore defined.
  • In accordance with the above methods, the modification, visualising, or activating or repressing transcription of a target nucleic acid may therefore be carried out in vitro and in a cell free environment; i.e. the method is carried out as a biochemical reaction whether free in solution or whether involving a solid phase. Target nucleic acid may be bound to a solid phase, for example.
  • In a cell free environment, the order of adding each of the target nucleic acid, the Cascade protein complex and the RNA molecule is at the option of the average skilled person. The three components may be added simultaneously, sequentially in any desired order, or separately at different times and in a desired order. Thus it is possible for the target nucleic acid and RNA to be added simultaneously to a reaction mix and then the Cascade protein complex of the invention to be added separately and later in a sequence of specific method steps.
  • The modification, visualising, or activating or repressing transcription of a target nucleic acid may be made in situ in a cell, whether an isolated cell or as part of a multicellular tissue, organ or organism. Therefore in the context of whole tissue and organs, and in the context of an organism, the method can be carried out in vivo or it can be carried out by isolating a cell from the whole tissue, organ or organism and then returning the cell treated with ribonucleoprotein complex to its former location, or a different location, whether within the same or a different organism. Thus the method would include allografts, autografts, isografts and xenografts.
  • In these embodiments, the ribonucleoprotein complex or the Cascade protein complex of the invention requires an appropriate form of delivery into the cell, which will be well known to persons of skill in the art, including microinjection, whether into the cell cytoplasm or into the nucleus.
  • Also when present separately, the RNA molecule requires an appropriate form of delivery into a cell, whether simultaneously, separately or sequentially with the Cascade protein complex. Such forms of introducing RNA into cells are well known to a person of skill in the art and may include in vitro or ex vivo delivery via conventional transfection methods. Physical methods, such as microinjection and electroporation, as well as calcium co-precipitation, and commercially available cationic polymers and lipids, and cell-penetrating peptides, cell-penetrating particles (gene-gun) may each be used. For example, viruses may be used as delivery vehicles, whether to the cytoplasm and/or nucleus—e.g. via the (reversible) fusion of Cascade protein complex of the invention or a ribonucleoprotein complex of the invention to the viral particle. Viral delivery (e.g. adenovirus delivery) or Agrobacterium-mediated delivery may be used.
  • The invention also includes a method of modifying visualising, or activating or repressing transcription of a target nucleic acid in a cell, comprising transfecting, transforming or transducing the cell with any of the expression vectors as hereinbefore described. The methods of transfection, transformation or transduction are of the types well known to a person of skill in the art. Where there is one expression vector used to generate expression of a Cascade complex of the invention and when the RNA is added directly to the cell then the same or a different method of transfection, transformation or transduction may be used. Similarly, when there is one expression vector being used to generate expression of a Cascade-functional fusion complex of the invention and when another expression vector is being used to generate the RNA in situ via expression, then the same or a different method of transfection, transformation or transduction may be used.
  • In other embodiments, mRNA encoding the Cascade complex of the invention is introduced into a cell so that the Cascade complex is expressed in the cell. The RNA which guides the Cascade complex to the desired target sequence is also introduced into the cell, whether simultaneously, separately or sequentially from the mRNA, such that the necessary ribonucleoprotein complex is formed in the cell.
  • In the aforementioned methods of modifying or visualising a target nucleic acid, the additional amino acid sequence may be a marker and the marker associates with the target nucleic acid; preferably wherein the marker is a protein; optionally a fluorescent protein, e.g. green fluorescent protein (GFP) or yellow fluorescent protein (YFP) or mCherry. Whether in vitro, ex vivo or in vivo, then methods of the invention can be used to directly visualise a target locus in a nucleic acid molecule, preferably in the form of a higher order structure such as a supercoiled plasmid or chromosome, or a single stranded target nucleic acid such as mRNA. Direct visualisation of a target locus may use electron micrography, or fluorescence microscopy.
  • Other kinds of label may be used to mark the target nucleic acid including organic dye molecules, radiolabels and spin labels which may be small molecules.
  • In methods of the invention described above, the target nucleic acid is DNA; preferably dsDNA although the target can be RNA; preferably mRNA.
  • In methods of the invention for modifying, visualising, activating transcription or repressing transcription of a target nucleic acid wherein the target nucleic acid is dsDNA, the additional amino acid sequence with nucleic acid or chromatin modifying activity may be a nuclease or a helicase-nuclease, and the modification is preferably a single stranded or a double stranded break at a desired locus. In this way unique sequence specific cutting of DNA can be engineered by using the Cascade-functional moiety complexes. The chosen sequence of the RNA component of the final ribonucleoprotein complex provides the desired sequence specificity for the action of the additional amino acid sequence.
  • Therefore, the invention also provides a method of non-homologous end joining of a dsDNA molecule in a cell at a desired locus to remove at least a part of a nucleotide sequence from the dsDNA molecule; optionally to knockout the function of a gene or genes, wherein the method comprises making double stranded breaks using any of the methods of modifying a target nucleic acid as hereinbefore described.
  • The invention further provides a method of homologous recombination of a nucleic acid into a dsDNA molecule in a cell at a desired locus in order to modify an existing nucleotide sequence or insert a desired nucleotide sequence, wherein the method comprises making a double or single stranded break at the desired locus using any of the methods of modifying a target nucleic acid as hereinbefore described.
  • The invention therefore also provides a method of modifying, activating or repressing gene expression in an organism comprising modifying, activating transcription or repressing transcription of a target nucleic acid sequence according to any of the methods hereinbefore described, wherein the nucleic acid is dsDNA and the functional moiety is selected from a DNA modifying enzyme (e.g. a demethylase or deacetylase), a transcription activator or a transcription repressor.
  • The invention additionally provides a method of modifying, activating or repressing gene expression in an organism comprising modifying, activating transcription or repressing transcription of a target nucleic acid sequence according to any of the methods hereinbefore described, wherein the nucleic acid is an mRNA and the functional moiety is a ribonuclease; optionally selected from an endonuclease, a 3′ exonuclease or a 5′ exonuclease.
  • In any of the methods of the invention as described above, the cell which is subjected to the method may be a prokaryote. Similarly, the cell may be a eukaryotic cell, e.g. a plant cell, an insect cell, a yeast cell, a fungal cell, a mammalian cell or a human cell. When the cell is of a mammal or human then it can be a stem cell (but may not be any human embryonic stem cell). Such stem cells for use in the invention are preferably isolated stem cells. Optionally in accordance with any method the invention a cell is transfected in vitro.
  • Preferably though, in any of the methods of the invention, the target nucleic acid has a specific tertiary structure, optionally supercoiled, more preferably wherein the target nucleic acid is negatively supercoiled. Advantageously, the ribonucleoprotein complexes of the invention, whether produced in vitro, or whether formed within cells, or whether formed within cells via expression machinery of the cell, can be used to target a locus which would otherwise be difficult to get access to in order to apply the functional activity of a desired component, whether labelling or tagging of a specific sequence, modification of nucleic acid structure, switching on or off of gene expression, or of modification of the target sequence itself involving single or double stranded cutting followed by insertion of one or more nucleotide residues or a cassette.
  • The invention also includes a pharmaceutical composition comprising a Cascade protein complex or a ribonucleoprotein complex of the invention as hereinbefore described.
  • The invention further includes a pharmaceutical composition comprising an isolated nucleic acid or an expression vector of the invention as hereinbefore described.
  • Also provided is a kit comprising a Casacade protein complex of the invention as hereinbefore described plus an RNA molecule of the invention as hereinbefore described.
  • The invention includes a Cascade protein complex or a ribonucleoprotein complex or a nucleic acid or a vector, as hereinbefore described for use as a medicament.
  • The invention allows a variety of possibilities to physically alter DNA of prokaryotic or eukaryotic hosts at a specified genomic locus, or change expression patterns of a gene at a given locus. Host genomic DNA can be cleaved or modified by methylation, visualized by fluorescence, transcriptionally activated or repressed by functional domains such as nucleases, methylases, fluorescent proteins, transcription activators or repressors respectively, fused to suitable Cascade-subunits. Moreover, the RNA-guided RNA-binding ability of Cascade permits the monitoring of RNA trafficking in live cells using fluorescent Cascade fusion proteins, and provides ways to sequester or destroy host mRNAs causing interference with gene expression levels of a host cell.
  • In any of the methods of the invention, the target nucleic acid may be defined, preferably so if dsDNA, by the presence of at least one of the following nucleotide triplets: 5′-CTT-3′, 5′-CAT-3′, 5′-CCT-3′, or 5′-CTC-3′ (or 5′-CUU-3′, 5′-CAU-3′, 5′-CCU-3′, or 5′-CTC-3′ if the target is an RNA). The location of the triplet is in the target strand adjacent to the sequence to which the RNA molecule component of a ribonucleoprotein of the invention hybridizes. The triplet marks the point in the target strand sequence at which base pairing with the RNA molecule component of the ribonucleoprotein does not take place in a 5′ to 3′ (downstream) direction of the target (whilst it takes place upstream of the target sequence from that point subject to the preferred length of the RNA sequence of the RNA molecule component of the ribonucleoprotein of the invention). In the context of a native type I CRISPR system, the triplets correspond to what is known as a “PAM” (protospacer adjacent motif). For ssDNA or ssRNA targets, presence of one of the triplets is not so necessary.
  • The invention will now be described in detail and with reference to specific examples and drawings in which:
  • FIG. 1 shows the results of gel-shift assays where Cascade binds negatively supercoiled (nSC) plasmid DNA but not relaxed DNA. A) Gel-shift of nSC plasmid DNA with J3-Cascade, containing a targeting (J3) crRNA. pUC-λ, was mixed with 2-fold increasing amounts of J3-Cascade, from a pUC-λ:Cascade molar ratio of 1:0.5 up to a 1:256 molar ratio. The first and last lanes contain only pUC-λ. B) Gel-shift as in (A) with R44-Cascade containing a non-targeting (R44) cRNA. C) Gel-shift as in (A) with Nt.BspQI nicked pUC-λ. D) Gel-shift as in (A) with PdmI linearized pUC-λ. E) Fit of the fraction pUC-λ bound to J3-Cascade plotted against the concentration of free J3-Cascade gives the dissociation constant (Kd) for specific binding. F) Fit of the fraction pUC-λ bound to R44-Cascade plotted against the concentration of free R44-Cascade gives the dissociation constant (Kd) for non-specific binding. G) Specific binding of Cascade to the protospacer monitored by restriction analysis, using the unique BsmI restriction site in the protospacer sequence. Lane 1 and 5 contain only pUC-λ. Lane 2 and 6 contain pUC-λ mixed with Cascade. Lane 3 and 7 contain pUC-λ mixed with Cascade and subsequent BsmI addition. Lane 4 and 8 contain pUC-λ mixed with BsmI. H) Gel-shift of pUC-λ bound to Cascade with subsequent Nt.BspQI cleavage of one strand of the plasmid. Lane 1 and 6 contain only pUC-λ. Lane 2 and 7 contain pUC-λ, mixed with Cascade. Lane 3 and 8 contain pUC-λ mixed with Cascade and subsequent Nt.BspQI nicking. Lane 4 and 9 contain pUC-λ mixed with Cascade, followed by addition of a ssDNA probe complementary to the displaced strand in the R-loop and subsequent nicking with Nt.BspQI. Lane 5 and 10 contain pUC-λ nicked with Nt.BspQI. H) Gel-shift of pUC-λ bound to Cascade with subsequent Nt.BspQI nicking of the plasmid. Lane 1 and 6 contain only pUC-λ. Lane 2 and 7 contain pUC-λ mixed with Cascade. Lane 3 and 8 contain pUC-λ mixed with Cascade and subsequent Nt.BspQI cleavage. Lane 4 and 9 contain pUC-λ mixed with Cascade, followed by addition of a ssDNA probe complementary to the displaced strand in the R-loop and subsequent cleavage with Nt.BspQI. Lane 5 and 10 contain pUC-λ cleaved with Nt.BspQI. I) Gel-shift of pUC-λ bound to Cascade with subsequent EcoRI cleavage of both strands of the plasmid. Lane 1 and 6 contain only pUC-λ. Lane 2 and 7 contain pUC-λ mixed with Cascade. Lane 3 and 8 contain pUC-λ mixed with Cascade and subsequent EcoRI cleavage. Lane 4 and 9 contain pUC-λ mixed with Cascade, followed by addition of a ssDNA probe complementary to the displaced strand in the R-loop and subsequent cleavage with EcoRI. Lane 5 and 10 contain pUC-λ cleaved with EcoRI.
  • FIG. 2 shows scanning force micrographs demonstrating how Cascade induces bending of target DNA upon protospacer binding. A-P) Scanning force microscopy images of nSC plasmid DNA with J3-Cascade containing a targeting (J3) crRNA. pUC-λ was mixed with J3-Cascade at a pUC-λ:Cascade ratio of 1:7. Each image shows a 500×500 nm surface area. White dots correspond to Cascade.
  • FIG. 3 shows how BiFC analysis reveals that Cascade and Cas3 interact upon target recognition. A) Venus fluorescence of cells expressing CascadeΔCse1 and CRISPR 7Tm, which targets 7 protospacers on the phage genome, and Cse1-N155Venus and Cas3-C85Venus fusion proteins. B) Brightfield image of the cells in (A). C) Overlay of (A) and (B). D) Venus fluorescence of phage λ infected cells expressing CascadeΔCse1 and CRISPR 7Tm, and Cse1-N155Venus and Cas3-C85Venus fusion proteins. E) Brightfield image of the cells in (G). F) Overlay of (G) and (H). G) Venus fluorescence of phage λ infected cells expressing CascadeΔCse1 and non-targeting CRISPR R44, and N155Venus and C85Venus proteins. H) Brightfield image of the cells in (J). I) Overlay of (J) and (K). J) Average of the fluorescence intensity of 4-7 individual cells of each strain, as determined using the profile tool of LSM viewer (Carl Zeiss).
  • FIG. 4 shows Cas3 nuclease and helicase activities during CRISPR-interference. A) Competent BL21-AI cells expressing Cascade, a Cas3 mutant and CRISPR J3 were transformed with pUC-λ. Colony forming units per microgram pUC-λ (cfu/μg DNA) are depicted for each of the strains expressing a Cas3 mutant. Cells expressing wt Cas3 and CRISPR J3 or CRISPR R44 serve as positive and negative controls, respectively. B) BL21-AI cells carrying Cascade, Cas3 mutant, and CRISPR encoding plasmids as well as pUC-λ are grown under conditions that suppress expression of the cas genes and CRISPR. At t=0 expression is induced. The percentage of cells that lost pUC-λ over time is shown, as determined by the ratio of ampicillin sensitive and ampicillin resistant cells.
  • FIG. 5 shows how a Cascade-Cas3 fusion complex provides in vivo resistance and has in vitro nuclease activity. A) Coomassie Blue stained SDS-PAGE of purified Cascade and Cascade-Cas3 fusion complex. B) Efficiency of plaquing of phage λ on cells expressing Cascade-Cas3 fusion complex and a targeting (J3) or non-targeting (R44) CRISPR and on cells expressing Cascade and Cas3 separately together with a targeting (J3) CRISPR. C) Gel-shift (in the absence of divalent metal ions) of nSC target plasmid with J3-Cascade-Cas3 fusion complex. pUC-λ was mixed with 2-fold increasing amounts of J3-Cascade-Cas3, from a pUC-λ:J3-Cascade-Cas3 molar ratio of 1:0.5 up to a 1:128 molar ratio. The first and last lane contain only pUC-λ. D) Gel-shift (in the absence of divalent metal ions) of nSC non-target plasmid with J3-Cascade-Cas3 fusion complex. pUC-p7 was mixed with 2-fold increasing amounts of J3-Cascade-Cas3, from a pUC-p7:J3-Cascade-Cas3 molar ratio of 1:0.5 up to a 1:128 molar ratio. The first and last lane contain only pUC-p7. E) Incubation of nSC target plasmid (pUC-λ, left) or nSC non-target plasmid (pUC-p7, right) with J3-Cascade-Cas3 in the presence of 10 mM MgCl2. Lane 1 and 7 contain only plasmid. F) Assay as in (E) in the presence of 2 mM ATP. G) Assay as in (E) with the mutant J3-Cascade-Cas3K320N complex. H) Assay as in (G) in the presence of 2 mM ATP.
  • FIG. 6 is a schematic diagram showing a model of the CRISPR-interference type I pathway in E. coli.
  • FIG. 7 is a schematic diagram showing how a Cascade-FokI fusion embodiment of the invention is used to create FokI dimers which cuts dsDNA to produce blunt ends as part of a process of non-homologous end joining or homologous recombination.
  • FIG. 8 shows how BiFC analysis reveals that Cascade and Cas3 interact upon target recognition. Overlay of Brightfield image and Venus fluorescence of cells expressing Cascade without Cse1, Cse1-N155Venus and Cas3-C85Venus and either CRISPR 7Tm, which targets 7 protospacers on the phage Lambda genome, or the non-targeting CRISPR R44. Cells expressing CRISPR 7Tm are fluorescent only when infected with phage Lambda, while cells expressing CRISPR R44 are non-fluorescent. The highly intense fluorescent dots (outside cells) are due to light-reflecting salt crystals. White bars correspond to 10 micron.
  • FIG. 9 shows pUC-λ, sequences of 4 clones [SEQ ID NOs: 39-42] encoding CRISPR J3, Cascade and Cas3 (wt or S483AT485A) indicate that these are escape mutants carrying (partial) deletions of the protospacer or carrying a single point mutation in the seed region, which explains the inability to cure these plasmids.
  • FIG. 10 shows sequence alignments of cas3 genes from organisms containing the Type I-E CRISPR/Cas system. Alignment of cas3-cse1 genes from Streptomyces sp. SPB78 (1st sequence, Accession Number: ZP_07272643.1) [SEQ ID NO: 43], in Streptomyces griseus (2nd sequence, Accession Number YP_001825054) [SEQ ID NO: 44], and in Catenulispora acidiphila DSM 44928 (3rd sequence, Accession Number YP_003114638) [SEQ ID NO: 45] and an artificial E. coli Cas3-Cse1 fusion protein [SEQ ID NO: 46] which includes the polypeptide linker sequence from S. griseus.
  • FIG. 11 shows the design of a CascadeKKR/ELD nuclease pair in which FokI nuclease domains are mutated such that only heterodimers consisting of KKR and ELD nuclease domains are and the distance between the opposing binding sites may be varied to determine the optimal distance between a Cascade nuclease pair.
  • FIG. 12 is a schematic diagram showing genome targeting by a Cascade-FokI nuclease pair.
  • FIG. 13 shows an SDS PAGE gel of Cascade-nuclease complexes.
  • FIG. 14 shows electrophoresis gels of in vitro cleavage assays of CascadeKKR/ELD on plasmid DNA.
  • FIG. 15 shows CascadeKKR/ELD cleavage patterns and frequency [SEQ ID NO: 47].
  • EXAMPLES Materials and Methods Used Strains, Gene Cloning, Plasmids and Vectors
  • E. coli BL21-AI and E. coli BL21 (DE3) strains were used throughout. Table 1 lists all plasmids used in this study. The previously described pWUR408, pWUR480, pWUR404 and pWUR547 were used for production of Strep-tag II R44-Cascade, and pWUR408, pWUR514 and pWUR630 were used for production of Strep-tag II J3-Cascade (Jore et al., (2011) Nature Structural & Molecular Biology 18, 529-536; Semenova et al., (2011) Proceedings of the National Academy of Sciences of the United States of America 108, 10098-10103.) pUC-λ (pWUR610) and pUC-p7 (pWUR613) have been described elsewhere (Jore et al., 2011; Semenova et al., 2011). The C85Venus protein is encoded by pWUR647, which corresponds to pET52b (Novagen) containing the synthetic GA1070943 construct (Table 2) (Geneart) cloned between the BamHI and NotI sites. The N155Venus protein is encoded by pWUR648, which corresponds to pRSF1b (Novagen) containing the synthetic GA1070941 construct (Table 2) (Geneart) cloned between the NotI and XhoI sites. The Cas3-C85Venus fusion protein is encoded by pWUR649, which corresponds to pWUR647 containing the Cas3 amplification product using primers BG3186 and BG3213 (Table 3) between the NcoI and BamHI sites. The CasA-N155Venus fusion protein is encoded by pWUR650, which corresponds to pWUR648 containing the CasA amplification product using primers BG3303 and BG3212 (Table 3) between the NcoI and BamHI sites. CRISPR 7Tm is encoded by pWUR651, which corresponds to pACYCDuet-1 (Novagen) containing the synthetic GA1068859 construct (Table 2) (Geneart) cloned between the NcoI and KpnI sites. The Cascade encoding pWUR400, the CascadeΔCse1 encoding WUR401 and the Cas3 encoding pWUR397 were described previously (Jore et al., 2011). The Cas3H74A encoding pWUR652 was constructed using site directed mutagenesis of pWUR397 with primers BG3093, BG3094 (Table 3).
  • TABLE 1
    Plasmids used
    Description and order Restriction
    Plasmids of genes (5′-3′) sites Primers Source
    pWUR397 cas3 in pRSF-1b, no 1
    tags
    pWUR400 casA-casB-casC-casD- 1
    casE in pCDF-1b, no
    tags
    pWUR401 casB-casC-casD-casE 1
    in pCDF-lb, no tags
    pWUR404 casE in pCDF-1b, no 1
    tags
    pWUR408 casA in pRSF-1b, no 1
    tags
    pWUR480 casB with Strep-tag II 1
    (N-term)-casC-casD in
    pET52b
    pWUR514 casB with Strep-tag II 2
    (N-term)-casC-casD-
    CasE in pET52b
    pWUR547 E. coli R44 CRISPR, 7x 2
    spacer nr. 2, in
    pACYCDuet-1
    pWUR613 pUC-p7; pUC19 2
    containing R44-
    protospacer on a 350 bp
    phage P7 amplicon
    pWUR630 CRISPR poly J3, 5x NcoI/KpnI This
    spacer J3 in study
    pACYCDuet-1
    pWUR610 pUC-λ;pUC19 3
    containing J3-
    protospacer on a 350 bp
    phage λ amplicon
    pWUR647 C85Venus; GA1070943 BamHI/NotI This
    (Table S1) in pET52b study
    pWUR648 N155Venus; NotI/XhoI This
    GA1070941 (Table S1) study
    in pRSF1b
    pWUR649 cas3-C85Venus; NcoI/BamHI BG3186 + This
    pWUR647 containing BG3213 study
    cas3 amplicon
    pWUR650 casA-N155Venus NcoI/NotI BG3303 + This
    pWUR648 containing BG3212 study
    casA amplicon
    pWUR651 CRISPR 7Tm; NcoI/KpnI This
    GA1068859 (Table S1) study
    in pACYCDuet-1
    casB with Strep-tag II This
    (N-term)-casC-casD- study
    CasE in pCDF-1b
    cas3-casA fusion This
    study
    cas3H74A-CasA fusion This
    study
    cas3D75A-CasA fusion This
    study
    cas3K320N-CasA This
    fusion study
    cas3D452N-CasA This
    fusion study
    • Source 1 in the table above is Brouns et al (2008) Science 321, 960-964.
    • Source 2 in the table above is Jore et al (2011) Nature Structural & Molecular Biology 18: 529-537.
  • TABLE 2
    Synthetic Constructs
    GA1070943
    [SEQ ID NO: 6]
    ACTGGAAAGCGGGCAGTGAAAGGAAGGCCCATGAGGCCAGTTAATTAA
    GCGGATCCTGGCGGCGGCAGCGGCGGCGGCAGCGACAAGCAGAAGAAC
    GGCATCAAGGCGAACTTCAAGATCCGCCACAACATCGAGGACGGCGGC
    GTGCAGCTCGCCGACCACTACCAGCAGAACACCCCCATCGGCGACGGC
    CCCGTGCTGCTGCCCGACAACCACTACCTGAGCTACCAGTCCGCCCTG
    AGCAAAGACCCCAACGAGAAGCGCGATCACATGGTCCTGCTGGAGTTC
    GTGACCGCCGCCGGGATCACTCTCGGCATGGACGAGCTGTACAAGTAA
    GCGGCCGCGGCGCGCCTAGGCCTTGACGGCCTTCCTTCAATTCGCCCT
    ATAGTGAG
    GA1070941
    [SEQ ID NO: 7]
    CACTATAGGGCGAATTGGCGGAAGGCCGTCAAGGCCGCATTTAATTAA
    GCGGCCGCAGGCGGCGGCAGCGGCGGCGGCAGCATGGTGAGCAAGGGC
    GAGGAGCTGTTCACCGGGGTGGTGCCCATCCTGGTCGAGCTGGACGGC
    GACGTAAACGGCCACAAGTTCAGCGTGTCCGGCGAGGGCGAGGGCGAT
    GCCACCTACGGCAAGCTGACCCTGAAGCTCATCTGCACCACCGGCAAG
    CTGCCCGTGCCCTGGCCCACCCTCGTGACCACCCTCGGCTACGGCCTG
    CAGTGCTTCGCCCGCTACCCCGACCACATGAAGCAGCACGACTTCTTC
    AAGTCCGCCATGCCCGAAGGCTACGTCCAGGAGCGCACCATCTTCTTC
    AAGGACGACGGCAACTACAAGACCCGCGCCGAGGTGAAGTTCGAGGGC
    GACACCCTGGTGAACCGCATCGAGCTGAAGGGCATCGACTTCAAGGAG
    GACGGCAACATCCTGGGGCACAAGCTGGAGTACAACTACAACAGCCAC
    AACGTCTATATCACGGCCTAACTCGAGGGCGCGCCCTGGGCCTCATGG
    GCCTTCCGCTCACTGCCCGCTTTCCAG
    GA1068859
    [SEQ ID NO: 8]
    CACTATAGGGCGAATTGGCGGAAGGCCGTCAAGGCCGCATGAGCTCCA
    TGGAAACAAAGAATTAGCTGATCTTTAATAATAAGGAAATGTTACATT
    AAGGTTGGTGGGTTGTTTTTATGGGAAAAAATGCTTTAAGAACAAATG
    TATACTTTTAGAGAGTTCCCCGCGCCAGCGGGGATAAACCGGGCCGAT
    TGAAGGTCCGGTGGATGGCTTAAAAGAGTTCCCCGCGCCAGCGGGGAT
    AAACCGCCGCAGGTACAGCAGGTAGCGCAGATCATCAAGAGTTCCCCG
    CGCCAGCGGGGATAAACCGACTTCTCTCCGAAAAGTCAGGACGCTGTG
    GCAGAGTTCCCCGCGCCAGCGGGGATAAACCGCCTACGCGCTGAACGC
    CAGCGGTGTGGTGAATGAGTTCCCCGCGCCAGCGGGGATAAACCGGTG
    TGGCCATGCACGCCTTTAACGGTGAACTGGAGTTCCCCGCGCCAGCGG
    GGATAAACCGCACGAACTCAGCCAGAACGACAAACAAAAGGCGAGTTC
    CCCGCGCCAGCGGGGATAAACCGGCACCAGTACGCGCCCCACGCTGAC
    GGTTTCTGAGTTCCCCGCGCCAGCGGGGATAAACCGCAGCTCCCATTT
    TCAAACCCAGGTACCCTGGGCCTCATGGGCCTTCCGCTCACTGCCCGC
    TTTCCAG
    GA1047360
    [SEQ ID NO: 9]
    GAGCTCCCGGGCTGACGGTAATAGAGGCACCTACAGGCTCCGGTAAAA
    CGGAAACAGCGCTGGCCTATGCTTGGAAACTTATTGATCAACAAATTG
    CGGATAGTGTTATTTTTGCCCTCCCAACACAAGCTACCGCGAATGCTA
    TGCTTACGAGAATGGAAGCGAGCGCGAGCCACTTATTTTCATCCCCAA
    ATCTTATTCTTGCTCATGGCAATTCACGGTTTAACCACCTCTTTCAAT
    CAATAAAATCACGCGCGATTACTGAACAGGGGCAAGAAGAAGCGTGGG
    TTCAGTGTTGTCAGTGGTTGTCACAAAGCAATAAGAAAGTGTTTCTTG
    GGCAAATCGGCGTTTGCACGATTGATCAGGTGTTGATTTCGGTATTGC
    CAGTTAAACACCGCTTTATCCGTGGTTTGGGAATTGGTAGATCTGTTT
    TAATTGTTAATGAAGTTCATGCTTACGACACCTATATGAACGGCTTGC
    TCGAGGCAGTGCTCAAGGCTCAGGCTGATGTGGGAGGGAGTGTTATTC
    TTCTTTCCGCAACCCTACCAATGAAACAAAAACAGAAGCTTCTGGATA
    CTTATGGTCTGCATACAGATCCAGTGGAAAATAACTCCGCATATCCAC
    TCATTAACTGGCGAGGTGTGAATGGTGCGCAACGTTTTGATCTGCTAG
    CGGATCCGGTACC
  • TABLE 3
    Primers
    BG3186 ATAGCGCCATGGAACCTTTTAAATATATATGCCATTA
    [SEQ ID NO: 10]
    BG3213 ACAGTGGGATCCGCTTTGGGATTTGCAGGGATGACTCTGGT
    [SEQ ID NO: 11]
    BG3303 ATAGCGTCATGAATTTGCTTATTGATAACTGGATTCCTGTACG
    [SEQ ID NO: 12]
    BG3212 ACAGTGGCGGCCGCGCCATTTGATGGCCCTCCTTGCGGTTTTAA
    [SEQ ID NO: 13]
    BG3076 CGTATATCAAACTTTCCAATAGCATGAAGAGCAATGAAAAATAAC
    [SEQ ID NO: 14]
    BG3449 ATGATACCGCGAGACCCACGCTC
    [SEQ ID NO: 15]
    BG3451 CGGATAAAGTTGCAGGACCACTTC
    [SEQ ID NO: 16]
  • Protein Production and Purification
  • Cascade was expressed and purified as described (Jore et al., 2011). Throughout purification a buffer containing 20 mM HEPES pH 7.5, 75 mM NaCl, 1 mM DTT, 2 mM EDTA was used for resuspension and washing. Protein elution was performed in the same buffer containing 4 mM desthiobiotin. The Cascade-Cas3 fusion complex was expressed and purified in the same manner, with washing steps being performed with 20 mM HEPES pH 7.5, 200 mM NaCl and 1 mM DTT, and elution in 20 mM HEPES pH 7.5, 75 mM NaCl, 1 mM DTT containing 4 mM desthiobiotin.
  • Electrophoretic Mobility Shift Assay
  • Purified Cascade or Cascade subsomplexes were mixed with pUC-λ in a buffer containing 20 mM HEPES pH 7.5, 75 mM NaCl, 1 mM DTT, 2 mM EDTA, and incubated at 37° C. for 15 minutes. Samples were run overnight on a 0.8% TAE Agarose gel and post-stained with SybR safe (Invitrogen) 1:10000 dilution in TAE for 30 minutes. Cleavage with BsmI (Fermentas) or Nt.BspQI (New England Biolabs) was performed in the HEPES reaction buffer supplemented with 5 mM MgCl2.
  • Scanning Force Microscopy
  • Purified Cascade was mixed with pUC-λ(at a ratio of 7:1, 250 nM Cascade, 35 nM DNA) in a buffer containing 20 mM HEPES pH 7.5, 75 mM NaCl, 0.2 mM DTT, 0.3 mM EDTA and incubated at 37° C. for 15 minutes. Subsequently, for AFM sample preparation, the incubation mixture was diluted 10× in double distilled water and MgCl2 was added at a final concentration of 1.2 mM. Deposition of the protein-DNA complexes and imaging was carried out as described before (Dame et al., (2000) Nucleic Acids Res. 28: 3504-3510).
  • Fluorescence Microscopy
  • BL21-AI cells carrying CRISPR en cas gene encoding plasmids, were grown overnight at 37° C. in Luria-Bertani broth (LB) containing ampicillin (100 μg/ml), kanamycin (50 μg/ml), streptomycin (50 μg/ml) and chloramphenicol (34 μg/ml). Overnight culture was diluted 1:100 in fresh antibiotic-containing LB, and grown for 1 hour at 37° C. Expression of cas genes and CRISPR was induced for 1 hour by adding L-arabinose to a final concentration of 0.2% and IPTG to a final concentration of 1 mM. For infection, cells were mixed with phage Lambda at a Multiplicity of Infection (MOI) of 4. Cells were applied to poly-L-lysine covered microscope slides, and analyzed using a Zeiss LSM510 confocal laser scanning microscope based on an Axiovert inverted microscope, with a 40× oil immersion objective (N.A. of 1.3) and an argon laser as the excitation source (514 nm) and detection at 530-600 nm. The pinhole was set at 203 μm for all measurements.
  • pUC-λ Transformation Studies
  • LB containing kanamycin (50 μg/ml), streptomycin (50 μg/ml) and chloramphenicol (34 μg/ml) was inoculated from an overnight pre-inoculum and grown to an OD600 of 0.3. Expression of cas genes and CRISPR was induced for 45 minutes with 0.2% L-arabinose and 1 mM IPTG. Cells were collected by centrifugation at 4° C. and made competent by resuspension in ice cold buffer containing 100 mM RbCl2, 50 mM MnCl2, 30 mM potassium acetate, 10 mM CaCl2 and 15% glycerol, pH 5.8. After a 3 hour incubation, cells were collected and resuspended in a buffer containing 10 mM MOPS, 10 mM RbCl, 75 mM CaCl2, 15% glycerol, pH 6.8. Transformation was performed by adding 80 ng pUC-λ, followed by a 1 minute heat-shock at 42° C., and 5 minute cold-shock on ice. Next cells were grown in LB for 45 minutes at 37° C. before plating on LB-agar plates containing 0.2% L-arabinose, 1 mM IPTG, ampicillin (100 μg/ml), kanamycin (50 μg/ml), streptomycin (50 μg/ml) and chloramphenicol (34 μg/ml).
  • Plasmid curing was analyzed by transforming BL21-AI cells containing cas gene and CRISPR encoding plasmids with pUC-λ, while growing the cells in the presence of 0.2% glucose to suppress expression of the T7-polymerase gene. Expression of cas genes and CRISPR was induced by collecting the cells and re-suspension in LB containing 0.2% arabinose and 1 mM IPTG. Cells were plated on LB-agar containing either streptomycin, kanamycin and chloramphenicol (non-selective for pUC-λ) or ampicillin, streptomycin, kanamycin and chloramphenicol (selective for pUC-λ). After overnight growth the percentage of plasmid loss can be calculated from the ratio of colony forming units on the selective and non-selective plates.
  • Phage Lambda Infection Studies
  • Host sensitivity to phage infection was tested using a virulent phage Lambda (λvir), as in (Brouns et al (2008) Science 321, 960-964.). The sensitivity of the host to infection was calculated as the efficiency of plaquing (the plaque count ratio of a strain containing an anti-λ CRISPR to that of the strain containing a non-targeting R44 CRISPR) as described in Brouns et al (2008).
  • Example 1 Cascade Exclusively Binds Negatively Supercoiled Target DNA
  • The 3 kb pUC19-derived plasmid denoted pUC-λ, contains a 350 bp DNA fragment corresponding to part of the J gene of phage λ, which is targeted by J3-Cascade (Cascade associated with crRNA containing spacer J3 (Westra et al (2010) Molecular Microbiology 77, 1380-1393). The electrophoretic mobility shift assays show that Cascade has high affinity only for negatively supercoiled (nSC) target plasmid. At a molar ratio of J3-Cascade to pUC-λ of 6:1 all nSC plasmid was bound by Cascade, (see FIG. 1A), while Cascade carrying the non-targeting crRNA R44 (R44-Cascade) displayed non-specific binding at a molar ratio of 128:1 (see FIG. 1B). The dissociation constant (Kd) of nSC pUC-λ was determined to be 13±1.4 nM for J3-Cascade (see FIG. 1E) and 429±152 nM for R44-Cascade (see FIG. 1F).
  • J3-Cascade was unable to bind relaxed target DNA with measurable affinity, such as nicked (see FIG. 1C) or linear pUC-λ (see FIG. 1D), showing that Cascade has high affinity for larger DNA substrates with a nSC topology.
  • To distinguish non-specific binding from specific binding, the BsmI restriction site located within the protospacer was used. While adding BsmI enzyme to pUC-λ gives a linear product in the presence of R44-Cascade (see FIG. 1G, lane 4), pUC-λ is protected from BsmI cleavage in the presence of J3-Cascade (see FIG. 1G, lane 7), indicating specific binding to the protospacer. This shows that Cas3 is not required for in vitro sequence specific binding of Cascade to a protospacer sequence in a nSC plasmid.
  • Cascade binding to nSC pUC-λ was followed by nicking with Nt.BspQI, giving rise to an OC topology. Cascade is released from the plasmid after strand nicking, as can be seen from the absence of a mobility shift (see FIG. 1H, compare lane 8 to lane 10). In contrast, Cascade remains bound to its DNA target when a ssDNA probe complementary to the displaced strand is added to the reaction before DNA cleavage by Nt.BspQI (see FIG. 1H, lane 9). The probe artificially stabilizes the Cascade R-loop on relaxed target DNA. Similar observations are made when both DNA strands of pUC-λ are cleaved after Cascade binding (see FIG. 1I, lane 8 and lane 9).
  • Example 2 Cascade Induces Bending of Bound Target DNA
  • Complexes formed between purified Cascade and pUC-λ were visualized. Specific complexes containing a single bound J3-Cascade complex were formed, while unspecific R44-Cascade yields no DNA bound complexes in this assay under identical conditions. Out of 81 DNA molecules observed 76% were found to have J3-Cascade bound (see FIGS. 2A-P). Of these complexes in most cases Cascade was found at the apex of a loop (86%), whereas a small fraction only was found at non-apical positions (14%). These data show that Cascade binding causes bending and possibly wrapping of the DNA, probably to facilitate local melting of the DNA duplex.
  • Example 3 Naturally Occurring Fusions of Cas3 and Cse1: Cas3 Interacts with Cascade Upon Protospacer Recognition
  • Figure S3 shows sequence analysis of cas3 genes from organisms containing the Type I-E CRISPR/Cas system reveals that Cas3 and Cse1 occur as fusion proteins in Streptomyces sp. SPB78 (Accession Number: ZP 07272643.1), in Streptomyces griseus (Accession Number YP_001825054), and in Catenulispora acidiphila DSM 44928 (Accession Number YP_003114638).
  • Example 4 Bimolecular Fluorescence Complementation (BiFC) Shows how a Cse1 Fusion Protein Forming Part of Cascade Continues to Interact with Cas3
  • BiFC experiments were used to monitor interactions between Cas3 and Cascade in vivo before and after phage λ infection. BiFC experiments rely on the capacity of the non-fluorescent halves of a fluorescent protein, e.g., Yellow Fluorescent Protein (YFP) to refold and to form a fluorescent molecule when the two halves occur in close proximity. As such, it provides a tool to reveal protein-protein interactions, since the efficiency of refolding is greatly enhanced if the local concentrations are high, e.g., when the two halves of the fluorescent protein are fused to interaction partners. Cse1 was fused at the C-terminus with the N-terminal 155 amino acids of Venus (Cse1-N155Venus), an improved version of YFP (Nagai et al (2002) Nature Biotechnology 20, 87-90). Cas3 was C-terminally fused to the C-terminal 85 amino acids of Venus (Cas3-C85Venus).
  • BiFC analysis reveals that Cascade does not interact with Cas3 in the absence of invading DNA (FIG. 3ABC, FIG. 3P and FIG. 8). Upon infection with phage λ, however, cells expressing CascadeΔCse1, Cse1-N155Venus and Cas3-C85Venus are fluorescent if they co-express the anti-2, CRISPR 7Tm (FIG. 3DEF, FIG. 3P and FIG. 8). When they co-express a non-targeting CRISPR R44 (FIG. 3GHI, FIG. 3P and FIG. 8), the cells remain non-fluorescent. This shows that Cascade and Cas3 specifically interact during infection upon protospacer recognition and that Cse1 and Cas3 are in close proximity of each other in the Cascade-Cas3 binary effector complex.
  • These results also show quite clearly that a fusion of Cse1 with an heterologous protein does not disrupt the ribonucleoprotein formation of Cascade and crRNA, nor does it disrupt the interaction of Cascade and Cas3 with the target phage DNA, even when the Cas3 itself is also a fusion protein.
  • Example 5 Preparing a Designed Cas3-Cse1 Fusion Gives a Protein with In Vivo Functional Activity
  • Providing in vitro evidence for Cas3 DNA cleavage activity required purified and active Cas3. Despite various solubilization strategies, Cas3 overproduced (Howard et al (2011) Biochem. J. 439, 85-95) in E. coli BL21 is mainly present in inactive aggregates and inclusion bodies. Cas3 was therefore produced as a Cas3-Cse1 fusion protein, containing a linker identical to that of the Cas3-Cse1 fusion protein in S. griseus (see FIG. 10). When co-expressed with CascadeΔCse1 and CRISPR J3, the fusion-complex was soluble and was obtained in high purity with the same apparent stoichiometry as Cascade (FIG. 5A). When functionality of this complex was tested for providing resistance against phage λ infection, the efficiency of plaquing (eop) on cells expressing the fusion-complex J3-Cascade-Cas3 was identical as on cells expressing the separate proteins (FIG. 5B).
  • Since the J3-Cascade-Cas3 fusion-complex was functional in vivo, in vitro DNA cleavage assays were carried out using this complex. When J3-Cascade-Cas3 was incubated with pUC-λ in the absence of divalent metals, plasmid binding was observed at molar ratios similar to those observed for Cascade (FIG. 5C), while a-specific binding to a non-target plasmid (pUC-p7, a pUC19 derived plasmid of the same size as pUC-λ, but lacking a protospacer) occurred only at high molar ratios (FIG. 5D), indicating that a-specific DNA binding of the complex is also similar to that of Cascade alone.
  • Interestingly, the J3-Cascade-Cas3 fusion complex displays magnesium dependent endonuclease activity on nSC target plasmids. In the presence of 10 mM Mg2+ J3-Cascade-Cas3 nicks nSC pUC-λ (FIG. 5E, lane 3-7), but no cleavage is observed for substrates that do not contain the target sequence (FIG. 5E, lane 9-13), or that have a relaxed topology. No shift of the resulting OC band is observed, in line with previous observations that Cascade dissociates spontaneously after cleavage, without requiring ATP-dependent Cas3 helicase activity. Instead, the helicase activity of Cas3 appears to be involved in exonucleolytic plasmid degradation. When both magnesium and ATP are added to the reaction, full plasmid degradation occurred (FIG. 5H).
  • The inventors have found that Cascade alone is unable to bind protospacers on relaxed DNA. In contrast, the inventors have found that Cascade efficiently locates targets in negatively supercoiled DNA, and subsequently recruits Cas3 via the Cse1 subunit. Endonucleolytic cleavage by the Cas3 HD-nuclease domain causes spontaneous release of Cascade from the DNA through the loss of supercoiling, remobilizing Cascade to locate new targets. The target is then progressively unwound and cleaved by the joint ATP-dependent helicase activity and HD-nuclease activity of Cas3, leading to complete target DNA degradation and neutralization of the invader.
  • Referring to FIG. 6 and without wishing to be bound to any particular theory, a mechanism of operation for the CRISPR-interference type I pathway in E. coli may involve (1) First, Cascade carrying a crRNA scans the nSC plasmid DNA for a protospacer, with adjacent PAM. Whether during this stage strand separation occurs is unknown. (2) Sequence specific protospacer binding is achieved through basepairing between the crRNA and the complementary strand of the DNA, forming an R-loop. Upon binding, Cascade induces bending of the DNA. (3) The Cse1 subunit of Cascade recruits Cas3 upon DNA binding. This may be achieved by Cascade conformational changes that take place upon nucleic acid binding. (4) The HD-domain (darker part) of Cas3 catalyzes Mg2+-dependent nicking of the displaced strand of the R-loop, thereby altering the topology of the target plasmid from nSC to relaxed OC. (5a and 5b) The plasmid relaxation causes spontaneous dissociation of Cascade. Meanwhile Cas3 displays ATP-dependent exonuclease activity on the target plasmid, requiring the helicase domain for target dsDNA unwinding and the HD-nuclease domain for successive cleavage activity. (6) Cas3 degrades the entire plasmid in an ATP-dependent manner as it processively moves along, unwinds and cleaves the target dsDNA.
  • Example 6 Preparation of Artificial Cas-Strep Tag Fusion Proteins and Assembly of Cascade Complexes
  • Cascade complexes are produced and purified as described in Brouns et al (2008) Science 321: 960-4 (2008), using the expression plasmids listed in Supplementary Table 3 of Jore et al (2011) Nature Structural & Molecular Biology 18: 529-537. Cascade is routinely purified with an N-terminal Strep-tag II fused to CasB (or CasC in CasCDE). Size exclusion chromatography (Superdex 200 HR 10/30 (GE)) is performed using 20 mM Tris-HCl (pH 8.0), 0.1 M NaCl, 1 mM dithiotreitol. Cascade preparations (˜0.3 mg) are incubated with DNase I (Invitrogen) in the presence of 2.5 mM MgCl2 for 15 min at 37° C. prior to size exclusion analysis. Co-purified nucleic acids are isolated by extraction using an equal volume of phenol:chloroform:isoamylalcohol (25:24:1) pH 8.0 (Fluka), and incubated with either DNase I (Invitrogen) supplemented with 2.5 mM MgCl2 or RNase A (Fermentas) for 10 min at 37° C. Cas subunit proteins fused to the amino acid sequence of Strep-Tag are produced.
  • Plaque assays showing the biological activity of the Strep-Tag Cascade subunits are performed using bacteriophage Lambda and the efficiency of plaquing (EOP) was calculated as described in Brouns et al (2008).
  • For purification of crRNA, samples are analyzed by ion-pair reversed-phased-HPLC on an Agilent 1100 HPLC with UV260nm detector (Agilent) using a DNAsep column 50 mm×4.6 mm I. D. (Transgenomic, San Jose, Calif.). The chromatographic analysis is performed using the following buffer conditions: A) 0.1 M triethylammonium acetate (TEAA) (pH 7.0) (Fluka); B) buffer A with 25% LC MS grade acetonitrile (v/v) (Fisher). crRNA is obtained by injecting purified intact Cascade at 75° C. using a linear gradient starting at 15% buffer B and extending to 60% B in 12.5 min, followed by a linear extension to 100% B over 2 min at a flow rate of 1.0 ml/min. Hydrolysis of the cyclic phosphate terminus was performed by incubating the HPLC-purified crRNA in a final concentration of 0.1 M HCl at 4° C. for 1 hour. The samples are concentrated to 5-10 μl on a vacuum concentrator (Eppendorf) prior to ESI-MS analysis.
  • Electrospray Ionization Mass spectrometry analysis of crRNA is performed in negative mode using an UHR-TOF mass spectrometer (maXis) or an HCT Ultra PTM Discovery instrument (both Bruker Daltonics), coupled to an online capillary liquid chromatography system (Ultimate 3000, Dionex, UK). RNA separations are performed using a monolithic (PS-DVB) capillary column (200 μm×50 mm I.D., Dionex, UK). The chromatography is performed using the following buffer conditions: C) 0.4 M 1,1,1,3,3,3,-Hexafluoro-2-propanol (HFIP, Sigma-Aldrich) adjusted with triethylamine (TEA) to pH 7.0 and 0.1 mM TEAA, and D) buffer C with 50% methanol (v/v) (Fisher). RNA analysis is performed at 50° C. with 20% buffer D, extending to 40% D in 5 min followed by a linear extension to 60% D over 8 min at a flow rate of 2 μl/min.
  • Cascade protein is analyzed by native mass spectrometry in 0.15 M ammonium acetate (pH 8.0) at a protein concentration of 5 μM. The protein preparation is obtained by five sequential concentration and dilution steps at 4° C. using a centrifugal filter with a cut-off of 10 kDa (Millipore). Proteins are sprayed from borosilicate glass capillaries and analyzed on a LCT electrospray time-of-flight or modified quadrupole time-of-flight instruments (both Waters, UK) adjusted for optimal performance in high mass detection (see Tahallah N et al (2001) Rapid Commun Mass Spectrom 15: 596-601 (2001) and van den Heuvel, R. H. et al. Anal Chem 78: 7473-83 (2006). Exact mass measurements of the individual Cas proteins were acquired under denaturing conditions (50% acetonitrile, 50% MQ, 0.1% formic acid). Sub-complexes in solution were generated by the addition of 2-propanol to the spray solution to a final concentration of 5% (v/v). Instrument settings were as follows; needle voltage ˜1.2 kV, cone voltage ˜175 V, source pressure 9 mbar. Xenon was used as the collision gas for tandem mass spectrometric analysis at a pressure of 1.5 10−2 mbar. The collision voltage varied between 10-200 V.
  • Electrophoretic mobility shift assays (EMSA) are used to demonstrate the functional activity of Cascade complexes for target nucleic acids. EMSA is performed by incubating Cascade, CasBCDE or CasCDE with 1 nM labelled nucleic acid in 50 mM Tris-Cl pH 7.5, 100 mM NaCl. Salmon sperm DNA (Invitrogen) is used as competitor. EMSA reactions are incubated at 37° C. for 20-30 min prior to electrophoresis on 5% polyacrylamide gels. The gels are dried and analyzed using phosphor storage screens and a PMI phosphor imager (Bio-Rad). Target DNA binding and cleavage activity of Cascade is tested in the presence of 1-10 mM Ca, Mg or Mn-ions.
  • DNA targets are gel-purified long oligonucleotides (Isogen Life Sciences or Biolegio), listed in Supplementary Table 3 of Jore et al (2011). The oligonucleotides are end-labeled using γ32P-ATP (PerkinElmer) and T4 kinase (Fermentas). Double-stranded DNA targets are prepared by annealing complementary oligonucleotides and digesting remaining ssDNA with Exonuclease I (Fermentas). Labelled RNA targets are in vitro transcribed using T7 Maxiscript or T7 Mega Shortscript kits (Ambion) with α32P-CTP (PerkinElmer) and removing template by DNase I (Fermentas) digestion. Double stranded RNA targets are prepared by annealing complementary RNAs and digesting surplus ssRNA with RNase T1 (Fermentas), followed by phenol extraction.
  • Plasmid mobility shift assays are performed using plasmid pWUR613 containing the R44 protospacer. The fragment containing the protospacer is PCR-amplified from bacteriophage P7 genomic DNA using primers BG3297 and BG 3298 (see Supplementary Table 3 of Jore et al (2011). Plasmid (0.4 μg) and Cascade were mixed in a 1:10 molar ratio in a buffer containing 5 mM Tris-HCl (pH 7.5) and 20 mM NaCl and incubated at 37° C. for 30 minutes.
  • Cascade proteins were then removed by proteinase K treatment (Fluka) (0.15 U, 15 min, 37° C.) followed by phenol/chloroform extraction. RNA-DNA complexes were then treated with RNaseH (Promega) (2 U, 1 h, 37° C.).
  • Strep-Tag-Cas protein subunit fusions which form Cascade protein complexes or active sub-complexes with the RNA component (equivalent to a crRNA), have the expected biological and functional activity of scanning and specific attachment and cleavage of nucleic acid targets. Fusions of the Cas subunits with the amino acid chains of fluorescent dyes also form Cascade complexes and sub-complexes with the RNA component (equivalent to crRNA) which retains biological and functional activity and allows visualisation of the location of a target nucleic acid sequence in ds DNA for example.
  • Example 7 A Cascade-Nuclease Pair and Test of Nuclease Activity In Vitro
  • Six mutations designated “Sharkey” have been introduced by random mutagenesis and screening to improve nuclease activity and stability of the non-specific nuclease domain from Flavobacterium okeanokoites restriction enzyme FokI (see Guo, J., et al. (2010) J. Mol. Biol. 400: 96-107). Other mutations have been introduced that reduce off-target cleavage activity. This is achieved by engineering electrostatic interactions at the FokI dimer interface of a ZFN pair, creating one FokI variant with a positively charged interface (KKR, E490K, I538K, H537R) and another with a negatively charged interface (ELD, Q486E, I499L, N496D) (see Doyon, Y., et al. (2011) Nature Methods 8: 74-9). Each of these variants is catalytically inactive as a homodimer, thereby reducing the frequency of off-target cleavage.
  • Cascade-Nuclease Design
  • We translationally fused improved FokI nucleases to the N-terminus of Cse1 to generate variants of Cse1 being FokIKKR-Cse1 and FokIELD-Cse1, respectively. These two variants are co-expressed with Cascade subunits (Cse2, Cas7, Cas5 and Cas6e), and one of two distinct CRISPR plasmids with uniform spacers. This loads the CascadeKKR complex with uniform P7-crRNA, and the CascadeELD complex with uniform M13 g8-crRNA. These complexes are purified using the N-terminally StrepII-tagged Cse2 as described in Jore, M. M., et al., (2011) Nat. Struct. Mol. Biol. 18(5): 529-536. Furthermore an additional purification step can be carried out using an N-terminally HIS-tagged FokI, to ensure purifying full length and intact Cascade-nuclease fusion complexes.
  • The nucleotide and amino acid sequences of the fusion proteins used in this example were as follows:
  • >nucleotide sequence of FokI-(Sharkey-ELD)-Cse1
    [SEQ ID NO: 18]
    ATGGCTCAACTGGTTAAAAGCGAACTGGAAGAGAAAAAAAGTGAACTGCGCCACAAACTGAAATATGTGCCGCATGAA
    TATATCGAGCTGATTGAAATTGCACGTAATCCGACCCAGGATCGTATTCTGGAAATGAAAGTGATGGAATTTTTTATG
    AAAGTGTACGGCTATCGCGGTGAACATCTGGGTGGTAGCCGTAAACCGGATGGTGCAATTTATACCGTTGGTAGCCCG
    ATTGATTATGGTGTTATTGTTGATACCAAAGCCTATAGCGGTGGTTATAATCTGCCGATTGGTCAGGCAGATGAAATG
    GAACGTTATGTGGAAGAAAATCAGACCCGTGATAAACATCTGAATCCGAATGAATGGTGGAAAGTTTATCCGAGCAGC
    GTTACCGAGTTTAAATTCCTGTTTGTTAGCGGTCACTTCAAAGGCAACTATAAAGCACAGCTGACCCGTCTGAATCAT
    ATTACCAATTGTAATGGTGCAGTTCTGAGCGTTGAAGAACTGCTGATTGGTGGTGAAATGATTAAAGCAGGCACCCTG
    ACCCTGGAAGAAGTTCGTCGCAAATTTAACAATGGCGAAATCAACTTTGCGGATCCCACCAACCGCGCGAAAGGCCTG
    GAAGCGGTGAGCGTGGCGAGCatgaatttgcttattgataactggattcctgtacgcccgcgaaacggggggaaagtc
    caaatcataaatctgcaatcgctatactgcagtagagatcagtggcgattaagtttgccccgtgacgatatggaactg
    gccgctttagcactgctggtttgcattgggcaaattatcgccccggcaaaagatgacgttgaatttcgacatcgcata
    atgaatccgctcactgaagatgagtttcaacaactcatcgcgccgtggatagatatgttctaccttaatcacgcagaa
    catccattatgcagaccaaaggtgtcaaagcaaatgatgtgactccaatggaaaaactgttggctggggtaagcggcg
    cgacgaattgtgcatttgtcaatcaaccggggcagggtgaagcattatgtggtggatgcactgcgattgcgttattca
    accaggcgaatcaggcaccaggttttggtggtggttttaaaagcggtttacgtggaggaacacctgtaacaacgttcg
    tacgtgggatcgatcttcgttcaacggtgttactcaatgtcctcacattacctcgtatcaaaaacaatttcctaatga
    atcacatacggaaaaccaacctacctggattaaacctatcaagtccaatgagtctatacctgcttcgtcaattgggtt
    tgtccgtggtctattctggcaaccagcgcatattgaattatgcgatcccattgggattggtaaatgttcttgctgtgg
    acaggaaagcaatttgcgttataccggttttcttaaggaaaaatttacctttacagttaatgggctatggccccatcc
    gcattccccttgtctggtaacagtcaagaaaggggaggttgaggaaaaatttcttgctttcaccacctccgcaccatc
    atggacacaaatcagccgagttgtggtagataagattattcaaaatgaaaatggaaatcgcgtggcggcggttgtgaa
    tcaattcagaaatattgcgccgcaaagtcctatgaattgattatggggggatatcgtaataatcaagcatctattctt
    gaacggcgtcatgatgtgttgatgtttaatcaggggtggcaacaatacggcaatgtgataaacgaaatagtgactgtt
    ggtttgggatataaaacagccttacgcaaggcgttatatacctttgcagaagggtttaaaaataaagacttcaaaggg
    gccggagtctctgttcatgagactgcagaaaggcatttctatcgacagagtgaattattaattcccgatgtactggcg
    aatgttaatttttcccaggctgatgaggtaatagctgatttacgagacaaacttcatcaattgtgtgaaatgctattt
    aatcaatctgtagctccctatgcacatcatcctaaattaataagcacattagcgcttgcccgcgccacgctatacaaa
    catttacgggagttaaaaccgcaaggagggccatcaaatggctga
    >protein sequence of FokI-(Sharkey-ELD)-Cse1
    [SEQ ID NO: 19]
    MAQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNPTQDRILEMKVMEFFMKVYGYRGEHLGGSRKPDGAIYTVGSP
    IDYGVIVDTKAYSGGYNLPIGQADEMERYVEENQTRDKHLNPNEWWKVYPSSVTEEKELEVSGHFKGNYKAQLTRLNH
    ITNCNGAVLSVEELLIGGEMIKAGTLTLEEVRRKENNGEINEADPTNRAKGLEAVSVASMNLLIDNWIPVRPRNGGKV
    QIINLQSLYCSRDQWRLSLPRDDMELAALALLVCIGQIIAPAKDDVEFRHRIMNPLTEDEFQQLIAPWIDMFYLNHAE
    HPFMQTKGVKANDVTPMEKLLAGVSGATNCAFVNQPGQGEALCGGCTAIALFNQANQAPGEGGGFKSGLRGGTPVTTF
    VRGIDLRSTVLLNVLTLPRLQKQFPNESHTENQPTWIKPIKSNESIPASSIGFVRGLFWQPAHIELCDPIGIGKCSCC
    GQESNLRYTGELKEKETFTVNGLWPHPHSPCLVTVKKGEVEEKELAFTTSAPSWTQISRVVVDKIIQNENGNRVAAVV
    NQFRNIAPQSPLELEVIGGYRNNQASILERRHDVLMENQGWQQYGNVINEIVTVGLGYKTALRKALYTEAEGEKNKDE
    KGAGVSVHETAERHFYRQSELLIPDVLANVNFSQADEVIADLRDKLHQLCEMLFNQSVAPYAHHPKLISTLALARATL
    YKHLRELKPQGGPSNG*
    >nucleotide sequence of FokI-(Sharkey -KKR)-Cse1
    [SEQ ID NO: 20]
    ATGGCTCAACTGGTTAAAAGCGAACTGGAAGAGAAAAAAAGTGAACTGCGCCACAAACTGAAATATGTGCCGCATGAA
    TATATCGAGCTGATTGAAATTGCACGTAATCCGACCCAGGATCGTATTCTGGAAATGAAAGTGATGGAATTTTTTATG
    AAAGTGTACGGCTATCGCGGTGAACATCTGGGTGGTAGCCGTAAACCGGATGGTGCAATTTATACCGTTGGTAGCCCG
    ATTGATTATGGTGTTATTGTTGATACCAAAGCCTATAGCGGTGGTTATAATCTGCCGATTGGTCAGGCAGATGAAATG
    CAGCGTTATGTGAAAGAAAATCAGACCCGCAACAAACATATTAACCCGAATGAATGGTGGAAAGTTTATCCGAGCAGC
    GTTACCGAGTTTAAATTCCTGTTTGTTAGCGGTCACTTCAAAGGCAACTATAAAGCACAGCTGACCCGTCTGAATCGT
    AAAACCAATTGTAATGGTGCAGTTCTGAGCGTTGAAGAACTGCTGATTGGTGGTGAAATGATTAAAGCAGGCACCCTG
    ACCCTGGAAGAAGTTCGTCGCAAATTTAACAATGGCGAAATCAACTTTGCGGATCCCACCAACCGCGCGAAAGGCCTG
    GAAGCGGTGAGCGTGGCGAGCatgaatttgcttattgataactggattcctgtacgcccgcgaaacggggggaaagtc
    caaatcataaatctgcaatcgctatactgcagtagagatcagtggcgattaagtttgccccgtgacgatatggaactg
    gccgctttagcactgctggtttgcattgggcaaattatcgccccggcaaaagatgacgttgaatttcgacatcgcata
    atgaatccgctcactgaagatgagtttcaacaactcatcgcgccgtggatagatatgttctaccttaatcacgcagaa
    catccctttatgcagaccaaaggtgtcaaagcaaatgatgtgactccaatggaaaaactgttggctggggtaagcggc
    gcgacgaattgtgcatttgtcaatcaaccggggcagggtgaagcattatgtggtggatgcactgcgattgcgttattc
    aaccaggcgaatcaggcaccaggttttggtggtggttttaaaagcggtttacgtggaggaacacctgtaacaacgttc
    gtacgtgggatcgatcttcgttcaacggtgttactcaatgtcctcacattacctcgtcttcaaaaacaatttcctaat
    gaatcacatacggaaaaccaacctacctggattaaacctatcaagtccaatgagtctatacctgcttcgtcaattggg
    tttgtccgtggtctattctggcaaccagcgcatattgaattatgcgatcccattgggattggtaaatgttcttgctgt
    ggacaggaaagcaatttgcgttataccggttttcttaaggaaaaatttacctttacagttaatgggctatggccccat
    ccgcattccccttgtctggtaacagtcaagaaaggggaggttgaggaaaaatttcttgctttcaccacctccgcacca
    tcatggacacaaatcagccgagttgtggtagataagattattcaaaatgaaaatggaaatcgcgtggcggcggttgtg
    aatcaattcagaaatattgcgccgcaaagtcctcttgaattgattatggggggatatcgtaataatcaagcatctatt
    cttgaacggcgtcatgatgtgttgatgttaatcaggggtggcaacaatacggcaatgtgataaacgaaatagtgactg
    ttggtttgggatataaaacagccttacgcaaggcgttatatacctttgcagaagggtttaaaaataaagacttcaaag
    gggccggagtctctgttcatgagactgcagaaaggcatttctatcgacagagtgaattattaattcccgatgtactgg
    cgaatgttaatttttcccaggctgatgaggtaatagctgatttacgagacaaacttcatcaattgtgtgaaatgctat
    ttaatcaatctgtagctccctatgcacatcatcctaaattaataagcacattagcgcttgcccgcgccacgctataca
    aacatttacgggagttaaaaccgcaaggagggccatcaaatggctga
    >protein sequence of FokI-(Sharkey-KKR)-Cse1
    [SEQ ID NO: 21]
    MAQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNPTQDRILEMKVMEFFMKVYGYRGEHLGGSRKPDGAIYTVGSP
    IDYGVIVDTKAYSGGYNLPIGQADEMQRYVKENQTRNKHINPNEWWKVYPSSVTEFKFLFVSGHFKGNYKAQLTRLNR
    KTNCNGAVLSVEELLIGGEMIKAGTLTLEEVRRKENNGEINFADPTNRAKGLEAVSVASMNLLIDNWIPVRPRNGGKV
    QIINLQSLYCSRDQWRLSLPRDDMELAALALLVCIGQIIAPAKDDVEFRHRIMNPLTEDEFQQLIAPWIDMFYLNHAE
    HPFMQTKGVKANDVTPMEKLLAGVSGATNCAFVNQPGQGEALCGGCTAIALFNQANQAPGEGGGEKSGLRGGTPVTTE
    VRGIDLRSTVLLNVLTLPRLQKQFPNESHTENQPTWIKPIKSNESIPASSIGFVRGLFWQPAHIELCDPIGIGKCSCC
    GQESNLRYTGELKEKETFTVNGLWPHPHSPCLVTVKKGEVEEKFLAFTTSAPSWTQISRVVVDKIIQNENGNRVAAVV
    NQFRNIAPQSPLELIMGGYRNNQASILERRHDVLMENQGWQQYGNVINEIVTVGLGYKTALRKALYTFAEGEKNKDFK
    GAGVSVHETAERHFYRQSELLIPDVLANVNFSQADEVIADLRDKLHQLCEMLFNQSVAPYAHHPKLISTLALARATLY
    KHLRELKPQGGPSNG*
    >nucleotide sequence of His6-Dual-monopartite NLS SV40-FokI-(Sharkey-KKR)-Cse1
    (“His6” disclosed as SEQ ID NO: 48)
    [SEQ ID NO: 22]
    ATGcatcaccatcatcaccacCCGAAAAAAAAGCGCAAAGTGGATCCGAAGAAAAAACGTAAAGTTGAAGATCCGAAA
    GACATGGCTCAACTGGTTAAAAGCGAACTGGAAGAGAAAAAAAGTGAACTGCGCCACAAACTGAAATATGTGCCGCAT
    GAATATATCGAGCTGATTGAAATTGCACGTAATCCGACCCAGGATCGTATTCTGGAAATGAAAGTGATGGAATTTTTT
    ATGAAAGTGTACGGCTATCGCGGTGAACATCTGGGTGGTAGCCGTAAACCGGATGGTGCAATTTATACCGTTGGTAGC
    CCGATTGATTATGGTGTTATTGTTGATACCAAAGCCTATAGCGGTGGTTATAATCTGCCGATTGGTCAGGCAGATGAA
    ATGCAGCGTTATGTGAAAGAAAATCAGACCCGCAACAAACATATTAACCCGAATGAATGGTGGAAAGTTTATCCGAGC
    AGCGTTACCGAGTTTAAATTCCTGTTTGTTAGCGGTCACTTCAAAGGCAACTATAAAGCACAGCTGACCCGTCTGAAT
    CGTAAAACCAATTGTAATGGTGCAGTTCTGAGCGTTGAAGAACTGCTGATTGGTGGTGAAATGATTAAAGCAGGCACC
    CTGACCCTGGAAGAAGTTCGTCGCAAATTTAACAATGGCGAAATCAACTTTGCGGATCCCACCAACCGCGCGAAAGGC
    CTGGAAGCGGTGAGCGTGGCGAGCatgaatttgcttattgataactggattcctgtacgcccgcgaaacggggggaaa
    gtccaaatcataaatattgggcaaattatcgccccggcaaaagatgacgttgaatttcgacatcgcataatgaatccg
    ctcactgaagatgagtttcaacaactcatcgcgccgtggatagatatgttctaccttaatcacgcagaacatcccttt
    atgcagaccaaaggtgtcaaagcaaatgatgtgactccaatggaaaaactgttggctggggtaagcggcgcgacgaat
    tgtgcatttgtcaatcaaccggggcagggtgaagcattatgtggtggatgcactgcgattgcgttattcaaccaggcg
    aatcaggcaccaggttttggtggtggttttaaaagcggtttacgtggaggaacacctgtaacaacgttcgtacgtggg
    atcgatcttcgttcaacggtgttactcaatgtcctcacattacctcgtcttcaaaaacaatttcctaatgaatcacat
    acggaaaaccaacctacctggattaaacctatcaagtccaatgagtctatacctgcttcgtcaattgggtttgtccgt
    ggtctattctggcaaccagcgcatattgaattatgcgatcccattgggattggtaaatgttcttgctgtggacaggaa
    agcaatttgcgttataccggttttcttaaggaaaaatttacctttacagttaatgggctatggcccatccgcattccc
    cttgtctggtaacagtcaagaaaggggaggttgaggaaaaatttcttgctttcaccacctccgcaccatcatggacac
    aaatcagccgagttgtggtagataagattattcaaaatgaaaatggaaatcgcgtggcggcggttgtgaatcaattca
    gaaatattgcgccgcaaagtcctcttgaattgattatggggggatatcgtaataatcaagcatctattcttgaacggc
    gtcatgatgtgttgatgtttaatcaggggtggcaacaatacggcaatgtgataaacgaaatagtgactgttggtttgg
    gatataaaacagccttacgcaaggcgttatatacctttgcagaagggtttaaaaataaagacttcaaaggggccggag
    tctctgttcatgagactgcagaaaggcatttctatcgacagagtgaattattaattcccgatgtactggcgaatgtta
    atttttcccaggctgatgaggtaatagctgatttacgagacaaacttcatcaattgtgtgaaatgctatttaatcaat
    ctgtagctccctatgcacatcatcctaaattaataagcacattagcgcttgcccgcgccacgctatacaaacatttac
    gggagttaaaaccgcaaggagggccatcaaatggctga
    >protein sequence of His6-Dual-monopartite NLS SV40-FokI-(Sharkey-KKR)-Cse1
    (“His6” disclosed as SEQ ID NO: 48)
    [SEQ ID NO: 23]
    MHHHHHHPKKKRKVDPKKKRKVEDPKDMAQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNPTQDRILEMKVMEFF
    MKVYGYRGEHLGGSRKPDGAIYTVGSPIDYGVIVDTKAYSGGYNLPIGQADEMQRYVKENQTRNKHINPNEWWKVYPS
    SVTEFKFLFVSGHFKGNYKAQLTRLNRKTNVNGAVLSVEELLIGGEMIKAGTLTLEEVRRKFNNGEINFADPTNRAKG
    LEAVSVASMNLLIDNWIPVRPRNGGKVQIINLQSLYCSRDQWRLSLPRDDMELAALALLVCIGQIIAPAKDDVEFRHR
    IMNPLTEDEFQQLIAPWIDMFYLNHAEHPFMQTKGVKANDVTPMEKLLAGVSGATNCAFVNQPGQGEALCGGCTAIAL
    FNQANQAPGFGGGFKSGLRGGTPVTTFVRGIDLRSTVLLNVLTLPRLQKQFPNESHTENQPTWIKPIKSNESIPASSI
    GFVRGLFWQPAHIELCDPIGIGKCSCCGQESNLRYTGFLKEKFTFTVNGLWPHPHSPCLVTVKKGEVEEKFLAFTTSA
    PSWTQISRVVVDKIIQNENGNRVAAVVNQFRNIAPQSPLELIMGGYRNNQASILERHHDVLMFNQGWQQYGNVINEIV
    TVGLGYKTALRKALYTFAEGFKNKDFKGAGVSVHETAERHFYRQSELLIPDVLANVNFSQADEVIADLRDKLHQLCEM
    LFNQSVAPYAHHPKLISTLALARATLYKHLRELKPQGGPSNG*
    >nucleotide sequence of His6-Dual-monopartite NLS SV40-FokI (Sharkey-ELD)-Cse1
    (“His6” disclosed as SEQ ID NO: 48)
    [SEQ ID NO: 24]
    ATGcatcaccatcatcaccacCCGAAAAAAAAGCGCAAAGTGGATCCGAAGAAAAAACGTAAAGTTGAAGATCCGAAA
    GACATGGCTCAACTGGTTAAAAGCGAACTGGAAGAGAAAAAAAGTGAACTGCGCCACAAACTGAAATATGTGCCGCAT
    GAATATATCGAGCTGATTGAAATTGCACGTAATCCGACCCAGGATCGTATTCTGGAAATGAAAGTGATGGAATTTTTT
    ATGAAAGTGTACGGCTATCGCGGTGAACATCTGGGTGGTAGCCGTAAACCGGATGGTGCAATTTATACCGTTGGTAGC
    CCGATTGATTATGGTGTTATTGTTGATACCAAAGCCTATAGCGGTGGTTATAATCTGCCGATTGGTCAGGCAGATGAA
    ATGGAACGTTATGTGGAAGAAAATCAGACCCGTGATAAACATCTGAATCCGAATGAATGGTGGAAAGTTTATCCGAGC
    AGCGTTACCGAGTTTAAATTCCTGTTTGTTAGCGGTCACTTCAAAGGCAACTATAAAGCACAGCTGACCCGTCTGAAT
    CATATTACCAATTGTAATGGTGCAGTTCTGAGCGTTGAAGAACTGCTGATTGGTGGTGAAATGATTAAAGCAGGCACC
    CTGACCCTGGAAGAAGTTCGTCGCAAATTTAACAATGGCGAAATCAACTTTGCGGATCCCACCAACCGCGCGAAAGGC
    CTGGAAGCGGTGAGCGTGGCGAGCatgaatttgcttattgataactggattcctgtacgcccgcgaaacggggggaaa
    gtccaaatcataaatctgcaatcgctatactgcagtagagatcagtggcgattaagtttgccccgtgacgatatggaa
    ctggccgctttagcactgctggtttgcattgggcaaattatcgccccggcaaaagatgacgttgaatttcgacatcgc
    ataatgaatccgctcactgaagatgagtttcaacaactcatcgcgccgtggatagatatgttctaccttaatcacgca
    gaacatccctttatgcagaccaaaggtgtcaaagcaaatgatgtgactccaatggaaaaactgttggctggggtaagc
    ggcgcgacgaattgtgcatttgtcaatcaaccggggcagggtgaagcattatgtggtggatgcactgcgattgcgtta
    ttcaaccaggcgaatcaggcaccaggttttggtggtggttttaaaagcggtttacgtggaggaacacctgtaacaacg
    ttcgtacgtgggatcgatcttcgttcaacggtgttactcaatgtcctcacattacctcgtcttcaaaaacaatttcct
    aatgaatcacatacggaaaaccaacctacctggattaaacctatcaagtccaatgagtctatacctgcttcgtcaatt
    gggtttgtccgtggtctattctggcaaccagcgcatattgaattatgcgatcccattgggattggtaaatgttcttgc
    tgtggacaggaaagcaatttgcgttataccggttttcttaaggaaaaatttacctttacagttaatgggctatggccc
    catccgcattccccttgtctggtaacagtcaagaaaggggaggttgaggaaaaatttcttgctttcaccacctccgca
    ccatcatggacacaaatcagccgagttgtggtagataagattattcaaaatgaaaatggaaatcgcgtggcggcggtt
    gtgaatcaattcagaaatattgcgccgcaaagtcctcttgaattgattatggggggatatcgtaataatcaagcatct
    attcttgaacggcgtcatgatgtgttgatgtttaatcaggggtggcaacaatacggcaatgtgataaacgaaatagtg
    actgttggtttgggatataaaacagccttacgcaaggcgttatatacctttgcagaagggtttaaaaataaagacttc
    aaaggggccggagtctctgttcatgagactgcagaaaggcatttctatcgacagagtgaattattaattcccgatgta
    ctggcgaatgttaatttttcccaggctgatgaggtaatagctgatttacgagacaaacttcatcaattgtgtgaaatg
    ctatttaatcaatctgtagctccctatgcacatcatcctaaattaataagcacattagcgcttgcccgcgccacgcta
    tacaaacatttacgggagttaaaaccgcaaggagggccatcaaatggctga
    >protein sequence of His6-Dual-monopartite NLS SV40-FokI-(Sharkey-ELD)-Cse1
    (“His6” disclosed as SEQ ID NO: 48)
    [SEQ ID NO: 25]
    MHHHHHHPKKKRKVDPKKKRKVEDPKDMAQLVKSELEEKKSELRHKLKYVPHEYIELIEIARNPTQDRILEMKVMEFF
    MKVYGYRGEHLGGSRKPDGAIYTVGSPIDYGVIVDTKAYSGGYNLPIGQADEMERYVEENQTRDKHLNPNEWWKVYPS
    SVTEFKFLFVSGHFKGNYKAQLTRLNHITNCNGAVLSVEELLIGGEMIKAGTLTLEEVRRKFNNGEINFADPTNRAKG
    LEAVSVASMNLLIDNWIPVRPRNGGKVQIINLQSLYCSRDQWRLSLPRDDMELAALALLVCIGQIIAPAKDDVEFRHR
    IMNPLTEDEFQQLIAPWIDMFYLNHAEHPFMQTKGVKANDVTPMEKLLAGVSGATNCAFVNQPGQGEALCGGCTAIAL
    FNQANQAPGFGGGFKSGLRGGTPVTTFVRGIDLRSTVLLNVLTLPRLQKQFPNESHTENQPTWIKPIKSNESIPASSI
    GFVRGLFWQPAHIELCDPIGIGKCSCCGQESNLRYTGFLKEKFTFTVNGLWPHPHSPCLVTVKKGEVEEKFLAFTTSA
    PSWTQISRVVVDKIIQNENGNRVAAVVNQFRNIAPQSPLELIMGGYRNNQASILERRHDVLMFNQGWQQYGNVINEIV
    TVGLGYKTALRKALYTFAEGFKNKDFKGAGVSVHETAERHFYRQSELLIPDVLANVNFSQADEVIADLRDKLHQLCEM
    LFNQSVAPYAHHPKLISTLALARATLYKHLRELKPQGGPSNG*
  • DNA Cleavage Assay
  • The specificity and activity of the complexes was tested using an artificially constructed target plasmid as a substrate. This plasmid contains M13 and P7 binding sites on opposing strands such that both FokI domains face each other (see FIG. 11). The distance between the Cascade binding sites varies between 25 and 50 basepairs with 5 bp increments. As the binding sites of Cascade need to be flanked by any of four known PAM sequences (5′-protospacer-CTT/CAT/CTC/CCT-3′ this distance range gives sufficient flexibility to design such a pair for almost any given sequence.
  • The sequences of the target plasmids used are as follows. The number indicated the distance between the M13 and P7 target sites. Protospacers are shown in bold, PAMs underlined:
  • Sequences of the target plasmids. The number indicates the distance between the M13 and P7 target sites. (protospacers in bold, PAMs underlined)
  • >50 bp
    [SEQ ID NO: 26]
    gaattcACAACGGTGAGCAAGTCACTGTTGGCAAGCCAGGATCTGAACA
    ATACCGT CTTGCTTTCGAGCGCTAGCTCTAGAACTAGTCCTCAGCCTAG
    GCCTCGTTCCGAAG CTGTCTTTCGCTGCTGAGGGTGACGATCCCGCATA
    GGCGGCCTTTAACTCggatcc
    >45 bp
    [SEQ ID NO: 27]
    gaattcACAACGGTGAGCAAGTCACTGTTGGCAAGCCAGGATCTGAACA
    ATACCGT CTTTTCGAGCGCTAGCTCTAGAACTAGTCCTCAGCCTAGGCC
    TCGTTCAAG CTGTCTTTCGCTGCTGAGGGTGACGATCCCGCATAGGCGG
    CCTTTAACTCggatcc
    >40 bp
    [SEQ ID NO: 28]
    gaattcACAACGGTGAGCAAGTCACTGTTGGCAAGCCAGGATCTGAACA
    ATACCGT CTTCGAGCGCTAGCTCTAGAACTAGTCCTCAGCCTAGGCCTC
    GAAG CTGTCTTTCGCTGCTGAGGGTGACGATCCCGCATAGGCGGCCTTT
    AACTCggatcc
    >35 bp
    [SEQ ID NO: 29]
    gaattcACAACGGTGAGCAAGTCACTGTTGGCAAGCCAGGATCTGAACA
    ATACCGT CTTGCGCTAGCTCTAGAACTAGTCCTCAGCCTAGGCCTAAG C
    TGTCTTTCGCTGCTGAGGGTGACGATCCCGCATAGGCGGCCTTTAACTC
    ggatcc
    >30 bp
    [SEQ ID NO: 30]
    gaattcACAACGGTGAGCAAGTCACTGTTGGCAAGCCAGGATCTGAACA
    ATACCGT CTTGCTAGCTCTAGAACTAGTCCTCAGCCTAGGAAG CTGTCT
    TTCGCTGCTGAGGGTGACGATCCCGCATAGGCGGCCTTTAACTCggatc
    c
    >25 bp
    [SEQ ID NO: 31]
    gaattcACAACGGTGAGCAAGTCACTGTTGGCAAGCCAGGATCTGAACA
    ATACCGT CTTCTCTAGAACTAGTCCTCAGCCTAGGAAG CTGTCTTTCGC
    TGCTGAGGGTGACGATCCCGCATAGGCGGCCTTTAACTCggatcc
  • Cleavage of the target plasmids was analysed on agarose gels, where negatively supercoiled (nSC) plasmid can be distinguished from linearized- or nicked plasmid. The cleavage site of the CascadeKKR/ELD pair in a target vector was determined by isolating linear cleavage products from an agarose gel and filling in the recessed 3′ ends left by FokI cleavage with the Klenow fragment of E. coli DNA polymerase to create blunt ends. The linear vector was self-ligated, transformed, amplified, isolated and sequenced. Filling in of recessed 3′ ends and re-ligation will lead to extra nucleotides in the sequence that represents the overhang left by FokI cleavage. By aligning the sequence reads to the original sequence, the cleavage sites can be found on a clonal level and mapped. Below, the additional bases incorporated into the sequence after filling in recessed 3′ ends left by FokI cleavage are underlined:
  • Figure US20160186152A1-20160630-C00001
  • Reading from top to bottom, the 5′-3′ sequences above are SEQ ID NOs: 32-35, respectively.
  • Cleavage of a Target Locus in Human Cells
  • The human CCR5 gene encodes the C—C chemokine receptor type 5 protein, which serves as the receptor for the human immunodeficiency virus (HIV) on the surface of white blood cells. The CCR5 gene is targeted using a pair of CascadeKKR/ELD nucleases in addition to an artificial GFP locus. A suitable binding site pair is selected on the coding region of CCR5. Two separate CRISPR arrays containing uniform spacers targeting each of the binding sites are constructed using DNA synthesis (Geneart).
  • The human CCR5 target gene selection and CRISPR designs used are as follows:
  • >Part of genomic human CCR5 sequence, containing
    whole ORF (position 347-1446).
    [SEQ ID NO: 36]
    GGTGGAACAAGATGGATTATCAAGTGTCAAGTCCAATCTATGACATCA
    ATTATTATACATCGGAGCCCTGCCAAAAAATCAATGTGAAGCAAATCG
    CAGCCCGCCTCCTGCCTCCGCTCTACTCACTGGTGTTCATCTTTGGTT
    TTGTGGGCAACATGCTGGTCATCCTCATCCTGATAAACTGCAAAAGGC
    TGAAGAGCATGACTGACATCTACCTGCTCAACCTGGCCATCTCTGACC
    TGTTTTTCCTTCTTACTGTCCCCTTCTGGGCTCACTATGCTGCCGCCC
    AGTGGGACTTTGGAAATACAATGTGTCAACTCTTGACAGGGCTCTATT
    TTATAGGCTTCTTCTCTGGAATCTTCTTCATCATCCTCCTGACAATCG
    ATAGGTACCTGGCTGTCGTCCATGCTGTGTTTG CTTTAAAAGCCAGGA
    CGGTCACCTTTGGGGTGGTGACAAG TGTGATCACTTGGGTGGTGGCTG
    TGTTTGCGTCTCTCCCAGGAATCATCTTTACCAGATCTCAAAAAGAAG
    GTCTTCATTACACCTGCAGCTCTCATTTTCCATACAGTCAGTATCAAT
    TCTGGAAGAATTTCCAGACATTAAAGATAGTCATCTTGGGGCTGGTCC
    TGCCGCTGCTTGTCATGGTCATCTGCTACTCGGGAATCCTAAAAACTC
    TGCTTCGGTGTCGAAATGAGAAGAAGAGGCACAGGGCTGTGAGGCTTA
    TCTTCACCATCATGATTGTTTATTTTCTCTTCTGGGCTCCCTACAACA
    TTGTCCTTCTCCTGAACACCTTCCAGGAATTCTTTGGCCTGAATAATT
    GCAGTAGCTCTAACAGGTTGGACCAAGCTATGCAGGTGACAGAGACTC
    TTGGGATGACGCACTGCTGCATCAACCCCATCATCTATGCCTTTGTCG
    GGGAGAAGTTCAGAAACTACCTCTTAGTCTTCTTCCAAAAGCACATTG
    CCAAACGCTTCTGCAAATGCTGTTCTATTTTCCAGCAAGAGGCTCCCG
    AGCGAGCAAGCTCAGTTTACACCCGATCCACTGGGGAGCAGGAAATAT
    CTGTGGGCTTGTGACACGGACTCAAGTGGGCTGGTGACCCAGTC
    Red1/2: chosen target sites (distance: 34 bp, PAM
    5′-CTT-3′). “Red 1 is first appearing underlined
    sequence in the above. Red2 is the second
    underlined sequence.
    >CRISPR array red1 (italics = spacers, bold =
    repeats)
    [SEQ ID NO: 37]
    ccatggTAATACGACTCACTATAGGGAGAATTAGCTGATCTTTAATAA
    TAAGGAAATGTTACATTAAGGTTGGTGGGTTGTTTTTATGGGAAAAAA
    TGCTTTAAGAACAAATGTATACTTTTAGAGAGTTCCCCGCGCCAGCGG
    GGATAAACCG CAAACACAGCATGGACGACAGCCAGGTACCTA GAGTTC
    CCCGCGCCAGCGGGGATAAACCG CAAACACAGCATGGACGACAGCCAG
    GTACCTA GAGTTCCCCGCGCCAGCGGGGATAAACCG CAAACACAGCAT
    GGACGACAGCCAGGTACCTA GAGTTCCCCGCGCCAGCGGGGATAAACC
    GAAAACAAAAGGCTCAGTCGGAAGACTGGGCCTTTTGTTTTAACCCCT
    TGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGggtacc
    >CRISPR array red2 (italics: spacers, bold:
    repeats)
    [SEQ ID NO: 38]
    ccatggTAATACGACTCACTATAGGGAGAATTAGCTGATCTTTAATAA
    TAAGGAAATGTTACATTAAGGTTGGTGGGTTGTTTTTATGGGAAAAAA
    TGCTTTAAGAACAAATGTATACTTTTAGAGAGTTCCCCGCGCCAGCGG
    GGATAAACCG TGTGATCACTTGGGTGGTGGCTGTGTTTGCGT GAGTTC
    CCCGCGCCAGCGGGGATAAACCG TGTGATCACTTGGGTGGTGGCTGTG
    TTTGCGT GAGTTCCCCGCGCCAGCGGGGATAAACCG TGTGATCACTTG
    GGTGGTGGCTGTGTTTGCGT GAGTTCCCCGCGCCAGCGGGGATAAACC
    GAAAACAAAAGGCTCAGTCGGAAGACTGGGCCTTTTGTTTTAACCCCT
    TGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGggtacc

    Delivery of CascadeKKR/ELD into the Nucleus of Human Cells
  • Cascade is very stable as a multi-subunit protein-RNA complex and is easily produced in mg quantities in E. coli. Transfection or micro-injection of the complex in its intact form as purified from E. coli is used as methods of delivery (see FIG. 12). As shown in FIG. 12, Cascade-FokI nucleases are purified from E. coli and encapsulated in protein transfection vesicles. These are then fused with the cell membrane of human HepG2 cells releasing the nucleases in the cytoplasm (step 2). NLS sequences are then be recognized by importin proteins, which facilitate nucleopore passage (step 3). CascadeKKR (open rectangle) and CascadeELD (filled rectangle) will then find and cleave their target site (step 4.), inducing DNA repair pathways that will alter the target site leading to desired changes. CascadeKKR/ELD nucleases need to act only once and require no permanent presence in the cell encoded on DNA.
  • To deliver Cascade into human cells, protein transfection reagents are used from various sources including Pierce, NEB, Fermentas and Clontech. These reagents have recently been developed for the delivery of antibodies, and are useful to transfect a broad range of human cell lines with efficiencies up to 90%. Human HepG2 cells are transfected. Also, other cell lines including CHO-K1, COS-7, HeLa, and non-embryonic stem cells, are transfected.
  • To import the CascadeKKR/ELD nuclease pair into the nucleus, a tandem monopartite nuclear localisation signal (NLS) from the large T-antigen of simian virus 40 (SV40) is fused to the N-terminus of FokI. This ensures import of only intact CascadeELD/KKR into the nucleus. (The nuclear pore complex translocates RNA polymerases (550 kDa) and other large protein complexes). As a check prior to transformations, the nuclease activity of the CascadeKKR/ELD nuclease pair is checked in vitro using purified complexes and CCR5 PCR amplicons to exclude transfecting non-productive CascadeKKR/ELD nuclease pairs.
  • Surveyor Assay
  • Transfected cells are cultivated and passaged for several days. The efficiency of in vivo target DNA cleavage is then assessed by using the Surveyor assay of Guschin, D. Y., et at (2010) Methods Mol. Biol., 649: 247-256. Briefly, PCR amplicons of the target DNA locus will be mixed 1:1 with PCR amplicons from untreated cells. These are heated and allowed to anneal, giving rise to mismatches at target sites that have been erroneously repaired by NHEJ. A mismatch nuclease is then used to cleave only mismatched DNA molecules, giving a maximum of 50% of cleavage when target DNA cleavage by CascadeKKR/ELD is complete. This procedure was then followed up by sequencing of the target DNA amplicons of treated cells. The assay allows for rapid assessment and optimization of the delivery procedure.
  • Production of Cascade-Nuclease Pairs
  • The Cascade-nuclease complexes were constructed as explained above. Affinity purification from E. coli using the StrepII-tagged Cse2 subunit yields a complex with the expected stoichiometry when compared to native Cascade. Referring to FIG. 13, this shows the stoichiometry of native Cascade (1), CascadeKKR with P7 CrRNA and CascadeELD with M13 CrRNA 24 h after purification using only Streptactin. Bands in native Cascade (1) are from top to bottom: Cse1, Cas7, Cas5, Cas6e, Cse2. CascadeKKR/ELD show the FokI-Cse1 fusion band and an additional band representing Cse1 with a small part of FokI as a result of proteolytic degradation.
  • Apart from an intact FokI-Cse1 fusion protein, we observed that a fraction of the FokI-Cse1-fusion protein is proteolytically cleaved, resulting in a Cse1 protein with only the linker and a small part of FokI attached to it (as confirmed by Mass Spectrometry, data not shown). In most protein isolations the fraction of degraded fusion protein is approximately 40%. The isolated protein is stably stored in the elution buffer (20 mM HEPES pH 7.5, 75 mM NaCl, 1 mM DTT, 4 mM desthiobiotin) with additional 0.1 % Tween 20 and 50% glycerol at −20° C. Under these storage conditions, integrity and activity of the complex have been found stable for at least three weeks (data not shown).
  • Introduction of a his6-Tag (SEQ ID NO: 48) and NLS to the Cascade-Nuclease
  • The Cascade nuclease fusion design was modified to incorporate a Nucleolar Localization Signal (NLS) to enable transport into the nucleus of eukaryotic cells. For this a tandem monopartite NLS from the large T-antigen of Simian Virus SV40 (sequence: PKKKRKVDPKKKRKV) (SEQ ID NO: 49) was translationally fused to the N-terminus of the FokI-Cse1 fusion protein, directly preceded by a His6-tag at the N-terminus. The His6-tag (sequence: MHHHHHH) (“His6” disclosed as SEQ ID NO: 48 and “MHHHHHH” disclosed as SEQ ID NO: 50) allows for an additional Ni2+-resin affinity purification step after StrepII purification. This additional step ensures the isolation of only full-length Cascade-nuclease fusion complex, and increases the efficiency of cleavage by eliminating the binding of non-intact Cascade complexes to the target site forming an unproductive nuclease pair.
  • In Vitro Cleavage Assay
  • CascadeKRR/ELD activity and specificity was assayed in vitro as described above. FIG. 14A shows plasmids with distances between protospacers of 25-50 bp (5 bp increments, lanes 1-6) incubated with CascadeKKR/ELD for 30 minutes at 37° C. Lane 10 contains the target plasmid in its three possible topologies: the lowest band represents the initial, negatively supercoiled (nSC) form of the plasmid, the middle band represents the linearized form (cleaved by XbaI), whilst the upper band represents the open circular (OC) form (after nicking with Nt.BbrCI). Lane 7 shows incubation of a plasmid with both binding sites removed (negative control). Therefore FIG. 14A shows a typical cleavage assay using various target plasmids in which the binding sites are separated by 25 to 50 base pairs in 5 bp increments (lanes 1 to 6). These plasmids with distances of 25-50 bp were incubated with CascadeKKR/ELD carrying anti P7 and M13 crRNA respectively. A plasmid containing no binding sites served as a control (lane 7). The original plasmid exists in negatively supercoiled form (nSC, control lane 8), and nicked or linearized products are clearly distinguishable. Upon incubation a linear cleavage product is formed when binding sites were separated by 30, 35 and 40 base pairs ( lanes 2, 3, 4). At 25, 45 and 50 base pairs distance ( lanes 1, 5, 6), the target plasmid appeared to be incompletely cleaved leading to the nicked form (OC). These results show the best cleavage in plasmids with distances between 30 and 40 bp, giving sufficient flexibility when designing a crRNA pair for any given locus. Both shorter and longer distances result in increased nicking activity while creating less DSBs. There is very little activity on a plasmid where the two protospacers have been removed, showing target specificity (lane 7).
  • Cleavage Conditions
  • To assess the optimal buffer conditions for cleavage assays, and to estimate whether activity of the complex is expected at physiological conditions, the following two buffers were selected: (1) NEB4 (New England Biolabs, 50 mM potassium acetate, 20 mM Tris-acetate, 10 mM magnesium acetate, 1 mM dithiothreitol, pH 7.9) and (2) Buffer 0 (Fermentas, 50 mM Tris-HCl, 10 mM MgCl2, 100 mM NaCl, 0.1 mg/mL BSA, pH 7.5). Of the two, NEB4 is recommended for optimal activity of the commercial intact FokI enzyme. Buffer 0 was chosen from a quick screen to give good activity and specificity (data not shown). FIG. 14B shows incubation with different buffers and different incubation times. Lanes 1-4 have been incubated with Fermentas Buffer 0 ( lane 1, 2 for 15 minutes, lane 3, 4 for 30 minutes), lanes 5, 6 have been incubated with NEB4 (30 minutes). Lanes 1, 3, 5 used the target plasmid with 35 bp spacing, lanes 2, 4, 6 used the non-target plasmid (no binding sites). Lanes 7, 8 have been incubated with only CascadeKKR or CascadeELD respectively (buffer 0). Lane 9 is the topology marker as in (A). Lane 10 and 11 show the target and non-target plasmid incubated without addition of Cascade. Therefore in FIG. 14B, activity was tested on the target plasmid with 35 base pairs distance ( lane 1, 3, 5) and a non-target control plasmid ( lane 2, 4, 6). There was a high amount of unspecific nicking and less cleavage in NEB4 (lane 5,6), whilst buffer 0 shows only activity in the target plasmid with a high amount of specific cleavage and little nicking (lane 1-4). The difference is likely caused by the NaCl concentration in buffer 0, higher ionic strength weakens protein-protein interactions, leading to less nonspecific activity. Incubation of 15 or 30 minutes shows little difference in both target and non-target plasmid ( lane 1,2 or 3,4 respectively). Addition of only one type of Cascade (P7KKR or M13ELD) does not result in cleavage activity (lane 7, 8) as expected. This experiment shows that specific Cascade nuclease activity by a designed pair occurs when the NaCl concentration is at least 100 mM, which is near the physiological saline concentration inside cells (137 mM NaCl). The Cascade nuclease pair is expected to be fully active in vivo, in eukaryotic cells, while displaying negligible off-target cleavage activity.
  • Cleavage Site
  • The site of cleavage in the target plasmid with a spacing of 35 bp (pTarget35) was determined. FIG. 15 shows how sequencing reveals up- and downstream cleavage sites by CascadeKKR/ELD in the target plasmid with 35 base pair spacing. In FIG. 15A) is shown the target region within pTarget35 with annotated potential cleavage sites. Parts of the protospacers are indicated in red and blue. B) The bar chart shows four different cleavage patterns and their relative abundance within sequenced clones. The blue bars represent the generated overhang, while the left and right border of each bar represents the left and right cleavage site (see B for annotation).
  • FIG. 15A shows the original sequence of pTarget35, with numbered cleavage sites from −7 to +7 where 0 lies in the middle between the two protospacers (indicated in red and blue). Seventeen clones were sequenced and these all show cleavage around position 0, creating varying overhangs between 3 and 5 bp (see FIG. 15B). Overhangs of 4 are most abundant (cumulatively 88%), while overhangs of 3 and 5 occur only once (6% each). The cleavage occurred exactly as expected with no clones showing off target cleavage.
  • Cleaving a Target Locus in Human Cells.
  • CascadeKKR/ELD nucleases were successfully modified to contain an N-terminal His6-tag (SEQ ID NO: 48) followed by a dual mono-partite Nucleolar Localisation Signal. These modified Cascade nuclease fusion proteins were co-expressed with either one of two synthetically constructed CRISPR arrays, each targeting a binding site in the human CCR5 gene. First the activity of this new nuclease pair is validated in vitro by testing the activity on a plasmid containing this region of the CCR5 gene. The nuclease pair is transfected to a human cell line, e.g. HeLa cell line. Efficiency of target cleavage is assessed using the Surveyor assay as described above.

Claims (19)

1-46. (canceled)
47. A composition comprising a designed fusion of (i) at least one clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein subunit, and (ii) a nuclease, or a mutant or an active portion thereof.
48. The composition of claim 47, wherein the nuclease is a ribonuclease, or a mutant or an active portion thereof.
49. The composition of claim 48, wherein the ribonuclease is an endonuclease, a 3′ exonuclease or a 5′ exonuclease.
50. The composition of claim 47, wherein the CRISPR-associated protein subunit is selected from the group consisting of Cas6, Cas5, Cse1, Cse2 and Cas7.
51. The composition of claim 50, wherein the CRISPR-associated protein subunit is Cse1.
52. The composition of claim 47, wherein the nuclease is an endonuclease, or mutant or an active portion thereof.
53. The composition of claim 52, wherein the endonuclease comprises FokI or a modified FokI.
54. The composition of claim 53, wherein the modified FokI is KKR Sharkey or ELD Sharkey or a combination thereof.
55. The composition of claim 47, wherein the nuclease is a FokI KKR Sharkey, further comprising a second composition comprising a second designed fusion of (i) at least one clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein subunit, and (ii) a nuclease, or a mutant or an active portion thereof, wherein the nuclease is a FokI ELD Sharkey.
56. The composition of claim 47, further comprising a second composition comprising a second designed fusion of (i) at least one clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein subunit, and (ii) a nuclease, or a mutant or an active portion thereof.
57. The composition of claim 50, wherein the nuclease comprises FokI or a modified FokI.
58. The composition of claim 57, wherein the modified FokI is KKR Sharkey or ELD Sharkey or a combination thereof.
59. The composition of claim 51, wherein the nuclease comprises FokI or a modified FokI.
60. The composition of claim 59, wherein the modified FokI is KKR Sharkey or ELD Sharkey or a combination thereof.
61. The composition of claim 47, wherein the designed fusion further comprises a linker polypeptide between (i) the clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein subunit, and (ii) the nuclease, or the mutant or the active portion thereof.
62. The composition of claim 47, wherein the clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein subunit and (ii) the nuclease, or the mutant or the active portion thereof, are covalently linked by chemical synthesis.
63. One or more nucleic acid molecules encoding the composition of claim 47.
64. One or more expression vectors comprising the nucleic acid molecules of claim 63.
US14/997,467 2011-12-30 2016-01-15 Modified cascade ribonucleoproteins and uses thereof Abandoned US20160186152A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/997,467 US20160186152A1 (en) 2011-12-30 2016-01-15 Modified cascade ribonucleoproteins and uses thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GB1122458.1 2011-12-30
GB201122458A GB201122458D0 (en) 2011-12-30 2011-12-30 Modified cascade ribonucleoproteins and uses thereof
PCT/EP2012/076674 WO2013098244A1 (en) 2011-12-30 2012-12-21 Modified cascade ribonucleoproteins and uses thereof
US201414240735A 2014-02-24 2014-02-24
US14/326,099 US20150024499A1 (en) 2011-12-30 2014-07-08 Modified cascade ribonucleoproteins and uses thereof
US14/997,467 US20160186152A1 (en) 2011-12-30 2016-01-15 Modified cascade ribonucleoproteins and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/326,099 Continuation US20150024499A1 (en) 2011-12-30 2014-07-08 Modified cascade ribonucleoproteins and uses thereof

Publications (1)

Publication Number Publication Date
US20160186152A1 true US20160186152A1 (en) 2016-06-30

Family

ID=45695084

Family Applications (8)

Application Number Title Priority Date Filing Date
US14/240,735 Abandoned US20140294773A1 (en) 2011-12-30 2012-12-21 Modified cascade ribonucleoproteins and uses thereof
US14/326,099 Abandoned US20150024499A1 (en) 2011-12-30 2014-07-08 Modified cascade ribonucleoproteins and uses thereof
US14/997,474 Active US9885026B2 (en) 2011-12-30 2016-01-15 Modified cascade ribonucleoproteins and uses thereof
US14/997,467 Abandoned US20160186152A1 (en) 2011-12-30 2016-01-15 Modified cascade ribonucleoproteins and uses thereof
US15/802,413 Active 2033-07-26 US10435678B2 (en) 2011-12-30 2017-11-02 Modified cascade ribonucleoproteins and uses thereof
US16/554,225 Active US10711257B2 (en) 2011-12-30 2019-08-28 Modified cascade ribonucleoproteins and uses thereof
US16/914,203 Active US10954498B2 (en) 2011-12-30 2020-06-26 Modified cascade ribonucleoproteins and uses thereof
US17/179,215 Active 2034-01-26 US11939604B2 (en) 2011-12-30 2021-02-18 Modified cascade ribonucleoproteins and uses thereof

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US14/240,735 Abandoned US20140294773A1 (en) 2011-12-30 2012-12-21 Modified cascade ribonucleoproteins and uses thereof
US14/326,099 Abandoned US20150024499A1 (en) 2011-12-30 2014-07-08 Modified cascade ribonucleoproteins and uses thereof
US14/997,474 Active US9885026B2 (en) 2011-12-30 2016-01-15 Modified cascade ribonucleoproteins and uses thereof

Family Applications After (4)

Application Number Title Priority Date Filing Date
US15/802,413 Active 2033-07-26 US10435678B2 (en) 2011-12-30 2017-11-02 Modified cascade ribonucleoproteins and uses thereof
US16/554,225 Active US10711257B2 (en) 2011-12-30 2019-08-28 Modified cascade ribonucleoproteins and uses thereof
US16/914,203 Active US10954498B2 (en) 2011-12-30 2020-06-26 Modified cascade ribonucleoproteins and uses thereof
US17/179,215 Active 2034-01-26 US11939604B2 (en) 2011-12-30 2021-02-18 Modified cascade ribonucleoproteins and uses thereof

Country Status (26)

Country Link
US (8) US20140294773A1 (en)
EP (2) EP2798060A1 (en)
JP (1) JP6408914B2 (en)
KR (1) KR101889589B1 (en)
CN (2) CN105732816B (en)
AU (1) AU2012360975B2 (en)
BR (1) BR112014016228B1 (en)
CA (1) CA2862018C (en)
CY (1) CY1120538T1 (en)
DE (1) DE212012000234U1 (en)
DK (1) DK3091072T3 (en)
ES (1) ES2689256T3 (en)
GB (3) GB201122458D0 (en)
HR (1) HRP20181150T1 (en)
HU (1) HUE039617T2 (en)
IL (1) IL233399A0 (en)
IN (1) IN2014DN05937A (en)
LT (1) LT3091072T (en)
MX (1) MX364830B (en)
PL (1) PL3091072T3 (en)
PT (1) PT3091072T (en)
RS (1) RS57604B1 (en)
RU (1) RU2014127702A (en)
SG (1) SG11201403713QA (en)
SI (1) SI3091072T1 (en)
WO (1) WO2013098244A1 (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9856497B2 (en) 2016-01-11 2018-01-02 The Board Of Trustee Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
US9888673B2 (en) 2014-12-10 2018-02-13 Regents Of The University Of Minnesota Genetically modified cells, tissues, and organs for treating disease
WO2018148647A3 (en) * 2017-02-10 2018-09-20 Lajoie Marc Joseph Genome editing reagents and their use
US10166255B2 (en) 2015-07-31 2019-01-01 Regents Of The University Of Minnesota Intracellular genomic transplant and methods of therapy
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10336807B2 (en) 2016-01-11 2019-07-02 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of immunotherapy
KR102023577B1 (en) * 2018-03-27 2019-09-24 (주)지플러스 생명과학 Pharmaceutical composition for treating cancers comprising guide rna and endonuclease
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
WO2019186275A1 (en) * 2018-03-27 2019-10-03 G+Flas Life Sciences Sequence-specific in vivo cell targeting
WO2020072253A1 (en) * 2018-10-01 2020-04-09 North Carolina State University Recombinant type i crispr-cas system and uses thereof for screening for variant cells
US10711267B2 (en) 2018-10-01 2020-07-14 North Carolina State University Recombinant type I CRISPR-Cas system
US10787654B2 (en) 2014-01-24 2020-09-29 North Carolina State University Methods and compositions for sequence guiding Cas9 targeting
US10912797B2 (en) 2016-10-18 2021-02-09 Intima Bioscience, Inc. Tumor infiltrating lymphocytes and methods of therapy
WO2021092210A1 (en) * 2019-11-06 2021-05-14 Locus Biosciences, Inc. Phage compositions comprising crispr-cas systems and methods of use thereof
US11098325B2 (en) 2017-06-30 2021-08-24 Intima Bioscience, Inc. Adeno-associated viral vectors for gene therapy
US11155823B2 (en) 2015-06-15 2021-10-26 North Carolina State University Methods and compositions for efficient delivery of nucleic acids and RNA-based antimicrobials
US11236313B2 (en) 2016-04-13 2022-02-01 Editas Medicine, Inc. Cas9 fusion molecules, gene editing systems, and methods of use thereof
US11261451B2 (en) 2015-05-29 2022-03-01 North Carolina State University Methods for screening bacteria, archaea, algae, and yeast using CRISPR nucleic acids
US11286480B2 (en) 2015-09-28 2022-03-29 North Carolina State University Methods and compositions for sequence specific antimicrobials
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11439712B2 (en) 2014-04-08 2022-09-13 North Carolina State University Methods and compositions for RNA-directed repression of transcription using CRISPR-associated genes
US11499169B2 (en) 2013-10-30 2022-11-15 North Carolina State University Compositions and methods related to a type-II CRISPR-Cas system in Lactobacillus buchneri
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
US11597924B2 (en) 2016-03-25 2023-03-07 Editas Medicine, Inc. Genome editing systems comprising repair-modulating enzyme molecules and methods of their use
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US11753651B2 (en) 2014-08-28 2023-09-12 North Carolina State University Cas9 proteins and guiding features for DNA targeting and genome editing
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
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation

Families Citing this family (178)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7790756B2 (en) 2006-10-11 2010-09-07 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoproliferative diseases and other proliferative diseases
BR112012028805A2 (en) 2010-05-10 2019-09-24 The Regents Of The Univ Of California E Nereus Pharmaceuticals Inc endoribonuclease compositions and methods of use thereof.
US10323236B2 (en) 2011-07-22 2019-06-18 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
IN2014MN00974A (en) 2011-12-16 2015-04-24 Targetgene Biotechnologies Ltd
US11021737B2 (en) 2011-12-22 2021-06-01 President And Fellows Of Harvard College Compositions and methods for analyte detection
WO2014163886A1 (en) 2013-03-12 2014-10-09 President And Fellows Of Harvard College Method of generating a three-dimensional nucleic acid containing matrix
GB201122458D0 (en) 2011-12-30 2012-02-08 Univ Wageningen Modified cascade ribonucleoproteins and uses thereof
WO2013163628A2 (en) 2012-04-27 2013-10-31 Duke University Genetic correction of mutated genes
ES2960803T3 (en) 2012-05-25 2024-03-06 Univ California Methods and compositions for RNA-directed modification of target DNA and for modulation of RNA-directed transcription
ES2926021T3 (en) 2012-10-23 2022-10-21 Toolgen Inc Composition for cleaving a target DNA comprising a target DNA-specific guide RNA and Cas protein-encoding nucleic acid or Cas protein, and use thereof
EP3617309A3 (en) 2012-12-06 2020-05-06 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
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
IL239344B1 (en) 2012-12-12 2024-02-01 Broad Inst Inc Engineering of systems, methods and optimized guide compositions for sequence manipulation
US20140310830A1 (en) 2012-12-12 2014-10-16 Feng Zhang CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
WO2014093709A1 (en) 2012-12-12 2014-06-19 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
SG10201912328UA (en) 2012-12-12 2020-02-27 Broad Inst Inc Delivery, Engineering and Optimization of Systems, Methods and Compositions for Sequence Manipulation and Therapeutic Applications
ES2576128T3 (en) 2012-12-12 2016-07-05 The Broad Institute, Inc. Modification by genetic technology and optimization of systems, methods and compositions for the manipulation of sequences with functional domains
CN113355357A (en) 2012-12-12 2021-09-07 布罗德研究所有限公司 Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
IL308158A (en) 2012-12-17 2023-12-01 Harvard College Rna-guided human genome engineering
AU2014235794A1 (en) 2013-03-14 2015-10-22 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids
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
WO2014204578A1 (en) 2013-06-21 2014-12-24 The General Hospital Corporation Using rna-guided foki nucleases (rfns) to increase specificity for rna-guided genome editing
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
US9902973B2 (en) 2013-04-11 2018-02-27 Caribou Biosciences, Inc. Methods of modifying a target nucleic acid with an argonaute
EP3603679B1 (en) 2013-06-04 2022-08-10 President and Fellows of Harvard College Rna-guided transcriptional regulation
US9267135B2 (en) 2013-06-04 2016-02-23 President And Fellows Of Harvard College RNA-guided transcriptional regulation
EP3725885A1 (en) 2013-06-17 2020-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions methods, screens and applications thereof
AU2014281028B2 (en) 2013-06-17 2020-09-10 Massachusetts Institute Of Technology Delivery and use of the CRISPR-Cas systems, vectors and compositions for hepatic targeting and therapy
WO2014204724A1 (en) 2013-06-17 2014-12-24 The Broad Institute Inc. Delivery, engineering and optimization of tandem guide systems, methods and compositions for sequence manipulation
EP3011034B1 (en) 2013-06-17 2019-08-07 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components
WO2014204725A1 (en) 2013-06-17 2014-12-24 The Broad Institute Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
SG10201913015XA (en) 2013-07-10 2020-02-27 Harvard College Orthogonal cas9 proteins for rna-guided gene regulation and editing
US9663782B2 (en) 2013-07-19 2017-05-30 Larix Bioscience Llc Methods and compositions for producing double allele knock outs
US11060083B2 (en) 2013-07-19 2021-07-13 Larix Bioscience Llc Methods and compositions for producing double allele knock outs
US10563225B2 (en) 2013-07-26 2020-02-18 President And Fellows Of Harvard College Genome engineering
CN103388006B (en) * 2013-07-26 2015-10-28 华东师范大学 A kind of construction process of site-directed point mutation
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
US20150044772A1 (en) * 2013-08-09 2015-02-12 Sage Labs, Inc. Crispr/cas system-based novel fusion protein and its applications in genome editing
AU2014308899B2 (en) 2013-08-22 2020-11-19 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
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
SG10201801658XA (en) * 2013-08-29 2018-03-28 Univ Temple Methods and compositions for rna-guided treatment of hiv infection
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9340799B2 (en) 2013-09-06 2016-05-17 President And Fellows Of Harvard College MRNA-sensing switchable gRNAs
DE202014010413U1 (en) 2013-09-18 2015-12-08 Kymab Limited Cells and organisms
WO2015065964A1 (en) 2013-10-28 2015-05-07 The Broad Institute Inc. Functional genomics using crispr-cas systems, compositions, methods, screens and applications thereof
US9074199B1 (en) 2013-11-19 2015-07-07 President And Fellows Of Harvard College Mutant Cas9 proteins
US10787684B2 (en) 2013-11-19 2020-09-29 President And Fellows Of Harvard College Large gene excision and insertion
CN111206032A (en) 2013-12-12 2020-05-29 布罗德研究所有限公司 Delivery, use and therapeutic applications of CRISPR-CAS systems and compositions for genome editing
WO2015089364A1 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Crystal structure of a crispr-cas system, and uses thereof
US9840699B2 (en) 2013-12-12 2017-12-12 President And Fellows Of Harvard College Methods for nucleic acid editing
WO2015089427A1 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Crispr-cas systems and methods for altering expression of gene products, structural information and inducible modular cas enzymes
CA2932472A1 (en) 2013-12-12 2015-06-18 Massachusetts Institute Of Technology Compositions and methods of use of crispr-cas systems in nucleotide repeat disorders
EP3080271B1 (en) 2013-12-12 2020-02-12 The Broad Institute, Inc. Systems, methods and compositions for sequence manipulation with optimized functional crispr-cas systems
AU2014368982B2 (en) 2013-12-19 2021-03-25 Amyris, Inc. Methods for genomic integration
EP3114227B1 (en) 2014-03-05 2021-07-21 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating usher syndrome and retinitis pigmentosa
CN111471675A (en) 2014-03-05 2020-07-31 国立大学法人神户大学 Method for modifying genome sequence of nucleic acid base for specifically converting target DNA sequence, and molecular complex used therefor
US11141493B2 (en) 2014-03-10 2021-10-12 Editas Medicine, Inc. Compositions and methods for treating CEP290-associated disease
WO2015138510A1 (en) 2014-03-10 2015-09-17 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
WO2015148863A2 (en) 2014-03-26 2015-10-01 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating sickle cell disease
US11318206B2 (en) 2014-03-28 2022-05-03 Aposense Ltd Compounds and methods for trans-membrane delivery of molecules
AU2015237746B2 (en) * 2014-03-28 2020-01-30 Aposense Ltd. Compounds and methods for trans-membrane delivery of molecules
KR102598819B1 (en) 2014-06-23 2023-11-03 더 제너럴 하스피탈 코포레이션 Genomewide unbiased identification of dsbs evaluated by sequencing (guide-seq)
MX2017000422A (en) 2014-07-11 2017-05-01 Du Pont Compositions and methods for producing plants resistant to glyphosate herbicide.
EP3177718B1 (en) 2014-07-30 2022-03-16 President and Fellows of Harvard College Cas9 proteins including ligand-dependent inteins
ES2730378T3 (en) 2014-08-27 2019-11-11 Caribou Biosciences Inc Procedures to increase the efficiency of the modification mediated by Cas9
CN108064129A (en) 2014-09-12 2018-05-22 纳幕尔杜邦公司 The generation in the site-specific integration site of complex character locus and application method in corn and soybean
CN107250148B (en) 2014-12-03 2021-04-16 安捷伦科技有限公司 Guide RNAs with chemical modifications
EP3889260A1 (en) 2014-12-12 2021-10-06 The Broad Institute, Inc. Protected guide rnas (pgrnas)
NZ733702A (en) 2015-01-28 2022-04-29 Caribou Biosciences Inc Crispr hybrid dna/rna polynucleotides and methods of use
EP3858990A1 (en) 2015-03-03 2021-08-04 The General Hospital Corporation Engineered crispr-cas9 nucleases with altered pam specificity
KR101675023B1 (en) * 2015-03-26 2016-11-10 한국생명공학연구원 Fok Composition for specific editing of a target gene in vivo using a target gene-specific nucleic acid probe and dimerized Fokendonuclease and uses thereof
EP3553178A1 (en) 2015-03-27 2019-10-16 E. I. du Pont de Nemours and Company Soybean u6 small nuclear rna gene promoters and their use in constitutive expression of small rna genes in plants
KR102648489B1 (en) 2015-04-06 2024-03-15 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Chemically modified guide RNA for CRISPR/CAS-mediated gene regulation
SG11201708653RA (en) 2015-04-24 2017-11-29 Editas Medicine Inc Evaluation of cas9 molecule/guide rna molecule complexes
AU2016270870A1 (en) 2015-06-02 2018-01-04 Monsanto Technology Llc Compositions and methods for delivery of a polynucleotide into a plant
WO2016196887A1 (en) 2015-06-03 2016-12-08 Board Of Regents Of The University Of Nebraska Dna editing using single-stranded dna
WO2016205759A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of cas9 orthologs and variants for sequence manipulation
CN109536474A (en) 2015-06-18 2019-03-29 布罗德研究所有限公司 Reduce the CRISPR enzyme mutant of undershooting-effect
AU2016297167A1 (en) * 2015-07-23 2018-02-22 Mayo Foundation For Medical Education And Research Editing mitochondrial DNA
US9580727B1 (en) 2015-08-07 2017-02-28 Caribou Biosciences, Inc. Compositions and methods of engineered CRISPR-Cas9 systems using split-nexus Cas9-associated polynucleotides
WO2017040348A1 (en) 2015-08-28 2017-03-09 The General Hospital Corporation Engineered crispr-cas9 nucleases
US9926546B2 (en) 2015-08-28 2018-03-27 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
US9512446B1 (en) 2015-08-28 2016-12-06 The General Hospital Corporation Engineered CRISPR-Cas9 nucleases
ES2938623T3 (en) 2015-09-09 2023-04-13 Univ Kobe Nat Univ Corp Method for converting a genome sequence of a gram-positive bacterium by specific nucleic acid base conversion of a targeted DNA sequence and the molecular complex used therein
CA2998087A1 (en) * 2015-09-09 2017-03-16 National University Corporation Kobe University Method for modifying genome sequence that specifically converts nucleobase of targeted dna sequence, and molecular complex used in said method
EP3347467B1 (en) 2015-09-11 2021-06-23 The General Hospital Corporation Full interrogation of nuclease dsbs and sequencing (find-seq)
AU2016331185A1 (en) 2015-09-30 2018-04-26 The General Hospital Corporation Comprehensive in vitro reporting of cleavage events by sequencing (CIRCLE-seq)
CN105331627B (en) * 2015-09-30 2019-04-02 华中农业大学 A method of prokaryotic gene group editor is carried out using endogenous CRISPR-Cas system
EP4089175A1 (en) * 2015-10-13 2022-11-16 Duke University Genome engineering with type i crispr systems in eukaryotic cells
EP3365356B1 (en) 2015-10-23 2023-06-28 President and Fellows of Harvard College Nucleobase editors and uses thereof
DK3350327T3 (en) 2015-10-23 2019-01-21 Caribou Biosciences Inc CONSTRUCTED CRISPR CLASS-2-NUCLEIC ACID TARGETING-NUCLEIC ACID
EP3371329A4 (en) 2015-11-03 2019-06-19 President and Fellows of Harvard College Method and apparatus for volumetric imaging of a three-dimensional nucleic acid containing matrix
EP3382019B1 (en) 2015-11-27 2022-05-04 National University Corporation Kobe University Method for converting monocot plant genome sequence in which nucleic acid base in targeted dna sequence is specifically converted, and molecular complex used therein
WO2017112620A1 (en) 2015-12-22 2017-06-29 North Carolina State University Methods and compositions for delivery of crispr based antimicrobials
RU2749050C2 (en) 2016-01-27 2021-06-03 Онкорус, Инк. Oncolytic viral vectors and use thereof
WO2017165862A1 (en) 2016-03-25 2017-09-28 Editas Medicine, Inc. Systems and methods for treating alpha 1-antitrypsin (a1at) deficiency
CN109415761B (en) 2016-04-25 2022-09-20 哈佛学院董事及会员团体 Hybrid chain reaction method for in situ molecular detection
US11293033B2 (en) 2016-05-18 2022-04-05 Amyris, Inc. Compositions and methods for genomic integration of nucleic acids into exogenous landing pads
AU2017274145B2 (en) 2016-06-02 2020-07-23 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
US20190323038A1 (en) * 2016-06-17 2019-10-24 Montana State Univesity Bidirectional targeting for genome editing
US11566263B2 (en) 2016-08-02 2023-01-31 Editas Medicine, Inc. Compositions and methods for treating CEP290 associated disease
GB2568182A (en) 2016-08-03 2019-05-08 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
AU2017308889B2 (en) 2016-08-09 2023-11-09 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
US20190225974A1 (en) 2016-09-23 2019-07-25 BASF Agricultural Solutions Seed US LLC Targeted genome optimization in plants
KR102622411B1 (en) 2016-10-14 2024-01-10 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 AAV delivery of nucleobase editor
AU2017341926B2 (en) 2016-10-14 2022-06-30 The General Hospital Corporation Epigenetically regulated site-specific nucleases
US9816093B1 (en) 2016-12-06 2017-11-14 Caribou Biosciences, Inc. Engineered nucleic acid-targeting nucleic acids
WO2018119359A1 (en) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Editing of ccr5 receptor gene to protect against hiv infection
JP7348064B2 (en) 2017-01-09 2023-09-20 アポセンス リミテッド Compounds and methods for transmembrane delivery of molecules
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
WO2018165629A1 (en) 2017-03-10 2018-09-13 President And Fellows Of Harvard College Cytosine to guanine base editor
EP3596217A1 (en) 2017-03-14 2020-01-22 Editas Medicine, Inc. Systems and methods for the treatment of hemoglobinopathies
WO2018170436A1 (en) 2017-03-16 2018-09-20 Jacobs Farm Del Cabo Basil with high tolerance to downy mildew
EP3601562A1 (en) 2017-03-23 2020-02-05 President and Fellows of Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins
US10876101B2 (en) 2017-03-28 2020-12-29 Locanabio, Inc. CRISPR-associated (Cas) protein
US11591589B2 (en) 2017-04-21 2023-02-28 The General Hospital Corporation Variants of Cpf1 (Cas12a) with altered PAM specificity
EP3622070A2 (en) 2017-05-10 2020-03-18 Editas Medicine, Inc. Crispr/rna-guided nuclease systems and methods
WO2018209320A1 (en) 2017-05-12 2018-11-15 President And Fellows Of Harvard College Aptazyme-embedded guide rnas for use with crispr-cas9 in genome editing and transcriptional activation
CN110997728A (en) 2017-05-25 2020-04-10 通用医疗公司 Bipartite Base Editor (BBE) structure and II-type-CAS 9 zinc finger editing
BR112019025717A2 (en) 2017-06-08 2020-09-01 Osaka University method to produce eukaryotic cell from edited dna and kit used in the same
CA3067382A1 (en) 2017-06-15 2018-12-20 The Regents Of The University Of California Targeted non-viral dna insertions
EP3650545A4 (en) 2017-06-20 2021-03-31 Jiangsu Hengrui Medicine Co., Ltd. Method for knocking out target gene in t cell in vitro and crrna used in the method
EP3658165A4 (en) 2017-07-26 2021-09-01 Oncorus, Inc. Oncolytic viral vectors and uses thereof
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)
KR102626503B1 (en) 2017-08-21 2024-01-17 토쿠시마 대학 Target sequence-specific modification technology using nucleotide target recognition
WO2019040650A1 (en) 2017-08-23 2019-02-28 The General Hospital Corporation Engineered crispr-cas9 nucleases with altered pam specificity
EP3676376A2 (en) 2017-08-30 2020-07-08 President and Fellows of Harvard College High efficiency base editors comprising gam
US11252928B2 (en) 2017-09-21 2022-02-22 The Condard-Pyle Company Miniature rose plant named ‘Meibenbino’
US11917978B2 (en) 2017-09-21 2024-03-05 The Conard Pyle Company Miniature rose plant named ‘meibenbino’
WO2019075197A1 (en) 2017-10-11 2019-04-18 The General Hospital Corporation Methods for detecting site-specific and spurious genomic deamination induced by base editing technologies
KR20200121782A (en) 2017-10-16 2020-10-26 더 브로드 인스티튜트, 인코퍼레이티드 Uses of adenosine base editor
SG11202003798TA (en) 2017-10-27 2020-05-28 Univ California Targeted replacement of endogenous t cell receptors
EP3737690A1 (en) 2018-01-12 2020-11-18 Basf Se Gene underlying the number of spikelets per spike qtl in wheat on chromosome 7a
US20210093679A1 (en) 2018-02-05 2021-04-01 Novome Biotechnologies, Inc. Engineered gut microbes and uses thereof
JP7422128B2 (en) * 2018-04-03 2024-01-25 ジーフラス ライフ サイエンシズ,インク. Sequence-specific in vivo cell targeting
JP7460539B2 (en) 2018-04-17 2024-04-02 ザ ジェネラル ホスピタル コーポレイション IN VITRO sensitive assays for substrate selectivity and sites of binding, modification, and cleavage of nucleic acids
GB2589246A (en) 2018-05-16 2021-05-26 Synthego Corp Methods and systems for guide RNA design and use
FR3082208A1 (en) * 2018-06-11 2019-12-13 Fondation Mediterranee Infection METHOD OF MODIFYING A TARGET NUCLEIC ACID SEQUENCE OF A HOST CELL
US10227576B1 (en) 2018-06-13 2019-03-12 Caribou Biosciences, Inc. Engineered cascade components and cascade complexes
US20210102183A1 (en) * 2018-06-13 2021-04-08 Caribou Biosciences, Inc. Engineered cascade components and cascade complexes
EP3601574A4 (en) * 2018-06-13 2020-03-18 Caribou Biosciences, Inc. Engineered cascade components and cascade complexes
WO2019246555A1 (en) * 2018-06-21 2019-12-26 Cornell University Type i crispr system as a tool for genome editing
CA3108922A1 (en) 2018-08-29 2020-03-05 Amyris, Inc. Cells and methods for selection based assay
GB201815820D0 (en) 2018-09-28 2018-11-14 Univ Wageningen Off-target activity inhibitors for guided endonucleases
WO2020092725A1 (en) * 2018-11-01 2020-05-07 Montana State University Gene modulation with crispr system type i
CA3117730A1 (en) * 2018-11-09 2020-05-14 Inari Agriculture, Inc. Rna-guided nucleases and dna binding proteins
US10934536B2 (en) 2018-12-14 2021-03-02 Pioneer Hi-Bred International, Inc. CRISPR-CAS systems for genome editing
US11946040B2 (en) 2019-02-04 2024-04-02 The General Hospital Corporation Adenine DNA base editor variants with reduced off-target RNA editing
WO2020176389A1 (en) 2019-02-25 2020-09-03 Caribou Biosciences, Inc. Plasmids for gene editing
BR112021018606A2 (en) 2019-03-19 2021-11-23 Harvard College Methods and compositions for editing nucleotide sequences
AU2020254078A1 (en) 2019-04-05 2021-12-02 Osaka University Method for producing knock-in cell
CN110438142A (en) * 2019-05-13 2019-11-12 安徽大学 A kind of transcriptional control method based on SviCas5-6-7 in I-B-Svi type CRISPR-Cas system
US20230022635A1 (en) * 2019-12-31 2023-01-26 Inari Agriculture Technology, Inc. Delivery of biological molecules to plant cells
JP6940086B1 (en) * 2020-01-24 2021-09-22 C4U株式会社 How to detect specific DNA in a sample
DE112021002672T5 (en) 2020-05-08 2023-04-13 President And Fellows Of Harvard College METHODS AND COMPOSITIONS FOR EDIT BOTH STRANDS SIMULTANEOUSLY OF A DOUBLE STRANDED NUCLEOTIDE TARGET SEQUENCE
CN111979240B (en) * 2020-06-11 2022-04-15 中山大学 Gene expression regulation method and system based on Type I-F CRISPR/Cas
CN111849978B (en) * 2020-06-11 2022-04-15 中山大学 Chromatin imaging method and chromatin imaging system based on Type I-F CRISPR/Cas
US10894812B1 (en) 2020-09-30 2021-01-19 Alpine Roads, Inc. Recombinant milk proteins
US11944063B2 (en) 2020-09-30 2024-04-02 Spring Meadow Nursery, Inc. Hydrangea ‘SMNHPH’
US10947552B1 (en) 2020-09-30 2021-03-16 Alpine Roads, Inc. Recombinant fusion proteins for producing milk proteins in plants
AU2021353004A1 (en) 2020-09-30 2023-04-13 Nobell Foods, Inc. Recombinant milk proteins and food compositions comprising the same
US11155884B1 (en) 2020-10-16 2021-10-26 Klemm & Sohn Gmbh & Co. Kg Double-flowering dwarf Calibrachoa
EP3984355B1 (en) 2020-10-16 2024-05-15 Klemm & Sohn GmbH & Co. KG Double-flowering dwarf calibrachoa
WO2022093977A1 (en) 2020-10-30 2022-05-05 Fortiphyte, Inc. Pathogen resistance in plants
EP4244347A1 (en) 2020-11-10 2023-09-20 Industrial Microbes, Inc. Microorganisms capable of producing poly(hiba) from feedstock
AU2022284804A1 (en) 2021-06-01 2023-12-07 Arbor Biotechnologies, Inc. Gene editing systems comprising a crispr nuclease and uses thereof
WO2023039491A2 (en) * 2021-09-09 2023-03-16 Proof Diagnostics, Inc. Coronavirus rapid diagnostics
KR20240055811A (en) 2021-09-10 2024-04-29 애질런트 테크놀로지스, 인크. Guide RNA with chemical modifications for prime editing
US20230279442A1 (en) 2021-12-15 2023-09-07 Versitech Limited Engineered cas9-nucleases and method of use thereof
CN117384867A (en) * 2022-09-16 2024-01-12 北京普译生物科技有限公司 Modified Cas3 translocation enzyme and application thereof
CN115851664B (en) * 2022-09-19 2023-08-25 中国药科大学 I-B CRISPR-Cascade-Cas3 gene editing system and application

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140295556A1 (en) * 2013-03-15 2014-10-02 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to Increase Specificity for RNA-Guided Genome Editing

Family Cites Families (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
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
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
FR2688514A1 (en) 1992-03-16 1993-09-17 Centre Nat Rech Scient Defective recombinant adenoviruses expressing cytokines and antitumour drugs containing them
US5625048A (en) 1994-11-10 1997-04-29 The Regents Of The University Of California Modified green fluorescent proteins
US5968738A (en) 1995-12-06 1999-10-19 The Board Of Trustees Of The Leland Stanford Junior University Two-reporter FACS analysis of mammalian cells using green fluorescent proteins
US20020182673A1 (en) 1998-05-15 2002-12-05 Genentech, Inc. IL-17 homologous polypedies and therapeutic uses thereof
US6306610B1 (en) 1998-09-18 2001-10-23 Massachusetts Institute Of Technology Biological applications of quantum dots
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
US6635422B2 (en) 1999-12-28 2003-10-21 Ribonomics, Inc. Methods for isolating and characterizing endogenous mRNA-protein (mRNP) complexes
US20020119570A1 (en) 2000-09-25 2002-08-29 Kyonggeun Yoon Targeted gene correction by single-stranded oligodeoxynucleotides
MXPA03003690A (en) 2000-10-27 2004-05-05 Chiron Spa Nucleic acids and proteins from streptococcus groups a b.
US20060253913A1 (en) 2001-12-21 2006-11-09 Yue-Jin Huang Production of hSA-linked butyrylcholinesterases in transgenic mammals
CA2474486C (en) 2002-01-23 2013-05-14 The University Of Utah Research Foundation Targeted chromosomal mutagenesis using zinc finger nucleases
AU2003218382B2 (en) 2002-03-21 2007-12-13 Sangamo Therapeutics, Inc. Methods and compositions for using zinc finger endonucleases to enhance homologous recombination
US7539579B2 (en) 2002-04-09 2009-05-26 Beattie Kenneth L Oligonucleotide probes for genosensor chips
AU2003298574B2 (en) 2002-09-05 2008-04-24 California Institute Of Technology Use of chimeric nucleases to stimulate gene targeting
US20120196370A1 (en) 2010-12-03 2012-08-02 Fyodor Urnov Methods and compositions for targeted genomic deletion
US7972854B2 (en) 2004-02-05 2011-07-05 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
CN101426931A (en) * 2004-04-07 2009-05-06 安克塞斯生物公司 Nucleic acid detection system
US7919277B2 (en) 2004-04-28 2011-04-05 Danisco A/S Detection and typing of bacterial strains
US7892224B2 (en) 2005-06-01 2011-02-22 Brainlab Ag Inverse catheter planning
JP2009502170A (en) 2005-07-26 2009-01-29 サンガモ バイオサイエンシズ インコーポレイテッド Targeted integration and expression of foreign nucleic acid sequences
DK3284833T3 (en) 2005-08-26 2022-02-07 Dupont Nutrition Biosci Aps USE OF CRISPR-ASSOCIATED GENES (CAS)
WO2007128338A1 (en) 2006-05-10 2007-11-15 Deinove Process for chromosomal engineering using a novel dna repair system
ES2373586T3 (en) 2006-05-19 2012-02-06 Danisco A/S MARKED MICROORGANISMS AND METHODS TO MARK.
EP2447279B1 (en) 2006-05-25 2014-04-09 Sangamo BioSciences, Inc. Methods and compositions for gene inactivation
CN101505607B (en) 2006-06-16 2013-06-26 杜邦营养生物科学有限公司 Bacterium
EP2054514A4 (en) 2006-08-04 2009-11-04 Univ Georgia State Res Found Enzyme sensors, methods for preparing and using such sensors, and methods of detecting protease activity
BRPI0808704B1 (en) 2007-03-02 2022-01-18 Dupont Nutrition Biosciences Aps METHOD TO GENERATE AN INITIAL CULTURE COMPRISING AT LEAST TWO BACTERIOPHAGE-RESISTANT VARIANTS, INITIATOR CULTURE AND FERMENTATION METHOD
ES2425066T3 (en) * 2007-04-05 2013-10-11 Speedx Pty Ltd Enzymes and nucleic acid complexes and methods for use
FR2925918A1 (en) 2007-12-28 2009-07-03 Pasteur Institut Typing or subtyping Salmonella bacteria comprises determining the variable sequence composition of a nucleic acid fragment amplified from the CRISPR1 and/or CRISPR2 locus
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
DK2334794T3 (en) 2008-09-15 2017-02-20 Children's Medical Center Corp MODULATION OF BCL11A FOR TREATMENT OF HEMOGLOBINOPATHIES
US20100076057A1 (en) * 2008-09-23 2010-03-25 Northwestern University TARGET DNA INTERFERENCE WITH crRNA
US9404098B2 (en) 2008-11-06 2016-08-02 University Of Georgia Research Foundation, Inc. Method for cleaving a target RNA using a Cas6 polypeptide
US10662227B2 (en) 2008-11-07 2020-05-26 Dupont Nutrition Biosciences Aps Bifidobacteria CRISPR sequences
US8771766B2 (en) 2008-12-12 2014-07-08 Dupont Nutrition Biosciences Aps Genetic cluster of strains of Streptococcus thermophilus having unique rheological properties for dairy fermentation
WO2010075424A2 (en) 2008-12-22 2010-07-01 The Regents Of University Of California Compositions and methods for downregulating prokaryotic genes
WO2010117464A1 (en) 2009-04-09 2010-10-14 Sangamo Biosciences, Inc. Targeted integration into stem cells
PL2424571T3 (en) 2009-04-30 2020-10-19 Ospedale San Raffaele S.R.L Gene vector
JP2012529287A (en) 2009-06-11 2012-11-22 トゥールゲン インコーポレイション Rearrange target genomes using site-specific nucleases
US20120192298A1 (en) 2009-07-24 2012-07-26 Sigma Aldrich Co. Llc Method for genome editing
WO2011011767A1 (en) 2009-07-24 2011-01-27 Sigma-Aldrich Co. Method for genome editing
JP5866283B2 (en) 2009-07-28 2016-02-17 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 treating trinucleotide repeat disease
KR102262704B1 (en) 2009-08-11 2021-06-09 상가모 테라퓨틱스, 인코포레이티드 Organisms homozygous for targeted modification
EP2292731B1 (en) 2009-08-13 2018-04-04 DuPont Nutrition Biosciences ApS Method for preparing complex cultures
EP3118320A1 (en) 2009-09-25 2017-01-18 BASF Plant Science Company GmbH Plants having enhanced yield-related traits and a method for making the same
US20110105364A1 (en) 2009-11-02 2011-05-05 Nugen Technologies, Inc. Compositions and methods for targeted nucleic acid sequence selection and amplification
BR112012012444A2 (en) 2009-11-27 2015-09-22 Basf Plant Science Co Gmbh "chimeric endonuclease, isolated polynucleotide, expression cassette, vector, non-human organism, method for providing a chimeric endonuclease, method for homologous polynucleotide recombination, method for targeted polynucleotide mutation, and method for homologous recombination or targeted mutation"
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
PT2816112T (en) 2009-12-10 2018-11-20 Univ Iowa State Res Found Inc Tal effector-mediated dna modification
US20110203012A1 (en) 2010-01-21 2011-08-18 Dotson Stanton B Methods and compositions for use of directed recombination in plant breeding
WO2011097036A1 (en) * 2010-02-08 2011-08-11 Sangamo Biosciences, Inc. Engineered cleavage half-domains
EP2534163B1 (en) 2010-02-09 2015-11-04 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
WO2011130346A1 (en) 2010-04-13 2011-10-20 Sigma-Aldrich Co. Methods for generating endogenously tagged protein
BR112012028805A2 (en) 2010-05-10 2019-09-24 The Regents Of The Univ Of California E Nereus Pharmaceuticals Inc endoribonuclease compositions and methods of use thereof.
CN103025344B (en) 2010-05-17 2016-06-29 桑格摩生物科学股份有限公司 Novel DNA-associated proteins and application 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
JP2013537410A (en) 2010-07-23 2013-10-03 シグマ−アルドリッチ・カンパニー・リミテッド・ライアビリティ・カンパニー Genome editing using targeted endonucleases and single-stranded nucleic acids
WO2012018697A1 (en) 2010-08-02 2012-02-09 Integrated Dna Technologies, Inc. Methods for predicting stability and melting temperatures of nucleic acid duplexes
DK2630156T3 (en) 2010-10-20 2018-12-17 Dupont Nutrition Biosci Aps CRISPR-CAS SEQUENCES OF LACTOCOCCUS
KR20120096395A (en) 2011-02-22 2012-08-30 주식회사 툴젠 Methods for enriching cells containing nuclease-induced gene disruptions
CN103492578B (en) 2011-04-27 2016-10-19 阿迈瑞斯公司 Method for genomic modification
US20140113376A1 (en) 2011-06-01 2014-04-24 Rotem Sorek Compositions and methods for downregulating prokaryotic genes
US8927218B2 (en) 2011-06-27 2015-01-06 Flir Systems, Inc. Methods and compositions for segregating target nucleic acid from mixed nucleic acid samples
SG10201811736QA (en) 2011-09-21 2019-02-27 Sangamo Therapeutics Inc Methods and compositions for regulation of transgene expression
US8450107B1 (en) 2011-11-30 2013-05-28 The Broad Institute Inc. Nucleotide-specific recognition sequences for designer TAL effectors
IN2014MN00974A (en) 2011-12-16 2015-04-24 Targetgene Biotechnologies Ltd
GB201122458D0 (en) 2011-12-30 2012-02-08 Univ Wageningen Modified cascade ribonucleoproteins and uses thereof
RU2650811C2 (en) 2012-02-24 2018-04-17 Фред Хатчинсон Кэнсер Рисерч Сентер Compositions and methods for treatment of hemoglobinopathies
EP2820159B1 (en) 2012-02-29 2019-10-23 Sangamo Therapeutics, Inc. Methods and compositions for treating huntington's disease
US9637739B2 (en) 2012-03-20 2017-05-02 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
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
BR112014026203A2 (en) 2012-04-23 2017-07-18 Bayer Cropscience Nv plant-directed genome engineering
KR102091298B1 (en) 2012-05-02 2020-03-19 다우 아그로사이언시즈 엘엘씨 Targeted modification of malate dehydrogenase
JP6559063B2 (en) 2012-05-07 2019-08-14 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 nuclease-mediated targeted integration of transgenes
WO2013169398A2 (en) 2012-05-09 2013-11-14 Georgia Tech Research Corporation Systems and methods for improving nuclease specificity and activity
ES2960803T3 (en) * 2012-05-25 2024-03-06 Univ California Methods and compositions for RNA-directed modification of target DNA and for modulation of RNA-directed transcription
US20140056868A1 (en) 2012-05-30 2014-02-27 University of Washington Center for Commercialization 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
CN104540382A (en) 2012-06-12 2015-04-22 弗·哈夫曼-拉罗切有限公司 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
EP2861737B1 (en) 2012-06-19 2019-04-17 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
EP3444342B1 (en) 2012-07-11 2020-05-27 Sangamo Therapeutics, Inc. Methods and compositions for the treatment of lysosomal storage diseases
CN116622704A (en) 2012-07-25 2023-08-22 布罗德研究所有限公司 Inducible DNA binding proteins and genomic disruption tools and uses thereof
KR102474010B1 (en) 2012-08-29 2022-12-02 상가모 테라퓨틱스, 인코포레이티드 Methods and compositions for treatment of a genetic condition
US9914930B2 (en) 2012-09-07 2018-03-13 Dow Agrosciences Llc FAD3 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
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
EP2906602B1 (en) 2012-10-12 2019-01-16 The General Hospital Corporation Transcription activator-like effector (tale) - lysine-specific demethylase 1 (lsd1) fusion proteins
ES2926021T3 (en) 2012-10-23 2022-10-21 Toolgen Inc Composition for cleaving a target DNA comprising a target DNA-specific guide RNA and Cas protein-encoding nucleic acid or Cas protein, and use thereof
JP2015533284A (en) 2012-10-30 2015-11-24 リコンビネティクス・インコーポレイテッドRecombinetics,Inc. Control of sexual maturity in animals
BR112015009812A2 (en) 2012-10-31 2017-08-22 Cellectis METHOD FOR SPECIFIC GENETIC INSERTION INTO A PLANT GENOME, TRANSFORMED PLANT CELL AND ITS USE, HERBICIDIDE RESISTANT PLANT, KIT, VECTOR, AND HOST CELL
US20140127752A1 (en) 2012-11-07 2014-05-08 Zhaohui Zhou Method, composition, and reagent kit for targeted genomic enrichment
EP3617309A3 (en) * 2012-12-06 2020-05-06 Sigma Aldrich Co. LLC Crispr-based genome modification and regulation
IL239344B1 (en) 2012-12-12 2024-02-01 Broad Inst Inc 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
ES2576128T3 (en) 2012-12-12 2016-07-05 The Broad Institute, Inc. Modification by genetic technology and optimization of systems, methods and compositions for the manipulation of sequences with functional domains
WO2014093709A1 (en) 2012-12-12 2014-06-19 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
US20140310830A1 (en) 2012-12-12 2014-10-16 Feng Zhang CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
CN113355357A (en) 2012-12-12 2021-09-07 布罗德研究所有限公司 Engineering and optimization of improved systems, methods and enzyme compositions for sequence manipulation
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
SG10201912328UA (en) 2012-12-12 2020-02-27 Broad Inst Inc Delivery, Engineering and Optimization of Systems, Methods and Compositions for Sequence Manipulation and Therapeutic Applications
EP3144390B1 (en) 2012-12-12 2020-03-18 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
IL308158A (en) 2012-12-17 2023-12-01 Harvard College Rna-guided human genome engineering
DK3491915T3 (en) 2012-12-27 2023-08-28 Keygene Nv Method of inducing a targeted translocation in a plant
CN104995302B (en) 2013-01-16 2021-08-31 爱默蕾大学 CAS 9-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
CN103224947B (en) 2013-04-28 2015-06-10 陕西师范大学 Gene targeting system
CA2914519A1 (en) * 2013-06-05 2014-12-11 Duke University Rna-guided gene editing and gene regulation
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
US10227576B1 (en) * 2018-06-13 2019-03-12 Caribou Biosciences, Inc. Engineered cascade components and cascade complexes

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140295556A1 (en) * 2013-03-15 2014-10-02 The General Hospital Corporation Using RNA-guided FokI Nucleases (RFNs) to Increase Specificity for RNA-Guided Genome Editing

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
Brouns et al. (2008) Small CRISPR RNAs Guide Antiviral Defense in Prokaryotes. Science, 321:960-964, and Supporting Online Material *
Caccavo et al. (1994) Geobacter sulfurreducens sp. nov., a Hydrogen- and Acetate-Oxidizing Dissimilatory Metal-Reducing Microorganism. Applied and Environmental Microbiology, 60(10):3752-3759 *
Choudhury et al. (2012) Engineering RNA endonucleases with customized sequence specificities. Nature Communications, 3(1147), pages 1-8 *
Kim et al. (1997) Site-specific cleavage of DNA-RNA hybrids by zinc finger/FokI cleavage domain fusions. Gene, 203:43-49 *
Klein et al. (2014) Design and characterization of structured protein linkers with differing flexibilities. Protein Engineering, Design & Selection, 27(10):325-330 *
Methe et al. (2003) Genome of Geobacter sulfurreducens: Metal Reduction in Subsurface Environments. Science, 302:1967-1969 *
NC_002939.4 (Geobacter sulfurreducens PCA chromosome, complete genome, nucleotide positions 72630-74309, priority to October 14, 2010, 2 pages) *
NC_002939.5 (Geobacter sulfurreducens PCA chromosome, complete genome, nucleotide positions 72629-74308, priority to August 3, 2016, 2 pages) *
NZ_GG657742.1 (Streptomyces sp. SPB78 genomic scaffold supercont1.1, whole genome shotgun sequence, NCBI Reference Sequence, nucleotide positions 3502816-3507630, priority to October 26, 2010, 3 pages) *
Pingoud et al. (2014) Type II restriction endonucleases–a historical perspective and more. Nucleic Acids Research, 42(12):7489-7527 *
Rittie et al. (2008) Enzymes used in molecular biology: a useful guide. Journal of Cell Communication and Signaling, 2(1):25-45 *
Sorek et al. (2012) CRISPR-Mediated Adaptive Immune Systems in Bacteria and Archaea. Annual Reviews Biochemistry, 82:237-266 *
van der Oost et al. (2009) CRISPR-based adaptive and heritable immunity in prokaryotes. Trends in Biochemical Sciences, 34(8):401-407 *
Zhao et al. (2008) Increasing the homogeneity, stability and activity of human serum albumin and interferon-a2b fusion protein by linker engineering. Protein Expression and Purification, 61:73-77 *

Cited By (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11499169B2 (en) 2013-10-30 2022-11-15 North Carolina State University Compositions and methods related to a type-II CRISPR-Cas system in Lactobacillus buchneri
US10640788B2 (en) 2013-11-07 2020-05-05 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
US10190137B2 (en) 2013-11-07 2019-01-29 Editas Medicine, Inc. CRISPR-related methods and compositions with governing gRNAS
US10787654B2 (en) 2014-01-24 2020-09-29 North Carolina State University Methods and compositions for sequence guiding Cas9 targeting
US11439712B2 (en) 2014-04-08 2022-09-13 North Carolina State University Methods and compositions for RNA-directed repression of transcription using CRISPR-associated genes
US11753651B2 (en) 2014-08-28 2023-09-12 North Carolina State University Cas9 proteins and guiding features for DNA targeting and genome editing
US11680268B2 (en) 2014-11-07 2023-06-20 Editas Medicine, Inc. Methods for improving CRISPR/Cas-mediated genome-editing
US10993419B2 (en) 2014-12-10 2021-05-04 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
US10278372B2 (en) 2014-12-10 2019-05-07 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
US11390884B2 (en) 2015-05-11 2022-07-19 Editas Medicine, Inc. Optimized CRISPR/cas9 systems and methods for gene editing in stem cells
US11261451B2 (en) 2015-05-29 2022-03-01 North Carolina State University Methods for screening bacteria, archaea, algae, and yeast using CRISPR nucleic acids
US11911415B2 (en) 2015-06-09 2024-02-27 Editas Medicine, Inc. CRISPR/Cas-related methods and compositions for improving transplantation
US11155823B2 (en) 2015-06-15 2021-10-26 North Carolina State University Methods and compositions for efficient delivery of nucleic acids and RNA-based antimicrobials
US11642374B2 (en) 2015-07-31 2023-05-09 Intima Bioscience, Inc. Intracellular genomic transplant and methods of therapy
US11925664B2 (en) 2015-07-31 2024-03-12 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
US11583556B2 (en) 2015-07-31 2023-02-21 Regents Of The University Of Minnesota Modified cells and methods of therapy
US11266692B2 (en) 2015-07-31 2022-03-08 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
US11147837B2 (en) 2015-07-31 2021-10-19 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
US11667911B2 (en) 2015-09-24 2023-06-06 Editas Medicine, Inc. Use of exonucleases to improve CRISPR/CAS-mediated genome editing
US11286480B2 (en) 2015-09-28 2022-03-29 North Carolina State University Methods and compositions for sequence specific antimicrobials
US11111287B2 (en) 2016-01-11 2021-09-07 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of immunotherapy
US10336807B2 (en) 2016-01-11 2019-07-02 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of immunotherapy
US11773411B2 (en) 2016-01-11 2023-10-03 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
US9856497B2 (en) 2016-01-11 2018-01-02 The Board Of Trustee Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
US10457961B2 (en) 2016-01-11 2019-10-29 The Board Of Trustees Of The Leland Stanford Junior University Chimeric proteins and methods of regulating gene expression
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
US10912797B2 (en) 2016-10-18 2021-02-09 Intima Bioscience, Inc. Tumor infiltrating lymphocytes and methods of therapy
US11154574B2 (en) 2016-10-18 2021-10-26 Regents Of The University Of Minnesota Tumor infiltrating lymphocytes and methods of therapy
WO2018148647A3 (en) * 2017-02-10 2018-09-20 Lajoie Marc Joseph Genome editing reagents and their use
US11866699B2 (en) 2017-02-10 2024-01-09 University Of Washington Genome editing reagents and their use
US11499151B2 (en) 2017-04-28 2022-11-15 Editas Medicine, Inc. Methods and systems for analyzing guide RNA molecules
US11098297B2 (en) 2017-06-09 2021-08-24 Editas Medicine, Inc. Engineered Cas9 nucleases
US10428319B2 (en) 2017-06-09 2019-10-01 Editas Medicine, Inc. Engineered Cas9 nucleases
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
US11491208B2 (en) 2018-03-27 2022-11-08 Gflas Life Sciences, Inc. Sequence-specific in vivo cell targeting
WO2019186275A1 (en) * 2018-03-27 2019-10-03 G+Flas Life Sciences Sequence-specific in vivo cell targeting
KR102302679B1 (en) * 2018-03-27 2021-09-16 (주)지플러스 생명과학 Pharmaceutical composition for treating cancers comprising guide rna and endonuclease
KR102023577B1 (en) * 2018-03-27 2019-09-24 (주)지플러스 생명과학 Pharmaceutical composition for treating cancers comprising guide rna and endonuclease
KR20190113687A (en) * 2018-03-27 2019-10-08 (주)지플러스 생명과학 Pharmaceutical composition for treating cancers comprising guide rna and endonuclease
TWI723363B (en) * 2018-03-27 2021-04-01 南韓商G+Flas生命科學有限公司 Pharmaceutical composition for treating cancers comprising guide rna and endonuclease
WO2019190198A1 (en) * 2018-03-27 2019-10-03 (주)지플러스 생명과학 Pharmaceutical composition for treating cancer, containing guide rna and endonuclease as active ingredients
CN111989113A (en) * 2018-03-27 2020-11-24 G+Flas生命科学有限公司 Pharmaceutical composition for treating cancer comprising guide RNA and endonuclease as active ingredients
CN112533627A (en) * 2018-03-27 2021-03-19 G+Flas 生命科学公司 Sequence specific in vivo cell targeting
US11865164B2 (en) 2018-03-27 2024-01-09 G+Flas Life Sciences Pharmaceutical composition for treating cancer, containing guide RNA and endonuclease as active ingredients
US11680259B2 (en) 2018-10-01 2023-06-20 North Carolina State University Recombinant type I CRISPR-CAS system
WO2020072253A1 (en) * 2018-10-01 2020-04-09 North Carolina State University Recombinant type i crispr-cas system and uses thereof for screening for variant cells
US10711267B2 (en) 2018-10-01 2020-07-14 North Carolina State University Recombinant type I CRISPR-Cas system
WO2021092210A1 (en) * 2019-11-06 2021-05-14 Locus Biosciences, Inc. Phage compositions comprising crispr-cas systems and methods of use thereof

Also Published As

Publication number Publication date
MX364830B (en) 2019-05-08
EP3091072A1 (en) 2016-11-09
IL233399A0 (en) 2014-08-31
US20210171926A1 (en) 2021-06-10
CA2862018C (en) 2019-12-03
KR101889589B1 (en) 2018-08-17
US20160186214A1 (en) 2016-06-30
DK3091072T3 (en) 2018-08-06
RS57604B1 (en) 2018-11-30
KR20140115335A (en) 2014-09-30
US20180119121A1 (en) 2018-05-03
CA2862018A1 (en) 2013-07-04
HUE039617T2 (en) 2019-01-28
US20200040318A1 (en) 2020-02-06
RU2014127702A (en) 2016-02-20
SG11201403713QA (en) 2014-07-30
US10435678B2 (en) 2019-10-08
HRP20181150T1 (en) 2019-10-04
EP3091072B1 (en) 2018-07-04
ES2689256T3 (en) 2018-11-12
GB2512246B (en) 2016-07-20
US9885026B2 (en) 2018-02-06
JP6408914B2 (en) 2018-10-17
GB201605069D0 (en) 2016-05-11
GB2512246A (en) 2014-09-24
GB2534074A (en) 2016-07-13
BR112014016228B1 (en) 2022-05-03
US10954498B2 (en) 2021-03-23
LT3091072T (en) 2018-08-27
AU2012360975B2 (en) 2017-10-05
BR112014016228A2 (en) 2018-09-25
PT3091072T (en) 2018-07-25
JP2015503535A (en) 2015-02-02
WO2013098244A1 (en) 2013-07-04
GB201411878D0 (en) 2014-08-20
CN105732816A (en) 2016-07-06
DE212012000234U1 (en) 2014-08-07
US11939604B2 (en) 2024-03-26
US20140294773A1 (en) 2014-10-02
CY1120538T1 (en) 2019-07-10
CN104321429A (en) 2015-01-28
SI3091072T1 (en) 2018-09-28
AU2012360975A1 (en) 2014-07-24
US20150024499A1 (en) 2015-01-22
MX2014007910A (en) 2015-02-12
US20200325462A1 (en) 2020-10-15
IN2014DN05937A (en) 2015-06-12
GB201122458D0 (en) 2012-02-08
US10711257B2 (en) 2020-07-14
PL3091072T3 (en) 2019-01-31
EP2798060A1 (en) 2014-11-05
CN105732816B (en) 2020-12-25

Similar Documents

Publication Publication Date Title
US10954498B2 (en) Modified cascade ribonucleoproteins and uses thereof
US11427818B2 (en) S. pyogenes CAS9 mutant genes and polypeptides encoded by same
CN107922931B (en) Thermostable Cas9 nuclease
US11913014B2 (en) S. pyogenes Cas9 mutant genes and polypeptides encoded by same
JP2023022254A (en) Crispr/cpf1 systems and methods
KR102647766B1 (en) Class II, type V CRISPR systems
US20240174998A1 (en) Modified cascade ribonucleoproteins and uses thereof
CN117264987A (en) Method for chemically controllable gene editing by engineering AcrIIA25.1 protein
KR20230054457A (en) Systems and methods for translocating cargo nucleotide sequences
WO2022013133A1 (en) Methods for modulating cas-effector activity

Legal Events

Date Code Title Description
AS Assignment

Owner name: WAGENINGEN UNIVERSITEIT, NETHERLANDS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VAN DER OOST, JOHN;BROUNS, STAN JOHAN JOZEF;SIGNING DATES FROM 20140409 TO 20140410;REEL/FRAME:041010/0272

Owner name: CARIBOU BIOSCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WAGENINGEN UNIVERSITEIT;REEL/FRAME:041010/0277

Effective date: 20140410

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION