RNA-DIRECTED DNA CLEAVAGE BY THE Cas9-crRNA COMPLEX

- VILNIUS UNIVERSITY

Isolation or in vitro assembly of the Cas9-crRNA complex of the Streptococcus thermophilus CRISPR3/Cas system and use for cleavage of DNA bearing a nucleotide sequence complementary to the crRNA and a proto-spacer adjacent motif. Methods for site-specific modification of a target DNA molecule in vitro or in vivo using an RNA-guided DNA endonuclease comprising RNA sequences and at least one of an RuvC active site motif and an HNH active site motif; for conversion of Cas9 polypeptide into a nickase cleaving one strand of double-stranded DNA by inactivating one of the active sites (RuvC or HNH) in the polypeptide by at least one point mutation; for assembly of active polypeptide-polyribonucleotides complex in vivo or in vitro; and for re-programming a Cas9-crRNA complex specificity in vitro and using a cassette containing a single repeat-spacer-repeat unit.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

This applications claims priority to co-pending U.S. application Ser. Nos. 61/613,373 filed Mar. 20, 2012, and 61/625,420 filed Apr. 17, 2012, each of which is expressly incorporated by reference herein in its entirety.

CRISPR/Cas systems provide adaptive immunity against viruses and plasmids in bacteria and archaea. The silencing of invading nucleic acids is executed by ribonucleoprotein (RNP) complexes pre-loaded with small interfering crRNAs that act as guides for foreign nucleic acid targeting and degradation. Here we describe an isolation of the Cas9-crRNA complex and demonstrate that it generates in vitro a double strand break at specific sites in target DNA molecules that are complementary to crRNA sequences and bear a short proto-spacer adjacent motif (PAM), in the direct vicinity of the matching sequence. We show that DNA cleavage is executed by two distinct active sites (RuvC and HNH) within Cas9, to generate site-specific nicks on opposite DNA strands. Sequence specificity of the Cas9-crRNA complex is dictated by the 42 nt crRNA which includes a 20 nt fragment complementary to the proto-spacer sequence in the target DNA. The complex can be assembled in vitro or in vivo. Altogether, our data demonstrate that the Cas9-crRNA complex functions as an RNA-guided endonuclease with sequence-specific target site recognition and cleavage through two distinct strand nicks.

BACKGROUND

Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) together with cas (CRISPR-associated) genes comprise an adaptive immune system that provides acquired resistance against invading foreign nucleic acids in bacteria and archaea (Barrangou et al., 2007. Science 315:1709-12). CRISPR consists of arrays of short conserved repeat sequences interspaced by unique variable DNA sequences of similar size called spacers, which often originate from phage or plasmid DNA (Barrangou et al., 2007. Science 315:1709-12; Bolotin et al., 2005. Microbiology 151:2551-61; Mojica et al., 2005. J Mol Evol 60:174-82). The CRISPR-Cas system functions by acquiring short pieces of foreign DNA (spacers) which are inserted into the CRISPR region and provide immunity against subsequent exposures to phages and plasmids that carry matching sequences (Barrangou et al., 2007. Science 315:1709-12; Brouns et al., 2008. Science 321: 960-4) The CRISPR-Cas immunity is generally carried out through three stages, referred to as i) adaptation/immunization/spacer acquisition, ii) CRISPR expression/crRNA biogenesis, iii) interference/immunity. (Horvath & Barrangou, 2010. Science 327:167-70; Deveau et al., 2010. Annu Rev Microbiol. 64:475-93; Marraffini & Sontheimer, 2010. Nat Rev Genet 11, 181-90; Bhaya et al., Annu Rev Genet 45:273-97; Wiedenheft et al., 2012. Nature 482:331-338). Here, we specifically focus on the interference/immunity step which enables crRNA-mediated silencing of foreign nucleic acids.

The highly diverse CRISPR-Cas systems are categorized into three major types, which are further subdivided into ten subtypes, based on core element content and sequences (Makarova et al., 2011. Nat Rev Microbiol 9:467-77). The structural organization and function of nucleoprotein complexes involved in crRNA-mediated silencing of foreign nucleic acids differ between distinct CRISPR/Cas types (Wiedenheft et al., 2012. Nature 482:331-338). In the Type I-E system, as exemplified by Escherichia coli, crRNAs are incorporated into a multisubunit effector complex called Cascade (CRISPR-associated complex for antiviral defence) (Brouns et al., 2008. Science 321: 960-4), which binds to the target DNA and triggers degradation by the signature Cas3 protein (Sinkunas et al., 2011. EMBO J 30:1335-42; Beloglazova et al., 2011. EMBO J 30:616-27). In Type III CRISPR/Cas systems of Sulfolobus solfataricus and Pyrococcus furiosus, Cas RAMP module (Cmr) and crRNA complex recognize and cleave synthetic RNA in vitro (Hale et al., 2012. Mol Cell 45:292-302; Zhang et al., 2012. Mol Cell, 45:303-13) while the CRISPR/Cas system of Staphylococcus epidermidis targets DNA in vivo (Marraffini & Sontheimer, Science. 322:1843-5).

RNP complexes involved in DNA silencing by Type II CRISPR/Cas systems, more specifically in the CRISPR3/Cas system of Streptococcus thermophilus DGCC7710 (Horvath & Barrangou, 2010. Science 327:167-70), consists of four cas genes cas9, cas1, cas2, and csn2, that are located upstream of 12 repeat-spacer units (FIG. 1A). Cas9 (formerly named cas5 or csn1) is the signature gene for Type II systems (Makarova et al., 2011. Nat Rev Microbiol 9:467-77). In the closely related S. thermophilus CRISPR1/Cas system, disruption of cas9 abolishes crRNA-mediated DNA interference (Barrangou et al., 2007. Science 315:1709-12). We have shown recently that the S. thermophilus CRISPR3/Cas system can be transferred into Escherichia coli, and that this heterologous system provides protection against plasmid transformation and phage infection, de novo (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82). The interference against phage and plasmid DNA provided by S. thermophilus CRISPR3 requires the presence, within the target DNA, of a proto-spacer sequence complementary to the spacer-derived crRNA, and a conserved PAM (Proto-spacer Adjacent Motif) sequence, NGGNG. located immediately downstream the proto-spacer (Deveau et al., 2008. J Bacteriol 190:1390-400; Horvath et al., 2008. J Bacteriol 190:1401-12; Mojica et al., 2009. Microbiology 155:733-40). Single point mutations in the PAM or defined proto-spacer positions allow the phages or plasmids to circumvent CRISPR-mediated immunity (Deveau et al., 2008. J Bacteriol 190:1390-400; Garneau et al., 2010. Nature 468:67-71; Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82). We have established that in the heterologous system, cas9 is the sole cas gene necessary for CRISPR-encoded interference (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82), suggesting that this protein is involved in crRNA processing and/or crRNA-mediated silencing of invasive DNA. Cas9 of S. thermophilus CRISPR3/Cas system is a large multi-domain protein comprised of 1,409 aa residues (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82). It contains two nuclease domains, a RuvC-like nuclease domain near the amino terminus, and a HNH-like nuclease domain in the middle of the protein. Mutational analysis has established that interference provided in vivo by Cas9 requires both the RuvC- and HNH-motifs (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82).

Isolation of the Cas9-crRNA complex of the S. thermophilus CRISPR3/Cas system as well as complex assembly in vitro from separate components and demonstration that it cleaves both synthetic oligodeoxynucleotide and plasmid DNA bearing a nucleotide sequence complementary to the crRNA, in a PAM-dependent manner, is provided. Furthermore, we provide experimental evidence that the PAM is recognized in the context of double-stranded DNA and is critical for in vitro DNA binding and cleavage. Finally, we show that the Cas9 RuvC- and HNH-active sites are responsible for the cleavage of opposite DNA strands. Taken together, our data demonstrate that the Cas9-crRNA complex functions as an RNA-guided endonuclease which uses RNA for the target site recognition and Cas9 for DNA cleavage. The simple modular organization of the Cas9-crRNA complex, where specificity for DNA targets is encoded by a small crRNA and the cleavage machinery consists of a single, multidomain Cas protein, provides a versatile platform for the engineering of universal RNA-guided DNA endonucleases. Indeed, we provide evidence that by altering the RNA sequence within the Cas9-crRNA complex, programmable endonucleases can be designed both for in vitro and in vivo applications, and we provide a proof of concept for this novel application. These findings pave the way for the development of novel molecular tools for RNA-directed DNA surgery.

SUMMARY OF THE INVENTION

A method for the site-specific modification of a target DNA molecule through contacting under suitable conditions, a target polydeoxynucleotide molecule; and an RNA-guided DNA endonuclease comprising at least one RNA sequences and at least one of an RuvC active site motif and an HNH active site motif; to result in the target polydeoxynucleotide molecule modified in a region that is determined by the complimentary binding of the RNA sequence to the target DNA molecule is provided. The method includes incubating under suitable conditions a composition that includes a target double stranded polydeoxynucleotide or single stranded polydeoxynucleotide; wherein a double stranded polydeoxynucleotide contains a short proto-spacer adjacent motif (PAM), which is non-obligatory for a single stranded polydeoxynucleotide; and where PAM comprises a 5′NGGNG-3′ sequence; a polyribonucleotide (crRNA) comprising a 3′ and 5′ regions wherein the 3′ region comprises at least 22 nt of the repeat present in a microbe containing CRISPR locus and 5′-region comprises of at least 20 nt of the spacer sequence immediately downstream of the repeat in the CRISPR locus, which is substantially complementary, optionally complementary, to a portion of the target polynucleotide, a polypeptide wherein the amino acid sequence of polypeptide and amino acid sequence of SEQ ID NO: 1 have at least 80% identity, isolated from S. thermophilus, or genetically modified microorganism, including a genetically modified E. coli, or wherein the polypeptide is produced by a method selected from recombinant DNA technology or chemical synthesis; a polyribonucleotide tracrRNA of nucleotide sequence SEQ ID NO: 5 (or have at least 80% identity) comprising a 5′ and 3′ regions wherein the 5′ region is comprised of at least 22 nucleotides is complementary to the 22 nucleotides 3′ region of crRNA, and 3′ region. Wherein polyribonucleotides are produced by in vitro transcription or chemical synthesis. Wherein, suitable conditions means conditions in vitro or in vivo where reaction might occur.

A method for the conversion of Cas9 polypeptide into a nickase, cleaving only one strand of double-stranded DNA, by inactivating one of the active sites (RuvC or HNH) in the polypeptide by at least on point mutation, exemplified by D31A (SEQ ID NO: 2), N891A (SEQ ID NO: 3) and H868A (SEQ ID NO: 4) point mutations is provided. RuvC motif mutant cleaves only bottom DNA strand in respect to 5′NGGNG-3′ motif, while HNH motif mutant cleaves top strand.

Polypeptide-polyribonucleotides complex might be isolated from a genetically modified microbe (for example Escherichia coli or Streptococcus thermophilus), or assembled in vitro from separate components. In the genetically modified microbe components of the complex might be encoded on the one, two or three separate plasmids containing host promoters of the genetically modified microbe or promoters from a native host genome.

A method for assembly of active polypeptide-polyribonucleotides complex in vitro, comprising incubating the components of the complex under conditions suitable for complex assembly is provided. The complex might be assembled using three or four components. Method for three components assembly comprises incubating the Cas9 polypeptide, 78 nt tracrRNA polyribonucleotide (SEQ ID NO: 5), and 42 nt crRNA polyribonucleotide (5′-NNNNNNNNNNNNNNNNNNNN GUUUUAGAGCUGUGUUGUUUCG-3′) (SEQ ID NO: 15) under conditions suitable for complex assembly. Method for four components assembly comprises incubating the Cas9 polypeptide; 102 nt tracrRNA polyribonucleotide (SEQ ID NO: 6); polyribonucleotide containing sequence 5′-NNNNNNNNNNNNNNNNNNNN GUUUUAGAGCUGUGUUGUUUCG-3′ (SEQ ID NO: 15) and flanking regions and RNase III polypeptide, cleaving double stranded RNA polynucleotide. The examples for polyribonucleotide containing sequence 5′-NNNNNNNNNNNNNNNNNNNN GUUUUAGAGCUGUGUUGUUUCG-3′ (SEQ ID NO: 15) are SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11 and SEQ ID NO: 12). Examples of source for suitable RNaseIII include Escherichia coli or Streptococcus thermophilus.

A method for re-programming of a Cas9-crRNA complex specificity by mixing separate components or using a cassette containing a single repeat-spacer-repeat unit is provided. Any sequence might be inserted between two repeats in the cassette using suitable restriction endonucleases. Cassette might be used to target sequences in vivo, or to produce RNA ribonucleotide suitable for complex assembly in vitro.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1 shows Cas9 protein co-purifies with crRNA. (A) Schematic representation of CRISPR3/Cas system of S. thermophilus. Four cas genes (cas9, cas1, cas2, csn2) are located upstream of the CRISPR repeat-spacer array, consisting of 13 repeat (R) sequences and 12 unique spacers (S1-S12). The tracrRNA, required for crRNA maturation in Type II CRISPR systems (Deltcheva et al., 2011. Nature 471:602-7), is located upstream the cas9 gene and encoded on the opposite DNA strand (showed by an arrow) in respect to the other elements of CRISPR3/Cas system. (B) Schematic representation of heterologous loci in two plasmids used for the co-expression of the Cas9-crRNA complex. E. coli RR1 strain contained pCas9(−)1 SP (encoding Cas1, Cas2, Csn2, SP1 and tracrRNA) and pASKIBA-Cas9 (encoding Strep-tagged version of Cas9) plasmids. (C) Northern analysis of Cas9-crRNA complexes using anti-crDNA oligonucleotide as a probe. M1-84 nt oligodeoxynucleotide corresponding to the spacer S1-repeat unit; M2-42 nt synthetic oligoribonucleotide corresponding to the predicted S. thermophilus CRISPR3 crRNA (See FIG. 4); crRNA (wt)-crRNA isolated from the wt Cas9 complex; K1-crRNA (wt) treated with Dnase I for 15 min; K2-crRNA (wt) treated with RNaseI for 15 min, D31A-crRNA purified from the Cas9 D31A mutant complex; N891A-crRNA purified from the Cas9 N891A mutant complex.

FIG. 2 shows DNA cleavage by Cas9-crRNA complexes obtained by Cas9 co-expression with full length CRISPR locus. (A) Schematic representation of CRISPR/Cas locus of recombinant pCas9(−) plasmid carrying indigenous 12 spacer-repeat array of SthCRISPR3/Cas system and pASKIBA-Cas9 plasmid carrying cas9 gene with a Strep-tag at the C-terminus. (B) Oligoduplex cleavage assay. Both pCas9(−) and pASKIBA-Cas9 plasmids were co-expressed in E. coli, Cas9-crRNA complexes were purified and subjected to cleavage analysis using SP1 (first proto-spacer) and SP2 (second proto-spacer) oligoduplexes labeled with 33P at the 5′-end of the (+) strand. Reaction products were analysed on PAA gel.

FIG. 3 shows immunity against plasmid transformation in E. coli cells provided by the SthCRISPR3/Cas system. (A) Schematic representation of CRISPR/Cas locus of recombinant plasmid pCRISPR3 carrying indigenous 12 spacer-repeat array of SthCRISPR3/Cas system and engineered pCRISPR3-SP1 plasmid carrying 1 spacer-repeat unit. (B) Interference of plasmid transformation by SthCRISPR3/Cas system in E. coli cells. Escherichia coli RR1 recipient strains carrying plasmids pACYC184, pCRISPR3 or pCRISPR3-SP1, were transformed with plasmid pSP1 carrying proto-spacers and PAM or pUC18 (1). Transformation efficiency is expressed as cfu per nanogram of plasmid DNA (mean±SD).

FIG. 4 shows comparison of Type IIA CRISPR/Cas systems from S. thermophilus DGCC7710, LMD-9 and S. pyogenes SF370 strains. (A) Schematic organization of the CRISPR/Cas systems. Nucleotide sequences corresponding to the tracrRNA required for the crRNA maturation in of S. pyogenes (2) are present in LMD-9 and DGCC7710. Percentage of identical and similar (in parenthesis) residues between corresponding protein sequences that are connected by dashed lines. (B). Alignment of the conserved repeat sequences and tracrRNA. Corresponding sequences from DGCC7710 and LMD-9 are identical. Nucleotide positions which are identical in all three strains are labeled with an asterisk below aligned sequences. FIG. 4(B) discloses SEQ ID NOS 50, 50-52, and 52-53, respectively, in order of appearance. (C) Comparison of crRNA sequences. The sequence and length of S. pyogenes crRNA was determined by deep sequencing analysis (2). The approximate length of crRNA from S. thermophilus LMD-9 (2) and DGCC7710 (this work) strains were determined by the northern blot analysis. FIG. 4(C) discloses SEQ ID NOS 54-56, respectively, in order of appearance.

FIG. 5 shows Cas9-crRNA complex cleaves in vitro double-stranded DNA within a proto-spacer. (A) Oligoduplex substrate used in the cleavage assay. 55 nt oligoduplex SP1 contains the proto-spacer1 (red letters), PAM (blue letters) and 10 nt flanking sequences on both sides identical to those in pSP1 plasmid. In the SP1 oligoduplex DNA strand complimentary to the 5-terminal fragment of crRNA (red letters) is named (+)strand, an opposite DNA strand is named (−)strand. FIG. 5(A) discloses SEQ ID NOS 31, 7, and 34, respectively, in order of appearance. (B) Oligoduplex SP1 cleavage. 2.5 nM of Cas9-crRNA complex and 1 nM SP1 oligoduplex labeled with 33P at the 5′-end of either (+) or (−) strand were incubated in the reaction buffer (10 mM Tris-HCl pH=7.5, 10 mM NaCl, 10 mM MgCl2, 0.1 mg/ml BSA) at 37° C. for varied time intervals (30 s to 10 min) and reaction products analysed in the 20% PAA gel. Lanes M1 and M2 contain chemically synthesized 5′-end 33P-labeled 37 nt and 18 nt oligodeoxynucleotides corresponding to the cleavage products of (−) and (+) DNA strands, respectively. Cleavage positions are designated by arrows. FIG. 5(B) discloses SEQ ID NO: 31. (C) Schematic representation of pSP1 plasmid (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) used in the plasmid cleavage assay. FIG. 5(C) discloses SEQ ID NO: 57. (D) pSP1 plasmid cleavage. Agarose gel analysis of pSP1 cleavage products (left panel). SC—super-coiled plasmid DNA, OC—open circular DNA nicked at one of the strands, FLL—full length linear DNA cut at both strands. Final reaction mixtures at 37° C. contained 2.5 nM of pSP1 plasmid and 2.5 nM of Cas9-crRNA complex in the reaction buffer (section B). Direct sequencing electropherograms (right panel) of (+) (upper part) and (−) (lower part) strands of pSP1 plasmid cleavage product. The non-templated addition of adenine (T in the reverse complement sequence shown here) at the extremity of sequence is a sequencing artefact caused by the polymerase. FIG. 5(D) discloses SEQ ID NOS 57-59, 58, and 60, respectively, in order of appearance.

FIG. 6 shows DNA binding and cleavage analysis of Cas9-Chis protein lacking crRNA. Electrophoretic mobility shift analysis (EMSA) of Cas9-Chis protein binding to (A) the double stranded SP1 oligoduplex and (B) the single stranded s(+)SP1 oligonucleotide. Electrophoretic mobility shift experiments were performed in the binding buffer (40 mM Tris-acetate, pH 8.3 at 25 C, 0.1 EDTA, 0.1 mg/ml BSA, 10% v/v glycerol). The reactions contained 0.5 nM of the 33P-labelled oligoduplex, and the protein at concentrations as indicated above each lane. (C). Oligonucleotide cleavage assay. 5 nM of Cas9-Chis protein was incubated in the reaction buffer (10 mM Tris-HCl, pH=7.5, 10 mM NaCl, 10 mM MgCl2, 0.1 mg/ml BSA) at 37° C. with 1 nM oligonucleotide. SP1 oligoduplex was labeled with 33P at the 5′-end of the (+) or (−) strand. Single stranded oligonucleotide s(+)SP1 was labeled with 33P at the 5′-end.

FIG. 7 shows reprograming of Cas9-crRNA complex. (A) Schematic representation of heterologous loci in two plasmids used for reprogramming of Cas9-crRNA complex. pCas(−)SPN were constructed from pCas9(−) plasmid (See FIG. 2A), by inserting new spacer sequence (SN) (5′-CC ACC CAG CAA AAT TCG GTT TTC TGG CTG-3′ (SEQ ID NO: 16)) and inactivating Cas9 gene as described in (1). (B) Agarose gel analysis of plasmid DNA cleavage products. pSP1 and pSP1+SPN (pSP1 plasmid with inserted new proto-spacer and PAM over AatII site were incubated at 2.5 nM concentration with 2 nM of Cas9-crRNA complex in the reaction buffer (10 mM Tris-HCl pH=7.5, 10 mM NaCl, 10 mM MgCl2, 0.1 mg/ml BSA) at 37° C. for varied time intervals and reaction products analysed in the agarose gel. SC—super-coiled plasmid DNA, OC—open circular DNA nicked at one of DNA strands, FLL—full length linear DNA cut at both strands. (C) Oligoduplex SP1 cleavage. 2.5 nM of Cas9-crRNA complex and 1 nM SPN oligoduplex (Table S2) labeled with 33P at the 5′-end of either (+) or (−)strand were incubated in the reaction buffer (10 mM Tris-HCl pH=7.5, 10 mM NaCl, 10 mM MgCl2, 0.1 mg/ml BSA) at 37° C. M1-18 nt length marker Lanes M1 and M2 contain chemically synthesized 5′-end 33P-labeled 18 nt and 37 nt oligodeoxynucleotides corresponding to the cleavage products of (+) and (−) DNA strands, respectively. (D) Schematic representation of SPN oligoduplex substrate and cleavage products. SPN oligoduplex contains the new proto-spacer (red letters), PAM (blue letters). Cleavage positions are designated by arrows. FIG. 7(D) discloses SEQ ID NO: 39.

FIG. 8 shows impact of spacer length on CRISPR-encoded immunity. (A) Schematic representation of shortened versions of proto-spacers inserted in the transformed plasmids. FIG. 8(A) discloses SEQ ID NOS 7 and 61-66, respectively, in order of appearance. (B) Effect of proto-spacer length on the plasmid transformation efficiency. Transformation efficiency is expressed as cfu per nanogram of plasmid DNA (mean±SD). (C). Schematic representation of oligoduplexes used in the in vitro cleavage and binding experiments. FIG. 8(C) discloses SEQ ID NOS 31 and 38, respectively, in order of appearance. (D) Time courses of the 27 bp oligoduplex (full length protospacer SP1, filled circles) and the 20 bp oligoduplex (truncated protospacer SP1-20, square) cleavage by the Cas9-crRNA complex. (E) Electrophoretic mobility shift assay of SP1 and SP1-20 oligoduplex binding by the Cas9-crRNA complex.

FIG. 9 shows PAM is required for in vitro DNA binding and cleavage by the Cas9-crRNA complex. (A) Agarose gel analysis of plasmid DNA cleavage products. Three different plasmids: PAM+Proto-spacer+ (pSP1 plasmid containing both the proto-spacer and PAM), PAM-Protospacer− (pUC18 plasmid containing multiple PAMs but no protospacer) and PAM-Protospacer+(pSP1-pΔ (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) containing a proto-spacer without PAM) were incubated at 2.5 nM concentration with 2 nM of Cas9-crRNA complex in the reaction buffer (10 mM Tris-HCl pH=7.5, 10 mM NaCl, 10 mM MgCl2, 0.1 mg/ml BSA) at 37° C. for varied time intervals and reaction products analysed in the agarose gel. SC—super-coiled plasmid DNA, OC—open circular DNA nicked at one of DNA strands, FLL—full length linear DNA cut at both strands. (B) Time courses of (+)strand hydrolysis in the single-stranded and double-stranded oligodeoxynucleotides. Reactions containing 2 nM Cas9-crRNA and 1 nM of oligodeoxynucleotide were conducted at 37° C. in the reaction buffer (section A). SP1 (filled circles) and SP1-pΔ (open squares) oligoduplexes were used as dsDNA. s(+)SP1 (open triangles) and s(+) SP1-pΔ (filled squeres) were used as ssDNA. (C) and (D) dsDNA and ssDNA (+)strand) binding by Cas9-crRNA complex. The reactions contained 0.5 nM of the 33P-labelled ssDNA or dsDNA oligonucleotide, and the protein at concentrations as indicated above each lane. After 15 min at room temperature, the samples were subjected to PAGE for 2 h and analysed as described in ‘Materials and Methods’

FIG. 10 shows RNA binding and cleavage analysis of Cas9-crRNA complex. (A) Electrophoretic mobility shift analysis (EMSA) of Cas9-crRNA complex binding to 84 nt RNA fragment containing proto-spacer-1, PAM and 24 nt flanking sequences on both sides. Left panel: RNA (−) strand; center panel: RNA (+) strand; right panel: double stranded RNA. RNA fragments used for analysis were generated by in vitro transcription (TranscriptAid™ T7 High Yield Transcription Kit, Fermentas) from PCR fragments with inserted T7 promoter at the front end of RNA coding sequence. PCR fragments coding (+) and (−) RNA strands were obtained from pSP1 plasmid (1) with following primer pairs accordingly: 5′ taatacgactcactataGggtaccgagctcgaattg 3′ (SEQ ID NO: 17)/5′ GGGAAACAGCTATGACCATGATTACGAATTC-3′ (SEQ ID NO: 18) and 5′ gggtaccgagctcgaattgaaattcTAAACG 3′ (SEQ ID NO: 19)/5′ taatacgactcactataGggAAACAGCTATGACCATGATTACG 3′ (SEQ ID NO: 20) (T7 RNA polymerase promoter underlined, transcription start on bold). The reactions contained 1 nM of the 33P-labelled RNA fragment, and the protein at concentrations as indicated above each lane. After 15 min at room temperature, the samples were subjected to PAGE for 2 h and analyzed as described in ‘Materials and Methods’. (B) RNA cleavage assay. 2.5 nM of Cas9-crRNA complex was incubated in the reaction buffer (10 mM Tris-HCl pH=7.5, 10 mM NaCl, 10 mM MgCl2, 0.1 mg/ml BSA,) at 37° C. in the presence of 1 nM (+) and (−) RNA strands(left panel) or double stranded RNA labeled on (+) or (−) strand (right panel). Reaction products were analysed on denaturing PAA gel.

FIG. 11 shows RuvC and HNH active site motifs of Cas9 contribute to the cleavage of opposite DNA strands. (A) Localization of the conserved active site motifs within Cas9 protein. Amino acid residues identified as crucial for Cas9 in vivo activity (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) are indicated. (B). Agarose gel analysis of pSP1 plasmid cleavage by Cas9 and mutant proteins. Reactions were performed as described in and ‘Materials and Methods’ (C) Strand preference of D31A mutant. Reactions were performed as described in FIG. 2A and ‘Materials and Methods’. D31 mutant cleaves only (+)strand of SP1 oligoduplex. FIG. 11(C) discloses SEQ ID NOS 31 and 67, respectively, in order of appearance. (D) Strand preference of N891A mutant. N891 mutant cleaves only (−)strand of SP1 oligoduplex. Cleavage positions are designated by arrows. FIG. 11(D) discloses SEQ ID NOS 31 and 68, respectively, in order of appearance.

FIG. 12 shows properties of Cas9 active site mutant-crRNA complexes. (A) Direct sequencing of reaction products obtained with Cas9 mutant D31A (RuvC-like active site motif). FIG. 12(A) discloses SEQ ID NOS 58, 59, 58, and 58, respectively, in order of appearance. (B) Direct sequencing of reaction products obtained with Cas9 N891A mutant (HNH-like active site motif). FIG. 12(B) discloses SEQ ID NOS 58, 58, 58, and 60, respectively, in order of appearance. (C) SP1 oligoduplex binding by the wt Cas9-crRNA and active site mutant complexes. (D) Cleavage of (+)SP1 strand by Cas9-crRNA mutant complexes.

FIG. 13 shows molecular mass of the wt Cas9-Chis protein. Gel filtration experiments were carried out at room temperature using Superdex 200 10/300 GL column (GE healthcare) pre-equilibrated with 10 mM sodium phosphate (pH 7.4) buffer containing 500 mM sodium chloride. The apparent Mw of Cas9 (black triangle) were calculated by interpolation from the standard curve obtained using a set of proteins of known Mw (black circles) (Bio-Rad Gel Filtration Standards).

FIG. 14 shows schematic arrangement and mechanism of crRNA-directed DNA cleavage by the Cas9-crRNA complex. Domain architecture of Cas9 is shown schematically on the top. Cas9-crRNA complex binds to the dsDNA containing PAM. crRNA binds to the complementary (+)strand resulting in DNA strand separation and the R-loop formation. In the ternary complex RuvC active site of Cas9 is positioned at the scissile phosphate on the unpaired (−)strand, while HNH active site is located at the scissile phosphate on the DNA (+)strand bound to crRNA. Coordinated action of both active sites results in the double strand break 4 nt away from the PAM generating blunt end DNA. FIG. 14 discloses SEQ ID NOS. 31 and 69, respectively, in order of appearance.

FIG. 15 shows native electrophoresis of Cas9-crRNA and cleavage products. The protein at concentrations as indicated above each lane, where incubated in the reaction buffer (10 mM Tris-HCl pH=7.5, 10 mM NaCl, 10 mM MgCl2, 0.1 mg/ml BSA) at 37° C. for 30 min in the presence of 0.5 nM SP1 oligoduplex. Samples was mixed with loading dye solution (0.01% bromphenol blue and 75 mM EDTA in 50% v/v glycerol) and analysed by non-denaturing PAGE. The gel lanes marked M—melted form of cleavage reactions products. The cartoons in each side of the gel illustrate protein-DNA complexes and DNA that correspond to each band, while cartoons below the gel illustrate major substrate form after reaction.

FIG. 16 shows plasmid DNA cleavage by Cas9-crRNA complex. (A) pSP1 and pUC18 plasmid DNA cleavage. Cas9-crRNA complex was incubated with pSP1 and pUC18 plasmids in a reaction buffer provided in the Example 1. pSP1 plasmid contained a proto-spacer1 sequence flanked by the the 5′-GGNG-3′PAM sequence. Proto-spacer1 sequence was not present in pUC18. Reaction products were analysed in the agarose gel. Under these conditions pSP1 plasmid is converted into a linear form while pUC18 plasmid lacking proto-spacer1 sequence is resistant to cleavage. (B) pSP1 cleavage reactions in the absence of one of the components. In the reaction mixes lacking one of the components (Cas9, crRNA or tracrRNA, respectively) pSP1 plasmid is not cleaved. SC—super-coiled plasmid DNA, OC—open circular DNA nicked at one of DNA strands, FLL—full length linear DNA cut at both strands.

FIG. 17 shows DNA oligoduplex cleavage by Cas9-crRNA complex. The strand of oligoduplex which is complementary to crRNA is marked as (+) strand, while the other strand—(−) strand. To monitor cleavage reactions either (+) or (−) strand of the oligoduplex was P33-labeled at the 5′-terminus. M1 and M2 are synthetic oligonucleotide markers corresponding to the 37 nt of (−) strand and 18 nt of (+) strand which were used to determine the size of the cleavage products and map the cleavage position. Cas9 protein cleaves both strands of oligoduplex inside the proto-spacer, after the 37th nucleotide, 4 nt upstream of the PAM (5′-GGNG-3′) leaving blunt ends. Both strands of non-specific substrate (K1 and K2) are not cleaved when incubated with Cas9-crRNA complex for 30 min. FIG. 17 discloses SEQ ID NO: 31.

FIG. 18 shows plasmid DNA cleavage by Cas9-crRNA complex assembled in the absence of RNaseIII. Cas9-crRNA complex was incubated with pSP1 plasmid and reaction products analysed in the agarose gels. The pSP1 plasmid is resistant for cleavage in the presence of complex assembled without crRNA (left panel). The pSP1 plasmid is converted into linear form in the presence of complex assembled using synthetic 42 nt crRNA (no RNAseIII) (middle panel). The pSP1 plasmid is converted into a mixture of linear and circular DNA forms in the presence of complex assembled using CRISPR RNA transcript (no RNAseIII) (right panel).

FIG. 19 shows DNA oligoduplex cleavage by Cas9-crRNA complex. The strand of oligoduplex which is complementary to crRNA is marked as (+) strand, while the other strand—(−)strand. To monitor cleavage reaction either (+) or (−) strand of the oligoduplex was P33-labeled at the 5′-terminus. M1 and M2 are synthetic oligonucleotide markers corresponding to the 37 nt of (−) strand and 18 nt of (+) strand which were used to determine the size of the cleavage products and map the cleavage position. Cas9 protein cleaves both strands of oligoduplex inside the proto-spacer, after the 37th nucleotide form the 5′-end, 4 nt upstream of the PAM (5′-GGNG-3′) leaving blunt ends. Both strands of non-specific substrate (K1 and K2) are not cleaved when incubated with Cas9-crRNA complex for 30 min. FIG. 19 discloses SEQ ID NO: 31.

FIG. 20 shows (A) Schematic representation of the CRISPR3/Cas system of S. thermophilus DGCC7710. Four cas genes (cas9, cas1, cas2, csn2) are located upstream of the CRISPR repeat-spacer array, consisting of 13 repeat (R) sequences and 12 unique spacers (S1-S12). The tracrRNA, required for crRNA maturation in Type 11 CRISPR/Cas systems (Deltcheva et al., 2011. Nature 471, 602-7), is located upstream the cas9 gene and encoded on the opposite DNA strand (shown by an arrow) with respect to the other elements of this system. (B) The pathways for a new spacer insertion in to CRISPR region and CRISPR RNA synthesis. Synthetic oligoduplex encoding desired spacer sequence and containing SapI and Eco31I restriction compatible ends was inserted between two repeats. The CRISPR region was amplified using PCR. The new spacer encoding CRISPR RNA was obtained by in vitro transcription. (C) In vitro assembly of Cas9-RNA complex. The CRISPR RNA and tracrRNA transcripts were assembled in to duplex. The Cas9 protein was first pre-incubated with RNA duplex, followed by the subsequent incubation with RNAseIII to generate a catalytically competent Cas9-RNA complex.

FIG. 21 shows A. Schematic representation of pUC18 plasmid. The distance between SapI and AatII restriction sites is 775 bp, while the distance between two spacers is 612 bp. B. pUC18 plasmid cleavage by re-programed Cas9-crRNA complexes. “1”—pUC18 plasmid; “2”—pUC18 cleaved with AatII: “3”—pUC18 cleaved with complex containing crRNA matching proto-spacer1; “4”—pUC18 cleaved with SapI; “5”—pUC18 cleaved with complex containing crRNA matching proto-spacer2; “6”—pUC18 cleaved with AatII and SapI; “7”—pUC18 cleaved with mix of the complexes used in the line 3 and 5.

FIG. 22 shows genomic DNA cleavage with in vitro assembled Cas9-RNA complex. (A) Agarose gel analysis of linear λ DNA cleavage products. Phage A DNA was incubated with Cas9-RNA complex in the reaction buffer for various time intervals. The target site for Cas9-RNA complex is located 8 kb away from the cos site. (B). Probe selection for Southern blot experiments. Genomic DNA was fragmented by treating with PstI enzyme. The proto-spacer is located between two PstI sites. If genomic DNA is cleaved with Cas9-RNA complex, 466 bp fragment should be detected. Otherwise the probe will hybridize with 1499 bp length fragment. (C) Southern blot analysis of genomic DNA fragments. C line—E. coli genomic DNA fragmented with PstI. Cas9-RNA—genomic DNA was incubated with Cas9-RNA complex before fragmentation. (D). Human genomic DNA cleavage by Cas9-crRNA complex. Relative amount of intact DNA DNA fragments were estimated by qPCR.

FIG. 23 schematically illustrates targeting sequences contained in the reporter plasmid (pMTC-DSR+eGFP). eGFP coding sequence is separated by an intron from GAPDH gene. The 5′ and 3′ RFP coding sequences are indicated. homol indicates homologous sequences in the RFP gene necessary for homologous recombination to occur. A, B, C, and D indicate four distinct target sites for Cas9-mediated cleavage. Targets A and B are located in the intron. Targets C and D are located in the coding regions of eGFP. Cre indicates a target site for Cre endonuclease and is located in the intronic sequence.

FIG. 24 shows reduction of eGFP-positive cells after introduction of Cas9/RNA complexes. CHO-K1 cells were transfected with the reporter plasmid and Cas9/RNA complexes containing crRNA targeting either eGFP sequence A (intronic), eGFP sequence C (coding), or a non-specific sequence K. The percentage of eGFP-positive cells was determined by flow cytometry. As negative controls, cells were untransfected (NC) or transfected with the reporter plasmid alone (DNA) or with reporter plasmid and Cas9 protein alone as well as with reporter plasmid and Cas9-nonspecific crRNA complex (DNA+K).

FIG. 25 shows cell images where appearance of RFP suggested Cas9IRNA-mediated double-strand break repair by homologous recombination (HR). Forty-eight hours after co-transfection with the reporter plasmid and Cas9/RNA complexes targeting eGFP sequence C, CHO-k1 cells were visualized by fluorescence microscopy for eGFP and RFP.

FIG. 26 schematically illustrates targeting sequences contained in the reporter plasmid (pMTC-DSR+eGFP). eGFP coding sequence is separated by GAPDH intron copied from genomic DNA. The RFP N- and C-coding sequences are as indicated. Homologous sequences in the RFP gene are necessary for homologous recombination to occur. Target E located within the intron of eGFP is indicated in bold.

FIG. 27 is a gel showing Cas9/RNA complexes using synthetic crRNA and tracRNA function similarly to Cas9/RNA complexes using synthetic crRNA and in vitro transcribed tracrRNA. Plasmids were visualized after agarose gel electrophoresis. Lane C: uncut plasmid. Lanes 1-3: plasmids cut with Cas9+crRNA and either 1: control in vitro-transcribed tracrRNA; 2: unmodified synthetic tracrRNA (89 nt); or 3: unmodified synthetic tracrRNA (74 nt).

FIGS. 28A-E schematically show targeting sequences contained in the reporter plasmid (pMTC-DSR+eGFP) and potential processing/gene rearrangement outcomes.

FIG. 29 shows reduction of eGFP-positive cells after introduction of Cas9/RNA complexes. CHO-K1 cells were transfected with the reporter plasmid and Cas9/RNA complexes containing crRNA targeting either eGFP sequence A (intronic), eGFP sequence C (coding), or a non-specific sequence K. The percentage of eGFP-positive cells was determined by flow cytometry. As negative controls, cells were untransfected (NC) or transfected with the reporter plasmid alone (DNA) or with reporter plasmid and Cas9 protein alone as well as with reporter plasmid and Cas9-nonspecific crRNA complex (DNA+K).

FIG. 30 schematically shows targeting sequences contained in the reporter plasmid (pMTC-DSR+eGFP). eGFP coding sequence is indicated in black and is separated by GAPDH intron copied from genomic DNA. The RFP N- and C-coding sequences are indicated in gray. Homologous sequences in the RFP gene (light grey) are necessary for homologous recombination to occur. Target E located within the intron of eGFP is indicated in bold.

The following non-limiting examples further describe the methods, compositions, uses, and embodiments.

DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS Example 1

In this example, we have isolated the Cas9-crRNA complex of S. thermophilus CRISPR3/Cas system and demonstrate that it cuts in a PAM dependent manner both synthetic oligodeoxynucleotide and plasmid DNA bearing a nucleotide sequence complementary to the crRNA. Furthermore, we provide experimental evidence that PAM is recognized in the context of double-stranded DNA and is critical for in vitro DNA binding and cleavage. Finally, we show that RuvC and HNH-motifs of Cas9 contribute to the cleavage of opposite DNA strands. Taken together, our data demonstrate that Cas9-crRNA complex functions as RNA-guided endonuclease which uses RNA module for the target site recognition and employs two separate active sites in the protein module for DNA cleavage. These findings pave the way for engineering of programmable Cas9-crRNA complexes as universal RNA-guided endonucleases.

Materials and Methods

DNA manipulations. Genomic DNA of Streptococcus thermophilus DGCC7710 strain was used as a template in PCR reactions to clone cas9. To generate a pASKIBA3-Cas9 plasmid which was used for the expression of the C-terminal Strep-tagged Cas9 protein variant, PCR fragment amplified with following primers: 5′-ACGTCTCAAATGTTGTTTAATAAGTGTATAATAATTTC-3′ (SEQ ID NO: 21) and 5′-ACGTCTCCGCGCTACCCTCTCCTAGTTTG-3′ (SEQ ID NO: 22) was cloned into the pASK-IBA3 expression vector via Esp3I sites. To generate a pBAD-Cas9 plasmid which was used for the expression of the C-terminal 6×His-tagged Cas9 protein variant (“6×His” disclosed as SEQ ID NO: 23), PCR fragment amplified with the following primer pair: 5′-ACGTCTCACATGACTAAGCCATACTCAATTGGAC-3′ (SEQ ID NO: 24) and 5′-ACTCGAGACCCTCTCCTAGTTTGGCAA-3′ (SEQ ID NO: 25) was cloned into the pBAD24-Chis expression vector via NcoI and XhoI sites. Full sequencing of cas9 gene in pASKIBA3-Cas9 and pBAD-Cas9 plasmids revealed no difference with the original cas9 sequence. To obtain plasmids pCas9(−)SP1 (FIG. 1B) and pCRISPR3-SP1 (FIG. 2A). bearing a single spacer1, PCR fragment amplified from pCRISPR3 plasmid with the following primer pair:5′ GACCACTTATTGAGGTAAATGAG 3′ (SEQ ID NO: 26)/5′ CAAACCAGGATCCAAGCTAATACAGCAG-3′ (SEQ ID NO: 27) ((BamHI (GGATCC) sites is underlined) was cloned into pCas9(−) and pCRISPR3 plasmids (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82), respectively.

Expression and purification of Cas9 protein and Cas9-crRNA complex. (His)6-tagged (“(His)6” disclosed as SEQ ID NO: 23) version of Cas9 protein was expressed and purified using a scheme described for the Cas3 protein from S. thermophilus CRISPR4/Cas system (Sinkunas et al., 2011. EMBO J 30:1335-42). For purification of the Cas9-crRNA complex, Strep-tagged version of the Cas9 protein was expressed in E. coli RR1 strain, bearing pCas9(−)SP1 plasmid (FIG. 1B). LB broth was supplemented with Ap (100 μg/ml) and Cm (10 μg/ml). E. coli cells for the Cas9-crRNA complex isolation were grown in two steps. First, 4 ml of cells culture were grown at 37° C. to OD600 of ˜0.5, and expression induced by adding 0.2 μg/ml of anhydrotetracycline (AHT) (Sigma). After for 4 h, 1/400 of the pre-induced culture was inoculated into fresh LB medium supplemented with Ap (100 μg/ml), Cm (12 μg/ml) and AHT (0.2 μg/ml) and was grown at 37° C. overnight. Harvested cells were disrupted by sonication and cell debris removed by centrifugation. The supernatant was loaded onto the 1 ml StrepTrap HP column (GE Healthcare) and eluted with 2.5 mM of desthiobiotin. Approximately 1.5 μg of the Cas9 protein was obtained in a single run from 1 L of E. coli culture. The fractions containing Cas9 were stored at +4° C. for several days. The homogeneity of protein preparations was estimated by SDS-PAGE. Protein concentrations in the Cas9-crRNA complexes were determined by densitometric analysis of SDS-PAGE gels containing samples of Strep-Tactin purified Cas9 proteins along with known amounts of His-tagged Cas9 protein. The concentration of the Cas9-crRNA complexes is expressed as Cas9 protein concentration assuming that Cas9 is a monomer and binds crRNA in a complex with 1:1 stoichiometry.

Northern blot analysis. Cas9-bound RNA was isolated from Strep-Tactin purified Cas9, co-expressed with pCas9(−)SP1 plasmid using the miRNeasy Mini kit (Qiagen). Northern blots were performed by running RNA on a 10% polyacrylamide gel with 7 M urea in 20 mM MOPS/NaOH pH 8 buffer. The RNA was transferred to a SensiBlot™ Plus Nylon Membrane (Fermentas) by semi-dry blotting using a Trans-blot SD (Bio-Rad). RNA was cross-linked to the membrane with 0.16 M l-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) (Pierce)/0.13 M 1-methylimidazole (Sigma) pH 8 at 60° C. for 1 h. The membrane was pre-hybridized with 2×SSC buffer containing 1% SDS and 0.1 mg/ml denatured DNA from fish testes (Ambion) for 1 h at 40° C. Blots were probed for 12 h with a 32P-5′-labelled 42 nt anti-crRNA DNA oligonucleotide containing 20 nt of spacer1 and 22 nt of the repeat sequence (5′-TCGAAACAACACAGCTCTAAAACTGTCCTCTTCCTCTTTAGC-3′ (SEQ ID NO: 28)). The blots were washed 3× for 15 min with 0.2×SSC buffer containing 0.2% SDS, and were visualized using phosphorimaging. A 42 nt synthetic oligoribonucleotide (5′-CGCUAAAGAGGAAGAGGACAGUUUUAGAGCUGUGUUGUUUCG-3′ (SEQ ID NO: 7)) and 84 nt DNA oligonucleotide.

Oligonucleotide substrates. All oligonucleotide substrates used in this study are given in Table 1. Oligodeoxyribonucleotides were purchased from Metabion (Martinsried, Germany). The 5′-ends of oligonucleotides were radiolabelled using PNK (Fermentas) and [γ-33P]ATP (Hartmann Analytic). Duplexes were made by annealing two oligonucleotides with complementary sequences (SP1, SP1-Δp, SP2). Radioactive label was introduced at the 5′ end of individual DNA strand prior to the annealing with unlabelled strand.

Reactions with oligonucleotide substrates. Reactions were typically carried out by adding 2 nM of Cas9-crRNA complex to 1 nM labeled oligonucleotide in 10 mM Tris-HCl (pH 7.5 at 37° C.), 10 mM NaCl, 0.1 mg/ml BSA and 10 mM MgCl2 at 37° C. Aliquots were removed at timed intervals and quenched with loading dye (95% v/v formamide, 0.01% bromphenol blue, 25 mM EDTA, pH 9.0) and subjected to denaturing gel electrophoresis through 20% polyacrylamide followed by a FLA-5100 phosphorimager (Fujilm) detection.

Reactions with plasmid substrates. Reactions on pUC18 plasmid and its derivatives (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) were conducted at 37° C. in the buffer used for reactions on oligonucleotide substrates. Reaction mixtures typically contained 2.5 nM supercoiled plasmid and 2 nM of Cas9-crRNA complex. The reactions were initiated by adding protein to the mixture of the other components. Aliquots were removed at timed intervals and quenched with phenol/chloroform. The aqueous phase was mixed with loading dye solution (0.01% bromphenol blue and 75 mM EDTA in 50% v/v glycerol) and analyzed by electrophoresis through agarose.

Plasmid cleavage position determination. To achieve complete cleavage of plasmid substrate, 8 nM of Cas9-crRNA complex was incubated with 2.5 nM of supercoiled plasmid in the reaction buffer at 37° C. for 10 min. Reaction products were purified and concentrated using GeneJET PCR Purification Kit (Fermentas). Spacer1 surrounding region of Cas9 linearized and nicked plasmids were directly sequenced with the following primers: 5′-ccgcatcaggcgccattcgcc-3′ (SEQ ID NO: 29) (sequencing of (+)strand) and 5′-gcgaggaagcggaagagcgccc-3′ (SEQ ID NO: 30) (sequencing of (−)strand).

Binding assay. Increasing amounts of protein-crRNA complex were mixed with 0.5 nM of 33P-labeled double-stranded and single-stranded DNA substrates (Table 1) in the binding buffer (40 mM Tris-acetate, pH 8.3 at 25 C, 0.1 EDTA, 0.1 mg/ml BSA, 10% v/v glycerol) and incubated for 15 min at room temperature. Free DNA and protein-DNA complexes were separated on the non-denaturing 8% polyacrylamide gel (ratio of acrylamide/N,N′-methylenebisacrylamide 29:1) using 40 mM Tris-acetate (pH 8.3) supplemented with 0.1 mM EDTA as the running buffer. Electrophoresis was run at room temperature for 3 h at 6 V/cm.

Mutagenesis. The mutants D31A and N891A were obtained by the site-directed mutagenesis as previously described (Tamulaitis et al., 2007. Nucleic Acids Res 35:4792-9). Sequencing of the entire gene for each mutant confirmed that only the designed mutation had been introduced.

TABLE 1 Oligonucleotide substrates. Proto-spacer sequence is underlined, PAM is on bold. Oligo- nucleotide Sequence Specification SP1 5′-GCTCGAATTGAAATTCTAAACGCTAAAGAGGAAGAGGACATGGTGAATTCGTAAT-3′ 55 bp oligoduplex  (SEQ ID NO: 3′-CGAGCTTAACTTTAAGATTTGCGATTTCTCCTTCTCCTGTACCACTTAAGCATTA-5′ substrate containing 31) proto-spacer1 and PAM SP1-pA (SEQ) 5′-GCTCGAATTGAAATTCTAAACGCTAAAGAGGAAGAGGACAAATTCGTAAT-3′ 50 bp oligoduplex ID NO: 32) 3′-CGAGCTTAACTTTAAGATTTGCGATTTCTCCTTCTCCTGTTTAAGCATTA-5′ substrate containing proto-spacer2 SP2 5′-GCTCGAATTGTACTGCTGTATTAGCTTGGTTGTTGGTTTGTGGTGAATTCGTAAT-3′ 55 bp oligoduplex  (SEQ ID NO: 3′-CGAGCTTAACATGACGACATAATCGAACCAACAACCAAACACCACTTAAGCATTA-5′ substrate containing 33) proto-spacer2 and PAM (oligodublex without proto-spacer1) s (+) SP1 5′-ATTACGAATTCACCATGTCCTCTTCCTCTTTAGCGTTTAGAATTTCAATTCGAGC-3′ 55 nt ssDNA oligo- (SEQ ID NO: nucleotide substrate 34) (+) strand of SP1 oligoduplex s (+) SP1-pΔ 5′-ATTACGAATTTGTCCTCTTCCTCTTTAGCGTTTAGAATTTCAATTCGAGC-3′ 50 nt ssDNA oligo- (SEQ ID NO: nucleotide substrate 35) (+) strand of SP1- pa oligoduplex s (+) SP2 5′-ATTACGAATTCACCACAAACCAACAACCAAGCTAATACAGCAGTACAATTCGAGC-3′ 55 nt ssDNA oligo- (SEQ ID NO: nucleotides ubstrate, 36) (+) strand of SP2 oligoduplex s (-) SP1 5′-GCTCGAATTGAAATTCTAAACGCTAAAGAGGAAGAGGACATGGTGAATTCGTAAT-3′ 55 nt ssDNA oligo- (SEQ ID NO: nucleotide substrate, 37) (-) strand of SP1 oligoduplx SP1-20 (SEQ 5′-GCTCGAATTGCGCTAAAGAGGAAGAGGACATGGTGAATTCGTAAT-3′ 45 nt oligoduplex ID NO: 38) 3′-CGAGCTTAACGCGATTTCTCCTTCTCCTGTACCACTTAAGCATTA-5′ substrate containing 20 nt of proto- spacer1 and PAM SPN (SEQ ID 5′-GCTCGAATTGCCACCCAGCAAAATTCGGTTTTCTGGCTGATGGTGAATTCGTAAT-3′ 55 bp oligoduplex NO: 39) 3′-CGAGCTTAACGGTGGGTCGTTTTAAGCCAAAAGACCGACTACCACTTAAGCATTA-5′ substrate containing proto-spacerN and PAM

Results

Expression and purification of the Cas9-crRNA complex. The cas9 gene from the CRISR3 system of S. thermophilus DGCC7710 strain was cloned into the pASK-IBA3 vector to produce a construct encoding a Cas9 protein fusion containing a C-terminal Strep(II)-tag (FIG. 1B). Initially, we have tried to purify Cas9-crRNA complex from E. coli strain RR1 expressing Cas9 protein on the pASK-IBA3 vector and other Cas proteins (except Cas9) on pCas9(−) plasmid (Sapranauskas et al, 2011). pCas9(−) also contained a complete CRISPR3 array comprised of 12 spacer-repeat units (FIG. 2A). To achieve simultaneous transcription of all target genes we performed cas9 gene expression in two steps. First, we induced Cas9 expression in a small volume of E. coli culture and after 4 h transferred an aliquot of pre-induced culture into a larger volume of fresh LB media already containing inductor and incubated overnight. Cas9 protein complex was purified from the crude cell extract using Strep-Tactin Sepharose. We managed to isolate a small amount of the Cas9-crRNA complex which showed only traces of nucleolytic activity on the oligoduplex SP1 containing a proto-spacer1 and PAM. We assumed that low cleavage activity could be due to the intrinsic heterogeneity of Cas9-crRNA complexes resulting from the transcription of 12 spacer-repeat units. If all spacer-repeat units are uniformly transcribed into a mature crRNA, the concentration of the Cas9 complex containing crRNA against spacer-1 will make 1/12th fraction of the total Cas9-crRNA concentration. The cleavage activity of the Cas9-crRNA preparation against the SP2 oligoduplex containing a proto-spacer-2 and PAM is consistent with the heterogeneity of Cas9-crRNA complexes (FIG. 2B). To increase the yield of the specific Cas9-crRNA complex we engineered a pCas9(−)SP1 plasmid which contains a single R-spacer1-R unit in the CRISPR array (FIG. 1B). Plasmid transformation interference assay confirmed that the CRISPR3/Cas system carrying a single spacer1 prevents plasmid pSP1 transformation in E. coli with the same efficiency as the CRISPR3/Cas system carrying a complete CRISPR region (FIG. 3B). We have isolated Cas9-crRNA complex following the procedure described above and analysed crRNA bound to Cas9 protein.

Cas9 protein co-purifies with crRNA. CRISPR3/Cas system of S. thermophilus belongs to the Type IIA subtype (former Nmeni or CASS4) of CRISPR/Cas systems (Makarova et al., 2011. Nat Rev Microbiol 9:467-77). It has been shown that in the Type IIA CRISPR/Cas system of Streptococcus pyogenes trans-encoded small RNA (tracrRNA) and bacterial RNaseIII are involved in the generation of crRNA (Deltcheva et al., 2011. Nature 471:602-7). Streptococcus pyogenes crRNA is only 42 nt in length and has no “5′-handle” which is conserved in crRNA's from Type I and III CRISPR systems (Hale et al., 2009. Cell 139:945-56; Jore et al., 2011. Nat Struct Mol Biol 18:529-36). According to the northern blot analysis crRNA of similar length is generated in the S. thermophilus LMD-9 CRISPR3/Cas system (Makarova et al., 2011. Nat Rev Microbiol 9:467-77), which is almost identical to the CRISPR3/Cas system of DGCC7710 strain (FIGS. 4A and B). We assumed that crRNA isolated from the Cas9-crRNA complex expressed in the heterologous E. coli strain (FIG. 1) may have the same length (FIG. 4). Therefore, to probe nucleic acids extracted from the Strep-Tactin purified Cas9 complex we used 42 nt anti-crRNA DNA oligonucleotide comprised of 22 nt region corresponding to the 3′-end of the repeat sequence and 20 nt at the 5′-end of SP1 fragment. Nucleic acid present in the Cas9 complex hybridized with anti-crRNA oligonucleotide, and was sensitive to RNAse but not DNAse treatment (FIG. 1C). The size of extracted crRNA was identical to the 42 nt synthetic oligoribonucleotide corresponding to the putative crRNA of the CRISPR3 system of S. thermophilus DGCC7710 strain (FIG. 3A, FIG. 4C). Taken together, these data confirm that Cas9 Strep-tag protein co-purifies with 42 nt crRNA, which is derived from CRISPR3 region.

Cas9 protein cleaves double-stranded DNA within a proto-spacer. To test in vitro activity of purified Cas9-crRNA complex we first used the SP1 oligoduplex (Table 1) containing the proto-spacer sequence identical to spacer SP1 in the CRISPR3 array, the PAM sequence 5′-TGGTG-3′ downstream of the proto-spacer, and 10 nt flanking sequences from pSP1 plasmid (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) (FIG. 5A). The oligoduplex strand complementary to crRNA is named (+) strand, while the opposite duplex strand is called the (−) strand. To monitor cleavage reaction either (+) or (−) strand of the SP1 oligoduplex was P33-labeled at the 5′-terminus. Data shown in FIG. 5B demonstrate that the Cas9-crRNA complex cleaves both strands of oligoduplex at fixed position. Mapping of the cleavage position using synthetic oligonucleotides as size markers revealed that the Cas9-crRNA complex cuts both strands of the SP1 oligoduplex within the proto-spacer 4 nt upstream of the PAM (FIG. 5B) leaving blunt ends. It is worth to note, that no cleavage is observed after the 2 h incubation of the SP1 oligoduplex with the Cas9 protein lacking crRNA (FIG. 6C).

To test whether the Cas9-crRNA complex can locate the proto-spacer and cut DNA in vitro in long DNA substrates mimicking in vivo invading foreign DNA we analyzed cleavage of pSP1 plasmid (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) (FIG. 5C) carrying proto-spacer1 and PAM. In the presence of Cas9-crRNA complex supercoiled form of pSP1 plasmid was converted into a linear form (FIG. 5D), while pUC18 plasmid lacking proto-spacer1 was not cleaved. This means that both strands of the pSC1 plasmid were cleaved specifically within the proto-spacer region. We used direct sequencing to determine the ends of linear DNA form formed after the Cas9-crRNA cleavage. Sequencing results confirmed that cleavage of plasmid DNA occurred 4 nt away from PAM sequence similarly to the SP1 oligoduplex cleavage (FIG. 5D). The cleavage positions identified in the in vitro experiments (FIG. 4) for the CRISPR3/Cas system of S. thermophilus are identical to those determined in the in vivo cleavage experiments for the CRISPR1/Cas system in S. thermophilus (Garneau et al., 2010. Nature 468:67-71). To check if Cas9-crRNA induced cleavage occurs at the same position in other proto-spacer sequences, we analysed cleavage of the SP2 oligoduplex carrying a protospacer-2 and PAM sequences by the heterogeneous Cas9-crRNA complex isolated from the host carrying 12 spacer-repeat units. We have found that this heterogeneous Cas9-crRNA complex cuts (+)strand of SP2 oligoduplex exactly at the same position as in the SP1 oligoduplex. Cas9-crRNA cleavage specificity is directed by the crRNA sequence. To demonstrate directly that Cas9-crRNA complex specificity can be re-programmed by changing crRNA in the ribonucleoprotein complex we inserted a new spacer (SN) instead of spacer S1 in the CRISPR region generating pCas(−)SN plasmid containing only a minimal CRISPR region and tracrRNA encoding sequence (FIG. 7), co-expressed this plasmid together with pASKIBA-Cas9 and purified Cas9-crRNA complex. The cleavage specificity of Cas9-crRNA complex was analysed using plasmids pSP1+SPN and pSP1. pSP1+SPN plasmid containing the proto-spacer sequence matching the SN spacer in the CRISPR region, was linearized by the Cas9-crRNA complex, while pSP1 plasmid which lacks complimentary sequence remained intact (FIG. 7B). To determine the cleavage position within the SPN spacer sequence, we performed experiments with SPN oligoduplex, containing proto-spacer complementary to spacer SN and PAM (FIG. 7D). Oligoduplex cleavage assay confirmed (FIGS. 7C and D) that Cas9-crRNA complex with re-engineered specificity cleaves both DNA strands within the SN proto-spacer 4 nt upstream of the PAM identically to other Cas9-crRNA complexes.

The length of the spacer in the CRISPR3 region of S. thermophilus is 30 nt. According to the data provided in the FIG. 1C, the mature crRNA copurified with the Cas9 protein is comprised of 42 nt. It means that only 20 nt of crRNA is complementary to the (+)strand of proto-spacer. To assess whether 5′-end of proto-spacer is important for the plasmid interference by the CRISPR3 system of S. thermophilus we engineered plasmids pSP1-27, pSP1-23, pSP1-19, pSP1-15, pSP1-11 with the 5′-truncated proto-spacer1 (the length of proto-spacer 27 bp, 23 bp, 19 bp, 15 bp, 11 bp, respectively), and analyzed transformation efficiency of the recipient strain containing pCRISPR3 (FIG. 8B). Plasmids containing 4 or 7 bp truncations at the 5′ end of proto-spacer1, had no effect on the recipient strain ability to interfere with plasmid transformation. Shorter versions of proto-spacer (11, 15, 19 bp) abolished recipient strain ability to prevent plasmid transformation. These data shows that 5′ end of the proto-spacer, which has no complementarity to mature crRNA is not important for CRISPR3/Cas function. In full support to the in vivo experiments, the SP1-20 oligoduplex containing only 20 nt of the protospacer-1 is efficiently cleaved by Cas9-crRNA (FIGS. 8 D and E).

PAM is required for DNA binding and cleavage by Cas9-crRNA. Plasmids carrying a proto-spacer but not PAM (pSP1-pΔ) or multiple PAM's but no proto-spacer (pUC18) are resistant for Cas9-crRNA cleavage (FIG. 8A). Hence, in accordance with in vivo data both PAM and proto-spacer are required for double-stranded DNA cleavage by Cas9-crRNA complex (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82). To find out, whether PAM is recognized in a context of a double-stranded or a single-stranded DNA, we analyzed Cas9-crRNA binding and cleavage of oligodeoxynucleotides i) SP1 (containing both proto-spacer and PAM), ii) SP1-Δp (contains only proto-spacer), and iii) SP2 (contains only PAM). The (+)strands of these oligodeoxynucleotides were used as single-stranded DNA substrates (s(+)SP1, s(+)SP1-Δp, s(+)SP2, accordingly) (Table 1).

Consistent with the plasmid cleavage experiments, oligoduplexes which have only proto-spacer, but not PAM are not cut by Cas9-crRNA (FIG. 9B). On the other hand, (+)strand in the single-stranded form is cut at the similar rate independently whether it has or has not PAM (FIG. 9B). These data clearly show that PAM is required only for a double-stranded but not for a single-stranded DNA cleavage.

To test if PAM is important for DNA binding by the Cas9-crRNA complex, electrophoretic mobility shift experiments were performed. To avoid cleavage, binding experiments were performed in the absence of Mg2+ ions which are necessary for cleavage. Cas9-crRNA showed different binding patterns for double-stranded and single-stranded oligonucleotides. In the case of the SP1 oligoduplex a low mobility complex is observed already at 1 nM concentration (FIG. 9C). On the other hand, no binding is observed under the same experimental conditions for oligoduplexes without PAM (SP1-Δp) or without proto-spacer (SP2). Moreover, no low mobility complex is observed in the case of Cas9 protein without crRNA (FIG. 6A), confirming that crRNA is important for complex formation. Thus, taken together binding experiments clearly show that the Cas9 protein complex is unable to bind double-stranded DNA in the absence of PAM, even if it contains crRNA complementary to proto-spacer. To put it into other words, double-stranded DNA substrates lacking PAM are not cleaved because PAM is required for Cas9-crRNA binding.

On the other hand, single-stranded oligonucleotides ((+)strand) are bound by Cas9-crRNA with the same affinity independently of the PAM presence (FIG. 9D). Again, no binding was observed for single-stranded DNA oligonucleotide without proto-spacer (FIG. 9D), or for Cas9 protein lacking crRNA (FIG. 6C). Taken together these data indicate that Cas9-crRNA complex discriminates PAM only in the double-stranded but not a single-stranded DNA.

Since some Type III CRISPR systems provide RNA rather than DNA interference, we have studied RNA binding and cleavage by the Cas9-crRNA complex. The Cas9-crRNA did not cleave specifically either single-stranded RNA, or double-stranded RNA bearing a proto-spacer and PAM (FIG. 10B). This finding confirms confirms once more that DNA is a primary target for the CRISPR3/Cas system of S. thermophilus. Cas9-crRNA complex binds a complementary RNA containing a proto-spacer, but this interaction is probably functionally not important, because single stranded RNA is not cleaved specifically by Cas9 within a proto-spacer.

Mutagenesis of Cas9 protein RuvC and HNH motifs. Plasmid transformation experiments indicate that RuvC and HNH motifs (FIG. 11A) are important for Cas9 function (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82). To test if these motifs are involved in the target DNA cleavage, we expressed and purified D31A and N891A mutants following procedure described for wt Cas9. Both mutants co-purified with crRNA identical to crRNA in the wt Cas9 complex (FIG. 11C). To test whether mutant proteins retained cleavage activity, we monitored pSP1 plasmid cleavage by mutant Cas9-crRNA complexes. Surprisingly, instead of linear reaction product observed for the wt Cas9 protein, both mutants produced nicked DNA form (FIG. 11B) indicating that both active sites mutants cleave only one DNA strand of plasmid substrate within a proto-spacer.

To determine whether mutant proteins exhibit a strand preference, we analysed D31A and N891A mutant cleavage of the SP1 oligoduplex. RuvC active site mutant (D31A) cut (+) strand of oligoduplex at the same position as wt Cas9-crRNA protein. while the (−)strand stayed intact (FIG. 11C). And vice versa, HNH active site mutant (N891A) cleaved only (−)strand, but not (+) strand of the SP1 oligoduplex (FIG. 11D). Taken together these data indicate that RuvC and HNH active sites act on opposite DNA strands to generate a double strand break. To test, whether the same cleavage pattern is conserved during the plasmid DNA cleavage, we sequenced proto-spacer regions of nicked plasmids. Run-off sequence data confirmed that RuvC active site mutant cut only (+) DNA strand while HNH/McrA mutant—only (−)strand (FIGS. 12A and B). Furthermore, we found that RuvC mutant cleaved (+) strand of a single-stranded DNA but no such cleavage was detected for the HNH mutant (FIG. 12D).

To test whether mutations altered DNA-binding affinity of mutant protein-crRNA complexes, DNA binding was studied using the electrophoretic mobility shift assay. Both mutant protein-crRNA complexes bound oligoduplex SP1 with the same affinity as wild type protein (FIG. 12C.). Thus, mutations in the putative active sites of Cas9 have no significant effect on double-stranded DNA-binding properties of the Cas9-crRNA complex. Since 42 nt crRNA was present in the mutant protein complexes (FIG. 12C), we conclude that mutant Cas9-crRNA complexes lost ability to cut one of the target DNA strand due to active site mutation. Since Cas9-HisTag protein is a monomer in solution (FIG. 13), it is likely that Cas9 protein is functional as a monomer and uses two active sites for the cleavage of opposite DNA strands. Similar strategy is exploited by some restriction endonucleases (Armalyte et al., 2005. J Biol Chem 280: 41584-94).

Discussion

Cas9-crRNA complex of CRISPR3/Cas system of S. thermophilus is crRNA-guided endonuclease. This work demonstrates that Cas9-crRNA complex of CRISPR3/Cas system of S. thermophilus is crRNA-directed endonuclease which cuts both DNA strands in the presence of Mg2+-ions within a protospacer 4 nt downstream of the PAM sequence to produce blunt end cleavage products. Sequence specificity of the Cas9-crRNA complex is dictated by the 42 nt crRNA which include ˜20 nt fragment complementary to the proto-spacer sequence in the target DNA. In this respect the mature crRNA in the Cas9 complex of CRISPR3/Cas system of S. thermophilus is similar to crRNA of Streptococcus pyogenes which has a 3-handle of repeat sequence but lacks part of the spacer sequence and 5′-handle corresponding to the repeat fragment (Deltcheva et al, 2011). Therefore, crRNA present in the Cas9-crRNA complex of CRISPR3/Cas system of S. thermophilus is complementary only to the part of the proto-spacer sequence distal to PAM. Not surprisingly, truncation of the 3-end of the proto-spacer sequence by 10 nucleotides has no effect on Cas9-crRNA cleavage of synthetic oligoduplexes or plasmid DNA (FIG. 8).

The cleavage machinery of Cas9-crRNA complex resides in the Cas9 protein which provides two active sites for the phosphodiester bond cleavage. The RuvC- and HNH-like active sites of Cas9 protein are located on different domains and act independently on individual DNA strands. Alanine replacement of the active site residues in the RuvC- and HNH-motifs transforms Cas9-crRNA complex into a strand-specific nicking endonucleases similar to the nicking enzymes (Chan et al., 2011. Nucleic Acids Res 39:1-18). Consistent with in vivo studies. a functional activity of the Cas9-crRNA complex in vitro is absolutely dependent on the presence of the proto-spacer adjacent motif NGGNG upstream of the proto-spacer sequence. Data presented in the FIG. 3 show that PAM is required for Cas9-crRNA binding to the double-stranded DNA. If PAM sequence is missing in double-stranded DNA, the Cas9-crRNA complex does not bind such DNA even if it contains a complementary proto-spacer sequence. On the other hand, Cas9-crRNA does not display DNA binding if PAM (or multiple PAM's) is present but proto-spacer sequence is absent. Thus, in consistence with the in vivo data, both PAM and proto-spacer sequences are necessary prerequisite for double-stranded DNA binding and subsequent cleavage. Contrary to the Cas9-crRNA binding to the double-stranded DNA, PAM sequence motif has no effect on the single-stranded DNA binding by: a single-stranded oligodeoxynucleotide containing proto-spacer with or without PAM sequence is bound equally well but with lower affinity than double-stranded DNA. In the presence of Mg2+ ions Cas9 cuts single-stranded DNA bound to the crRNA using its HNH-active site.

Mechanism of DNA interference in the Type II systems. Our results establish a simple model for the mechanism of double-stranded DNA cleavage by Cas9-crRNA complex in the S. thermophilus CRISPR3/Cas system (FIG. 14). Cas9-crRNA complexes using a mechanism that yet has to be defined locates and binds to a proto-spacer sequence within the double-stranded DNA in a PAM-dependent process. It is possible that PAM in the double-stranded DNA serves as an initiation site (signal) for the strand separation and promotes subsequent pairing of crRNA to the complementary (+)strand of DNA. It remains to be established whether a Cas9 protein module or Cas9-bound crRNA (for example, using nucleotides in the conserved the “3′-handle” of the conserved repeat sequence) recognizes the PAM sequence. Despite of the lack of these mechanistic details, our data clearly demonstrate that PAM is recognized by Cas9-crRNA in the context of double-stranded DNA. The Cas9-crRNA binding to the target sequence in the ds DNA presumably results in the R-loop structure where (−)strand is displaced and the complementary (+) DNA strand is paired to the crRNA. In the presence of Mg2+ ions phosphodiester bond cleavage occurs on both strands 4 nt 5′-upstream of the PAM sequence to generate blunt DNA ends. DNA cleavage analysis by the RuvC- or HNH-motif mutants demonstrate that RuvC- and HNH-like active sites of Cas9 protein act on the (−) and (+)strands, respectively. Therefore, in the catalytically competent the Cas9-crRNA complex, the N-terminal domain containing the catalytic D31A residue of the RuvC motif is positioned at the displaced (−) DNA strand. while the central part of Cas9 containing the HNH motif is located in the vicinity of the scissile phosphodiester bond of (+) DNA strand paired to crRNA. After DNA cleavage Cas9-crRNA remains bound to the reaction products (FIG. 15). Taken together data presented here suggest a first molecular mechanism for the DNA interference step by the CRISPR3/Cas system of S. thermophilus. Since cas9 is a signature gene (Makarova et al., 2011. Nat Rev Microbiol 9:467-77) for Type IIA and Type IIB systems the cleavage mechanism proposed here is likely to be conserved in other Type IIA and Type IIB systems. Stand-alone versions of Cas9-like proteins which are not a part of the CRISPR system were identified by bioinformatics (Makarova et al., 2011. Biol Direct 6: 38). In the light of the data provided here we suggest that these proteins can provide interference against foreign DNA similarly to Cas9 if loaded with small crRNA molecules which may be generated through the pathway different from CRISPR.

Comparison to other RNA interference complexes. The mechanism proposed here for the double-stranded DNA cleavage by the Cas9-crRNA complex differs significantly from that for the Type I-E (former E. coli or CASS2) system (Jore et al., 2011. Nat Struct Mol Biol 18:529-36). In the E. coli system crRNA and Cas proteins assemble into a large ribonucleoprotein complex named Cascade that facilitates target recognition by enhancing sequence-specific hybridization between the CRISPR RNA and complementary target sequences (Jore et al., 2011. Nat Struct Mol Biol 18:529-36). Target recognition is dependent on PAM and governed by the “seed” crRNA sequence located at the 5′-end of the spacer region (Semenova et al., 2011. Proc Natl Acad Sci USA 108:10098-103). However, while Cascade-crRNA complex alone is able to bind double-stranded DNA containing PAM and proto-spacer, it requires an accessory Cas3 protein for DNA cleavage. Cas3 is a single-stranded DNA nuclease and helicase which is able to cleave single-stranded DNA producing multiple cuts (Sinkunas et al., 2011. EMBO J 30:1335-42). The mechanistic details of the Cas3 action on a proper biological substrate (e.g., Cascade-crRNA bound to the double-stranded DNA in the R-loop like complex) have yet to be established. However, it has been demonstrated recently that Cas3 of M. jannaschii alone is able to cut both DNA strands in the synthetic substrate mimicking R-loop (Beloglazova et al., 2011. EMBO J 30:616-27). It is proposed that Cas3 may follow similar mechanism for DNA cleavage in the presence of Cascade-crRNA complex. Thus, current data clearly show that mechanistic details of the interference step for the Type I-E system differs from that of CRISPR3 system both by the catalytic machinery and mechanism and complexity.

In the III-B subtype CRISPR systems present in many archea and some bacteria, Cas module RAMP (Cmr) proteins and cRNA assemble into the effector complex that targets invading RNA (Hale et al., 2009. Cell 139:945-56; Hale et al., 2012. Mol Cell 45:292-302). In Pyrococcus furiosus RNA silencing complex comprised of six Cmr1-6 proteins and crRNA binds to the target RNA and cuts it at fixed distance in respect to 3′-end the psiRNA. The cleavage activity depends on Mg2+-ions however individual Cmr protein(-s) responsible for target RNA cleavage has yet to be identified. The effector complex of Sulfolobus solfataricus comprised of seven Cmr1-7 proteins and crRNA cuts invading RNA in an endonucleolytic reaction at UA dinucleotides (Zhang et al., 2012. Mol Cell 45: 303-13). Importantly, both Cmr-crRNA complexes perform RNA cleavage in a PAM independent manner.

The data provided here show that Cas9-crRNA complex of CRISPR3 system is so far the most simple DNA interference system comprised of a single Cas9 protein bound to the crRNA molecule. The simple modular organization of the Cas9-crRNA complex where specificity for DNA target is encoded by the crRNA and cleavage machinery is brought by the Cas protein provides a versatile platform for engineering of universal RNA-guided DNA endonucleases.

Example 2

In Vitro Assembly of Cas9-crRNA Complex from 4 Components

In this example we demonstrate that the catalytically active Cas9-crRNA complex can be assembled in vitro by mixing 4 individual components: the C-terminal (His)6-tagged variant of Cas9 protein (“(His)6” disclosed as SEQ ID NO: 23), tracrRNA transcript (SEQ ID NO: 5), CRISPR RNA transcript (SEQ ID NO: 8) and E. coli RNAseIII (Abgene). Cas9 protein is first pre-incubated with tracrRNA and CRISPR RNA transcripts, followed by the subsequent incubation with RNAselII to generate a catalytically competent Cas9-crRNA complex which is used for the site-specific DNA cleavage.

More specifically, RNA fragments required for complex assembly were produced by in vitro transcription (TranscriptAid™ T7 High Yield Transcription Kit, Fermentas) of PCR-generated fragment containing a T7 promoter at the proximal end of RNA coding sequence. PCR-generated DNA fragments encoding CRISPR RNA and tracrRNA were produced using pCas9(−)SP1 plasmid as a template with a following primer pair: 5′-taatacgactcactataGggtagaaaagatatcctacgagg-3′ (SEQ ID NO: 40)/5′-CAACAACCAAGCTAATACAGCAG-3′ (SEQ ID NO: 41) and 5′-aaaaacaccgaatcggtgccac-3′ (SEQ ID NO: 42)/5′-taatacgactcactataGggTAATAATAATTGTGGTTTGAAACCATTC-3′ (SEQ ID NO: 43) (T7 RNA polymerase promoter underlined, transcription start shown in bold). The 150 nt CRISPR RNA transcript is comprised of 102 nt Repeat-Spacer1-Repeat sequences flanked by the 23 nt upstream and 25 nt downstream regions required for primer annealing. The 105 nt transcript of tracrRNA is comprised of a 38 nt stretch partially complimentary to the S. thermophilus DCGG7710 CRISPR3 repeat sequence fragment (anti-repeat sequence), flanked by the 16 nt upstream and 51 nt downstream region. RNA fragments produced by in vitro transcription were purified using RNeasy MinElute Cleanup Kit (Qiagen).

For in vitro assembly of catalytically competent Cas9-crRNA complex, the (His)6-tagged Cas9 protein (“(His)6” disclosed as SEQ ID NO: 23) was mixed with CRISPR RNA and tracrRNA transcripts at 1:0.5:1 molar ratio and pre-incubated in a buffer containing 10 mM Tris-HCl (pH 7.5 at 37° C.), 100 mM NaCl at 37° C. for 30 min followed by addition of RNAselII (Ambion), MgCl2 and DTT and subsequent incubation for additional 30 min. The final concentrations of the components in the assembly mix were the following: 100 nM of (His)6-tagged Cas9 protein (“(His)6” disclosed as SEQ ID NO: 23), 50 nM of CRISPR RNA, 100 nM of tracrRNA, 50 nM RNAselII, 10 mM MgCl2 and 1 mM DTT.

Below we provide experimental evidences that in vitro assembled Cas9-crRNA complex guided by the crRNA sequence cleaves DNA at the specific site to generate blunt ends. In this respect Cas9-crRNA complex can be used an alternative for a restriction endonuclease or meganuclease for the site-specific DNA cleavage in vitro. The sequence specificity of the complex is dictated by the crRNA sequence which can be engineered to address a desirable DNA target.

First, the DNA cleavage activity of the in vitro assembled Cas9-crRNA complex was assayed on the plasmid substrates pSP1 and pUC18. The pSP1 plasmid contained a proto-spacer1 sequence flanked by the 5′-GGNG-3′PAM sequence. Proto-spacer1 sequence was not present in pUC18. Reactions on pUC18 and pSP1 plasmids (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) were conducted at 37° C. in the 10 mM Tris HCl (pH 7.5 at 37° C.), 50 mM NaCl, 0.05 mg/ml BSA, 0.5 mM DTT and 10 mM MgCl2. Reaction mixtures typically contained 3.0 nM of supercoiled plasmid DNA. The reactions were initiated by mixing 50 μl volumes of Cas9-crRNA complex and plasmid DNA (1:1 v/v ratio) in a reaction buffer. Aliquots were removed at timed intervals and quenched with phenol/chloroform. The aqueous phase was mixed with loading dye solution (0.01% bromphenol blue and 75 mM EDTA in 50% v/v glycerol) and reaction products analyzed by electrophoresis through agarose (FIG. 16). To check whether the pSP1 plasmid pre-cleaved by Cas9-crRNA complex can be re-ligated, we purified linear pSP1 cleavage product from agarose gel using GeneJET gel extraction Kit (Fermentas) and re-ligated using T4 DNA ligase (Fermentas). After transformation of E. coli cells by the ligation mix, five individual clones were selected from resulting transformants, plasmid DNA was purified and subjected to sequencing using the following primers: 5′-ccgcatcaggcgccattcgcc-3′ (SEQ ID NO: 29) (sequencing of (+)strand) and 5′-gcgaggaagcggaagagcgccc-3′ (SEQ ID NO: 30) (sequencing of (−)strand). Sequence analysis revealed that the DNA sequence of the pSP1 plasmid in the locus that was cleaved by Cas9-crRNA complex and re-ligated was identical to the sequence of the non-treated plasmid. E. coli transformation by the ligation mix in the absence of T4 DNA ligase did not produce transformants indicating that no traces of supercoiled plasmid are co-purified with the linear reaction product.

Next, the cleavage activity of the in vitro assembled Cas9-crRNA complex was assayed on a synthetic 55 bp oligodeoxynucleotide duplex SP1 containing a proto-spacer sequence matching to the spacer sequence of crRNA (FIG. 17). Reactions conditions were identical to those described above for the plasmid DNA cleavage, except that 1 nM of oligoduplex was used. Reaction product analysis revealed that in vitro assembled Cas9-crRNA complex cleaved both strands of the oligoduplex at fixed position, inside the proto-spacer, after the 37th nucleotide from the 5′-terminus, 4 nt upstream of the PAM sequence 5′-GGNG-3′ leaving blunt ends (FIG. 17).

Example 3

In Vitro Assembly of Cas9-crRNA Complex from 3 Components

In this example we demonstrate that active Cas9-crRNA complex can be assembled in vitro by mixing 3 individual components: the C-terminal (His)6-tagged variant of Cas9 protein (“(His)6” disclosed as SEQ ID NO: 23), tracrRNA transcript provided in Example 1 (SEQ ID NO: 5 and SEQ ID NO: 6), and CRISPR RNA transcript (SEQ ID NO: 8) provided in Example 1 or synthetic crRNA (SEQ ID NO: 8) which corresponds to the putative crRNA of CRISPR3/Cas system of S. thermophilus DGCC7710 strain. Synthetic 42 nt oligoribonucleotide is comprised of 20 nt of identical to the spacer1 of CRISPR3 region at the 5′ terminus and 22 nt of repeat sequence at the 3′ end. More specifically, tracrRNA and CRISPR RNA transcripts were obtained as described in Example 1. To generate the Cas9-crRNA complex the (His)6-tagged Cas9 protein (“(His)6” disclosed as SEQ ID NO: 23) was mixed with tracrRNA and CRISPR RNA transcript, or 42 nt synthetic crRNA, at 1:0.5:1 molar ratio and incubated in a buffer containing 10 mM Tris-HCl (pH 7.5 at 37° C.). 100 mM NaCl at 37° C. for 1 h. The final concentrations of the components in the assembly mix were the following: 100 nM of (His)6-tagged Cas9 protein (“(His)6” disclosed as SEQ ID NO: 23), 50 nM of CRISPR RNA or 42 nt synthetic crRNA, 100 nM of tracrRNA.

Below we provide experimental evidences that in vitro assembled Cas9-crRNA complex guided by the crRNA sequence cleaves DNA at the specific site to generate blunt ends. In this respect Cas9-crRNA complex can be used an alternative for a restriction endonuclease or meganuclease for the site-specific DNA cleavage in vitro. The sequence specificity of the complex is dictated by the crRNA sequence which can be engineered to address a desirable DNA target.

First, the DNA cleavage activity of the in vitro assembled Cas9-crRNA complex was assayed on the plasmid substrates pSP1 and pUC18. The pSP1 plasmid contained a proto-spacer1 sequence flanked by the 5′-GGNG-3′PAM sequence. Proto-spacer1 sequence was not present in pUC18. Reactions on plasmid substrates (Sapranauskas et al., 2011. Nucleic Acids Res 39:9275-82) were conducted at 37° C. in the 10 mM Tris-HCl (pH 7.5 at 37° C.), 50 mM NaCl, 0.05 mg/ml BSA, 0.5 mM of DTT and 10 mM MgCl2. Reaction mixtures typically contained 3.0 nM of supercoiled plasmid DNA. The reactions were initiated by mixing 50 μl volumes of Cas9-crRNA complex and plasmid DNA (1:1 v/v ratio) in a reaction buffer. Aliquots were removed at timed intervals and quenched with phenol/chloroform. The aqueous phase was mixed with loading dye solution (0.01% bromphenol blue and 75 mM EDTA in 50% v/v glycerol) and reaction products analyzed by electrophoresis through agarose (FIG. 18).

Next, the cleavage activity of the in vitro assembled Cas9-crRNA complex was assayed on a synthetic 55 bp oligodeoxynucleotide duplex SP1 containing a a proto-spacer sequence matching to the spacer sequence of crRNA (FIG. 19). Reactions conditions were identical to those described above for the plasmid DNA cleavage, except that 1 nM of oligoduplex was used. Reaction product analysis revealed that in vitro assembled Cas9-crRNA complex cleaved both strands of the oligoduplex at fixed position, inside the proto-spacer, after the 37th nucleotide form the 5′-end, 4 nt upstream of the PAM sequence 5′-GGNG-3′ leaving blunt ends (FIG. 19).

Example 4

Interchangeable Spacer Cassette for the Re-Programing of the Cas9-crRNA Complex Specificity.

In this example we describe an interchangeable spacer cassette which allows to produce crRNA carrying a nucleotide sequence against any desirable DNA target to be used for assembly of the Cas9-crRNA complex described in Examples 1 and 2 (FIG. 20B). The cassette caries a single repeat-spacer-repeat unit which allows insertion of the oligoduplex carrying the new spacer sequence required to generate a desired crRNA. To engineer a cassette, first we constructed a cassette containing a leader sequence, a repeat sequence and a unique SapI recognition site in the vicinity of the repeat sequence followed by BamHI site (FIG. 20C). To generate CRISPR region containing the unique desired spacer, we inserted a synthetic oligoduplex containing a unique spacer sequence and a repeat unit into the plasmid precleaved with SapI and BamHI restriction enzymes. Using this cassette we produced crRNA transcripts which contained nucleotide sequences complementary to the proto-spacers N1 and N2 present in pUC18 plasmid (see below).

As proof of the principle demonstration, we used an interchangeable spacer cassette to generate crRNA1 and crRNA2 which were engineered to target pUC18 plasmid at proto-spacer1 and proto-spacer2, respectively, incorporated crRNA1 and crRNA2 into Cas9 complex as described in the Example 1 and used these complexes for the cleavage of pUC18 plasmid. The proto-spacer N1 is located near the SapI restriction endonuclease site, while the proto-spacer N2 is in the vicinity of AatII site. The distance between SapI and AatII restriction sites is 775 bp, while the distance between the putative Cas9-crRNA complex cleavage sites located in the spacers N1 and N2 is 612 bp (FIG. 21A). The crRNA1 and crRNA2 PCR fragments containing T7 promoter at the proximal end were obtained from the corresponding interchangeable spacer cassette plasmids and used to produce by in vitro transcription CRISPR RNA transcripts carrying sequences matching spacer N1 or spacer N2 sequences. The catalytically active complexes of Cas9 with crRNA1 and crRNA2 were assembled for DNA cleavage as described in Example 1. In vitro assembled complexes containing either crRNA1 or crRNA2 linearized pUC18 plasmid (FIG. 21B). When both complexes were incubated with the pUC18plasmid, two DNA fragments (2074 and 612 bp) were obtained (FIG. 21B), indicating that plasmid cleavage occurred at sites targeted by the crRNA molecules present in the complexes.

Example 5

Cloning Procedure Using Cas9-crRNA Complex.

In this example we demonstrate that Cas9-crRNA complex may be used to prepare a vector for cloning procedure. First we demonstrated that cleavage products obtained by the Cas9-crRNA complex can be re-ligated by DNA ligase. We purified linear pSP1 cleavage product from agarose gel and re-ligated it using DNA ligase. After transformation of E. coli cells by the ligation mix, five individual clones were selected from resulting transformants, plasmid DNA was purified and subjected to sequencing. Sequence analysis revealed that the DNA sequence of the pSP1 plasmid in the locus that was cleaved by Cas9-RNA complex and re-ligated was identical to the sequence of the non-treated plasmid. E. coli transformation by the ligation mix in the absence of T4 DNA ligase did not produce transformants indicating that no traces of supercoiled plasmid are co-purified with the linear reaction product. This result illustrates, that the DNA ends generated by the Cas9 cleavage are substrates for T4 DNA ligase, and therefore must contain a phosphate at the 5′ terminus and a free OH group at the 3′ terminus (Lehman, 1974).

Next we analyzed cleavage of pUC18 plasmid with Cas9 complex loaded with crRNA1 and crRNA2 described in Example 5 (FIG. 21A). First, pUC18 was cleaved with one complex, purified and re-ligated. Sequencing of 10 clones in each case confirmed, that sequence of cleaved and re-ligated plasmid was identical to the sequence of the non-treated plasmid (FIG. 21C). This experiment suggests that additional mutations are not introduced after cleavage by Cas9-crRNA complex and ligation, and the Cas9-crRNA complex can be used for cloning experiments. When both complexes were incubated with the pUC18 plasmid, two DNA fragments (2074 and 612 bp) were obtained (FIG. 21B), indicating that plasmid cleavage occurred at sites targeted by the crRNA molecules present in the complexes. To demonstrate that the pUC18 plasmid cleaved with Cas9-RNA complexes is suitable for a genetic engineering we cloned PCR fragment containing a promoter and a tetracycline resistance gene from the pACYC184 plasmid to the pUC18 vector pre-cleaved with the Cas9 complex mix containing both crRNA1 or crRNA2. The clones were selected on the media enriched by tetracycline and ampicillin. Sequencing of 4 selected clones confirmed that the intact PCR fragment was inserted into a desired position ((FIG. 21C).

More specifically, the 2 μg pUC18 was incubated with the mix of separately assembled Cas9-RNA complexes (250 nM each) containing different crRNAs for 1 hour at 37° C. in 100 μl reaction volume (10 mM Tris-HCl (pH 7.5 at 37° C.), 100 mM NaCl, 1 mM DTT and 10 mM MgCl2). Obtained vector fragment was purified from agarose gel using GeneJET gel extraction Kit (Thermo Fisher scientific) and divided in to two equal parts. One part of pre-cleaved vector was dephosphorylated with the FastAP alkaline phosphatase while another part was untreated. 1282 bp insert containing a promoter and a tetracycline resistance gene was obtained from the pACYC184 plasmid by PCR. After purification using the GeneJET PCR Purification Kit (Thermo Fisher scientific), a solution containing the PCR fragment was divided in to two parts. One part was phosphorylated with T4 polynucleotide kinase (Thermo Fisher scientific) while another part remained untreated. Untreated vector was ligated with the untreated PCR fragment, while a dephosphorylated vector was ligated with a phosphorylated fragment using the T4 DNA ligase (Thermo Fisher scientific). Clones were selected on a media supplemented with 100 μg/ml of Ap and 25 μg/ml Tc.

Example 6

Cleavage of Long DNA Substrates by Cas9 crRNA Complex.

In this example we demonstrate that Cas9-crRNA may be addressed to cleave targets in long DNA molecules, including phage Δ, E. coli and human genomic DNAs.

More specifically, we addressed Cas9-RNA complex to cleave specific sites in A bacteriophage (48 kb), E. coli BL-21 strain (4.6 Mb) and human (3.2 Gb) genomic DNAs. Cas9-crRNA complex was assembled as described in Examples 2 and 3. We used 42 nt long synthetic crRNAs, 150 nt pre-crRNAs and tracrRNAs synthesized using in vitro transcription from templates generated as described in Example 4.

A DNA cleavage reactions were initiated by mixing A DNA (Thermo Fisher Scientific) with assembled Cas9-RNA complex (1:1 v/v ratio) and incubating at 37° C. Final reaction mixture contained 2 μg A DNA, 50 nM Cas9-RNA complex, 10 mM Tris-HCl (pH 7.5 at 37° C.), 100 mM NaCl, 1 mM DTT and 10 mM MgCl2 in 100 μl reaction volume. Aliquots were removed at timed intervals and quenched with phenol/chloroform. The aqueous phase was mixed with 3× loading dye solution (0.01% bromphenol blue and 75 mM EDTA in 50% v/v glycerol) and reaction products analyzed by electrophoresis through agarose gels and ethidium bromide staining. The analysis of linear A phage genomic DNA cleavage products in agarose gel confirmed that ˜40 bp length DNA is efficiently cleaved at a single site (FIG. 22A).

DNA from E. coli BL21 (DE3) strain was isolated using the Genomic DNA purification kit (Thermo Fisher Scientific). For cleavage assay, E. coli genomic DNA was combined with assembled Cas9-RNA complex (1:1 v/v ratio) and incubated for 3 hours at 37° C. Final reaction mixture contained 30 μg genomic DNA, 1 μM Cas9-RNA complex, 10 mM Tris-HCl (pH 7.5 at 37° C.), 100 mM NaCl, 1 mM DTT and 10 mM MgCl2 in 300 μl reaction volume. Following incubation, 30 μl of FastDigest PstI (Thermo Fisher Scientific) was added and the reaction mix was incubated for additional 16 hours at 37° C. The reaction was terminated by heating the reaction mixture for 30 min at 55° C. with Proteinase K (0.5 mg/ml; Thermo Fisher Scientific) and SDS (0.5%, w/v) followed by 30 min incubation at room temperature with RNase A (0.25 mg/ml; Thermo Fisher Scientific). After phenol/chloroform extraction, DNA was precipitated by isopropanol and dissolved in TE buffer (10 mM Tris-HCl, pH 8.0 and 1 mM EDTA). 10 μg of DNA was mixed with 3× loading dye solution (0.01% bromphenol blue and 75 mM EDTA in 50% v/v glycerol) and electrophoresed on 1% agarose gel.

To analyse Cas9-crRNA cleavage products of E. coli genomic DNA, we designed a probe against DNA fragment containing a Cas9-RNA complex target (a proto-spacer) (FIG. 22B) and performed Southern blot analysis. Southern blot analysis was performed as described in (Sambrook et al, 1989. Molecular Cloning: A Laboratory Manual) with the following modifications. Fractionated DNA was transferred from agarose gel onto SensiBlot Plus Nylon membrane (Thermo Fisher Scientific) via semi-dry transfer. DNA was denatured and fixed on the membrane by placing it on paper towel saturated with 0.4 M NaOH for 10 min, rinsed with 2×SSC and air dried. The membrane was prehybridized with 6×SSC buffer containing 0.5% SDS and 100 μg/ml denatured salmon sperm DNA (Amresco) for 1 h at 65° C. The hybridization probe was generated by PCR using the genomic E. coli BL21(DE3) DNA as a template yielding 397 bp product. 5′-ends were dephosphorylated with FastAP phosphatase (Thermo Fisher Scientific) and radiolabelled by incubating with [γ-2P]ATP (Hartmann Analytic) and T4 PNK (Thermo Fisher Scientific). The labeled probe was purified using GeneJET PCR Purification Kit (Thermo Fisher Scientific), denatured by heating to 95° C. for 5 min, rapidly cooled on ice and added directly to the prehybridization solution. The membrane was probed for 16 hours at 65° C. and washed twice with 2×SSC, 0.5% SDS and twice with 2×SSC, 0.1% SDS at room temperature, air dried and visualized by phosphorimaging (FLA-5100; Fujifilm).

The probe was designed to target DNA fragment containing a target (a proto-spacer) for the Cas9-RNA complex (FIG. 22B). The distance between two PstI targets is ˜1500 bp, while the distance between proto-spacer and left PstI target is 466 bp. After cleavage with Cas9 complex we detected only 466 bp DNA fragment (FIG. 22C), which means that all DNA targets were cleaved by Cas9 protein in the desired position. These data clearly demonstrates that Cas9 protein effectively finds targets in very long and complex molecules such as viral and bacterial DNA.

To analyze Cas9-crRNA cleavage products of human genomic DNA we used DNA extracted from human brain. Human genomic DNA was combined with assembled Cas9-crRNA complex (1:1 v/v ratio) and incubated for 30 min at 37° C. Final reaction mixture contained 1 μg genomic DNA, 100 nM Cas9, 10 mM Tris-HCl (pH 7.5 at 37° C.), 100 mM NaCl. 1 mM DTT and 10 mM MgCl2 in 100 μl reaction volume. Cas9-crRNA-HS1 (SeqID #13) and Cas9-crRNA-HS2 (SeqID #14) complexes were assembled to target RASGEF1C or ARL15 loci, respectively. Cleavage products were analyzed using qPCR (FIG. 22D). After treatment with Cas9-crRNA complex, the amount of intact DNA targets decreased more than 25 times. The analysis of the results obtained from qPCR data revealed that Cas9-RNA complexes cleave human genomic DNA efficiently in the desired loci. These data clearly demonstrates that Cas9 protein effectively finds targets in very long and complex molecules such as viral, bacterial and mammal DNA.

Example 7

Evidence for Gene Editing of a Reporter Plasmid in Mammalian Cells after Transfection of Cas9/RNA Complexes.

A reporter plasmid was constructed to monitor double-strand break repair either through non-homologous end-joining (NHEJ) or homologous recombination (HR). The plasmid contained GFP with an intron and flanking the eGFP sequences are 5′ and 3′ sequences of RFP as well as sites of homology (FIG. 23). The reduction of eGFP fluorescence using this reporter plasmid was an indication of NHEJ in which a Cas9/RNA-mediated double-strand break at targets C or D was repaired imperfectly by NHEJ, thereby disrupting the eGFP coding sequence. Targeting of intronic targets A and B and repair by NHEJ would likely not result in a reduction in eGFP fluorescence because the mutations induced by NHEJ usually delete or insert <20 bps and would therefore not affect the eGFP coding regions or splice site junctions. The appearance of RFP fluorescence, on the other hand, was an indication of HR where the Cas9/RNA-mediated double strand break is repaired by HR using the homologous sequences of RFP indicated.

The crRNA targeting used 42 nucleotide RNA molecules, as described above, having 22 nucleotides that are the repeat sequence, and 20 nucleotides (spacer sequence) are for the specific target. As described above, the target DNA needs the S. thermophilus motif or PAM which is “NGGNG” downstream of the protospacer in the target. GFP was not “engineered” to contain this PAM motif; several target sequences within eGFP naturally occur with the PAM sequence and crRNAs were designed to target the adjacent spacer sequences. RFP was a marker for homologous recombination after a double strand break in eGFP was created by Cas9/RNA.

FIG. 28A shows reporter gene construct for Cas9 protein activity analysis in eukaryotic cells in vivo. Intron sequence contains three cas9 target sites (A, E, B); GFP gene contains two (C, D) cas9 target sites. The RFP gene is split at Y196 position, where RFP fluorescence is abolished. FIG. 28B shows that GFP fluorescence is observed following intron processing in vivo. FIG. 28C shows that the Cas9/crRNA complex facilitated dsDNA breaks in any of aforementioned nuclease target sites may induce HR, result in reassembly of RFP gene and appearance of RFP fluorescence. FIGS. 28D and E show that the Cas9/crRNA complex facilitated dsDNA breaks in any of aforementioned nuclease target sites may induce NHEJ. Mutations in GFP gene sequence would result in lost or diminished GFP fluorescence; mutations in intron may have no affect on GFP fluorescence, however, in distinct cases may yield mature messenger RNA with improperly spliced intron sequences and result in lost or diminished GFP fluorescence.

S. thermophilus Cas9 protein, purified from E. coli, was complexed with in vitro-transcribed tracrRNA and synthetic unmodified crRNA targeting either sequence A (intronic) or sequence C (coding) of eGFP. For transfection, the Cas9/RNA complexes (either targeting A or C) were incubated with the transfection reagent TurboFECT and the reporter plasmid DNA was also incubated with TurboFECT in separate tubes and they were both added to CHO-K1 cells. The percentage of eGFP-positive cells was determined by flow cytometry. As shown in FIGS. 24 and 29, when cells were transfected with the reporter plasmid alone or with the reporter plasmid with Cas9 protein alone, the percentage of GFP-positive cells was about 40-50%, indicative of the overall transfection efficiency. However, when Cas9/RNA complexes targeting sequence C of eGFP were added to cells along with the reporter plasmid, the percentage of eGFP-positive cells was reduced to about 15%. This decrease in eGFP-positive cells was seen only with Cas9/RNA complexes targeting sequence C and there was no significant decrease in eGFP-positive cells seen with the Cas9/RNA complexes targeting sequence A or with a non-specific RNA. This result indicated that the Cas9/RNA targeting sequence C of eGFP resulted in gene editing of eGFP by introduction of a double-strand break and imperfect correction by NHEJ, creating a deletion in the coding sequence of eGFP.

In addition to analyzing the percentage of eGFP-positive cells, transfected cells were also visualized by fluorescent microscopy to monitor the appearance of RFP-positive cells, an indication of repair of Cas9-mediated double strand break by HR rather than NHEJ. As seen in FIG. 25, RFP is seen in some cells after transfection with the reporter plasmid and Cas9/RNA complexes targeting eGFP sequence C, suggesting double-strand break repair by HR.

Example 8

Cas9/RNA Complexes Made Using Synthetic Unmodified tracrRNAs and crRNAs are Functional In Vitro.

The experiments described in Example 7 above used Cas9/RNA complexes comprised of purified Cas9, synthetic crRNAs, and in vitro-transcribed tracrRNA. To determine whether Cas9/RNA complexes were functional when made using fully synthetic RNA components (crRNA and tracrRNA), unmodified S. thermophilus tracrRNAs (both endogenous 89-mer and a shorter 74-mer version that is expected to maintain functionality) were synthesized. The unmodified synthetic crRNAs were generated against target E (see FIGS. 26 and 30) located within the intron of eGFP in the reporter plasmid described above and Cas9/RNA (crRNA and tracrRNA) complexes were generated. To test these complexes, the reporter plasmid used above was incubated with the complexes in vitro and monitored for restriction by gel electrophoresis.

As seen in FIG. 27, Cas9/RNA complexes comprised of fully synthetic RNAs were equally functional in the in vitro assay as Cas9/RNA complexes comprised of synthetic crRNA and in vitro-transcribed tracrRNA.

Sequences SEQ ID NO: 1 WT Cas9 S. thermophilus DGCC7710 CRISPR3-Cas strain One letter: mlfnkciiisinldfsnkekcmtkpysigldigtnsvgwavitdnykvpskkmkvlgntskkyikknllgvllfdsgitaegrrlkrtarrrytrrr nrilylqeifstematlddaffqrlddsflvpddkrdskypifgnlveekvyhdefptiyhlrkyladstkkadlrlvylalahmikyrghfliegef nsknndiqknfqdfldtynaifesdlslenskqleeivkdkisklekkdrilklfpgeknsgifseflklivgnqadfrkcfnldekaslhfskesy dedletllgyigddysdvflkakklydaillsgfltvtdneteaplssamikrynehkedlallkeyirnislktynevfkddtkngyagyidgktn qedfyvylknllaefegadyflekidredflrkqrtfdngsipyqihlqemraildkqakfypflaknkeriekiltfripyyvgplargnsdfaws irkrnekitpwnfedvidkessaeafinrmtsfdlylpeekvlpkhsllyetfnvyneltkvrfiaesmrdyqfldskqkkdivrlyfkdkrkvtd kdiieylhaiygydgielkgiekqfnsslstyhdllniindkeflddssneaiieeiihtltifedremikqrlskfenifdksvlkklsrrhytgwgkl saklingirdeksgntildyliddgisnrnfmqlihddalsfkkkiqkaqiigdedkgnikevvkslpgspaikkgilqsikivdelvkvmggrk pesivvemarenqytnqgksnsqqrlkrlekslkelgskilkenipaklskidnnalqndrlylyylqngkdmytgddldidrlsnydidhiip qaflkdnsidnkvlvssasnrgksddfpslevvkkrktfwyqllksklisqrkfdnltkaerggllpedkagfiqrqlvetrqitkhvarlldekfn nkkdennravrtvkiitlkstlvsqfrkdfelykvreindfhhahdaylnaviasallkkypklepefvygdypkynsfrerksatekvyfysni mnifkksisladgrvierplievneetgesvwnkesdlatvrrvlsypqvnvvkkveeqnhgldrgkpkglfnanlsskpkpnsnenlvgak eyldpkkyggyagisnsfavlvkgtiekgakkkitnvlefqgisildrinyrkdklnfIlekgykdieliielpkyslfelsdgsrrmlasilstnnkr geihkgnqiflsqkfvkllyhakrisntinenhrkyvenhkkefeelfyyilefnenyvgakkngkllnsafqswqnhsidelcssfigptgser kglfeltsrgsaadfeflgvkipryrdytpssllkdatlihqsvtglyetridlaklgeg Three letters: MetLeuPheAsnLysCysIleIleIleSerIleAsnLeuAspPheSerAsnLysGluLys ValIleThrAspAsnTyrLysValProSerLysLysMetLysValLeuGlyAsnThrSer LysLysTyrIleLysLysAsnLeuLeuGlyValLeuLeuPheAspSerGlyIleThrAla GluGlyArgArgLeuLysArgThrAlaArgArgArgTyrThrArgArgArgAsnArgIle LeuTyrLeuGlnGluIlePheSerThrGluMetAlaThrLeuAspAspAlaPhePheGln ArgLouAspAspSerPhcLcuValProAspAspLysArgAspSerLysTyrProIlcPhe GlyAsnLeuValGluGluLysValTyrHisAspGluPheProThrIleTyrHisLeuArg LysTyrLeuAlaAspSerThrLysLysAlaAspLeuArgLeuValTyrLeuAlaLeuAla HisMetIleLysTyrArgGlyHisPheLeuIleGluGlyGluPheAsnSerLysAsnAsn AspIleGlnLysAsnPheGlnAspPheLeuAspThrTyrAsnAlaIlePheGluSerAsp LeuSerLeuGluAsnSerLysGlnLeuGluGluIleValLysAspLysIleSerLysLeu GluLysLysAspArgIleLeuLysLeuPheProGlyGluLysAsnSerGlyIlePheSer GluPheLeuLysLeuIleValGlyAsnGlnAlaAspPheArgLysCysPheAsnLeuAsp GluLysAlaSerLeuHisPheSerLysGluSerTyrAspGluAspLeuGluThrLeuLeu GlyTyrIleGlyAspAspTyrSerAspValPheLeuLysAlaLysLysLeuTyrAspAla IlcLeuLeuSerGlyPheLcuThrValThrAspAsnGluThrGluAlaProLcuScrSer AlaMetIleLysArgTyrAsnGluHisLysGluAspLeuAlaLeuLeuLysGluTyrIle ArgAsnIleSerLeuLysThrTyrAsnGluValPheLysAspAspThrLysAsnGlyTyr AlaGlyTyrIleAspGlyLysThrAsnGlnGluAspPheTyrValTyrLeuLysAsnLeu LeuAlaGluPheGluGlyAlaAspTyrPheLeuGluLysIleAspArgGluAspPheLeu ArgLysGlnArgThrPheAspAsnGlySerIleProTyrGlnIleHisLeuGlnGluMet ArgAlaIleLeuAspLysGlnAlaLysPheTyrProPheLeuAlaLysAsnLysGluArg IleGluLysIleLeuThrPheArgIleProTyrTyrValGlyProLeuAlaArgGlyAsn SerAspPheAlaTrpSerIleArgLysArgAsnGluLysIleThrProTrpAsnPheGlu AspValIleAspLysGluSerSerAlaGluAlaPheIleAsnArgMetThrSerPheAsp LeuTyrLeuProGluGluLysValLeuProLysHisSerLeuLeuTyrGluThrPheAsn ValTyrAsnGluLeuThrLysValArgPheIleAlaGluSerMetArgAspTyrGlnPhe LeuAspSerLysGlnLysLysAspIleValArgLeuTyrPheLysAspLysArgLysVal ThrAspLysAspIleIleGluTyrLeuHisAlaIleTyrGlyTyrAspGlyIleGluLeu LysGlyIleGluLysGlnPheAsnSerSerLeuSerThrTyrHisAspLeuLeuAsnIle IleAsnAspLysGluPheLeuAspAspSerSerAsnGluAlaIleIleGluGluIleIle HisThrLeuThrIlePheGluAspArgGluMetIleLysGlnArgLeuSerLysPheGlu AsnIlePheAspLysSerValLeuLysLysLeuSerArgArgHisTyrThrGlyTrpGly LysLeuSerAlaLysLeuIleAsnGlyIleArgAspGluLysSerGlyAsnThrIleLeu AspTyrLeuIleAspAspGlyIleSerAsnArgAsnPheMetGlnLeuIleHisAspAsp AlaLeuSerPheLysLysLysIleGlnLysAlaGlnIleIleGlyAspGluAspLysGly AsnIlcLysGluValValLysScrLcuProGlySerProAlaIleLysLysGlyIlcLeu GlnSerIleLysIleValAspGluLeuValLysValMetGlyGlyArgLysProGluSer IleValValGluMetAlaArgGluAsnGlnTyrThrAsnGlnGlyLysSerAsnSerGln GlnArgLeuLysArgLeuGluLysSerLeuLysGluLeuGlySerLysIleLeuLysGlu AsnIleProAlaLysLeuSerLysIleAspAsnAsnAlaLeuGlnAsnAspArgLeuTyr LeuTyrTyrLeuGlnAsnGlyLysAspMetTyrThrGlyAspAspLcuAspIlcAspArg LeuSerAsnTyrAspIleAspHisIleIleProGlnAlaPheLeuLysAspAsnSerIle AspAsnLysValLeuValSerSerAlaSerAsnArgGlyLysSerAspAspPheProSer LeuGluValValLysLysArgLysThrPheTrpTyrGlnLeuLeuLysSerLysLeuIle SerGlnArgLysPheAspAsnLeuThrLysAlaGluArgGlyGlyLeuLeuProGluAsp LysAlaGlyPheIleGlnArgGlnLeuValGluThrArgGlnIleThrLysHisValAla ArgLeuLeuAspGluLysPheAsnAsnLysLysAspGluAsnAsnArgAlaValArgThr ValLysIleIleThrLeuLysSerThrLeuValSerGlnPheArgLysAspPheGluLeu TyrLysValArgGluIleAsnAspPheHisHisAlaHisAspAlaTyrLeuAsnAla Val IleAlaSerAlaLeuLeuLysLysTyrProLysLeuGluProGluPheValTyrGlyAsp TyrProLysTyrAsnScrPhcArgGluArgLysScrAlaThrGluLysValTyrPhcTyr SerAsnIleMetAsnIlePheLysLysSerIleSerLeuAlaAspGlyArgValIleGlu ArgProLeuIleGluValAsnGluGluThrGlyGluSerValTrpAsnLysGluSerAsp LeuAlaThrValArgArgValLeuSerTyrProGlnValAsn ValValLysLysValGlu GluGlnAsnHisGlyLeuAspArgGlyLysProLysGlyLeuPheAsnAlaAsnLeuSer SerLysProLysProAsnSerAsnGluAsnLeuValGlyAlaLysGluTyrLeuAspPro LysLysTyrGlyGlyTyrAlaGlyIleSerAsnSerPheAlaValLeuValLysGlyThr IleGluLysGlyAlaLysLysLysIleThrAsnValLeuGluPheGlnGlyIleSerIle LeuAspArgIleAsnTyrArgLysAspLysLeuAsnPheLeuLeuGluLysGlyTyrLys AspIleGluLeuIleIleGluLeuProLysTyrSerLeuPheGluLeuSerAspGlySer ArgArgMetLeuAlaSerIleLeuSerThrAsnAsnLysArgGlyGluIleHisLysGly AsnGlnIlePheLeuSerGinLysPheValLysLeuLeuTyrHisAlaLysArgIleSer AsnThrIleAsnGluAsnHisArgLysTyrValGluAsnHisLysLysGluPheGluGlu LeuPheTyrTyrIleLeuGluPheAsnGluAsnTyrValGlyAlaLysLysAsnGlyLys LeuLeuAsnSerAlaPheGlnSerTrpGlnAsnHisSerIleAspGluLeuCysSerSer PheIleGlyProThrGlySerGluArgLysGlyLeuPheGluLeuThrSerArgGlySer AlaAlaAspPheGluPheLeuGly ValLysIleProArgTyrArgAspTyrThrProSer SerLeuLeuLysAspAlaThrLeuIleHisGlnSerValThrGlyLeuTyrGluThrArg IleAspLeuAlaLysLeuGlyGluGly SEQ ID NO: 2 D31A mutant One letter: mlfnkciiisinldfsnkckcmtkpysiglaigtnsvgwavitdnykvpskkmkvlgntskkyikknllgvllfdsgitaegrrlkrtarrrytrrr nrilylqeifstematlddaffqrlddsflvpddkrdskypifgnlveekvyhdefptiyhlrkyladstkkadlrlvylalahmikyrghfliegef nsknndiqknfqdfldtynaifesdlslenskqleeivkdkisklekkdrilklfpgeknsgifseflklivgnqadfrkcfnldekaslhfskesy dedletllgyigddysdvflkakklydaillsgfltvtdneteaplssamikrynehkedlallkeyirnislktynevfkddtkngyagyidgktn qedfyvylknllaefegadyflekidredflrkqrtfdngsipyqihlqemraildkqakfypflaknkeriekiltfripyyvgplargnsdfaws irkrnekitpwnfedvidkessaeafinrmtsfdlylpeekvlpkhsllyetfnvyneltkvrfiaesmrdyqfldskqkkdivrlyfkdkrkvtd kdiieylhaiygydgielkgiekqfnsslstyhdllniindkeflddssneaiieeiihtltifedremikqrlskfenifdksvlkklsrrhytgwgkl saklingirdeksgntildyliddgisnrfmqlihddalsfkkkiqkaqiigdedkgnikevvkslpgspaikkgilqsikivdelvkvmggrk pesivvemarenqytnqgksnsqqrlkrlekslkelgskilkenipaklskidnnalqndrlylyylqngkdmytgddldidrlsnydidhiip qaflkdnsidnkvlvssasnrgksddfpslevvkkrktfwyqllksklisqrkfdnltkaerggllpedkagfiqrqlvetrqitkhvarlldekfn nkkdennravrtvkiitlkstlvsqfrkdfclykvrcindfhhahdaylnaviasallkkypklcpcfvygdypkynsfrerksatckvyfysni mnifkksisladgrvierplievneetgesvwnkesdlatvrrvlsypqvnvvkkveeqnhgldrgkpkglfnanlsskpkpnsnenlvgak eyldpkkyggyagisnsfavlvkgtiekgakkkitnvlefqgisildrinyrkdklnfIlekgykdieliielpkyslfelsdgsrrmlasilstnnkr geihkgnqiflsqkfvkllyhakrisntinenhrkyvenhkkefeelfyyilefnenyvgakkngkllnsafqswqnhsidelcssfigptgser kglfeltsrgsaadfeflgvkipryrdytpssllkdatlihqsvtglyetridlaklgeg Three letters: MetLeuPheAsnLysCysIleIleIleSerIleAsnLeuAspPheSerAsnLysGluLys CysMetThrLysProTyrSerIleGlyLeuAlaIleGlyThrAsnSerValGlyTrpAla ValIleThrAspAsnTyrLysValProSerLysLysMetLysValLeuGlyAsnThrSer LysLysTyrIleLysLysAsnLeuLeuGlyValLeuLeuPheAspSerGlyIleThrAla GluGlyArgArgLeuLysArgThrAlaArgArgArgTyrThrArgArgArgAsnArgIle LeuTyrLeuGlnGluIlePheSerThrGluMetAlaThrLeuAspAspAlaPhePheGln ArgLeuAspAspSerPheLeuValProAspAspLysArgAspSerLysTyrProIlePhe GlyAsnLeuValGluGluLysValTyrHisAspGluPheProThrIleTyrHisLeuArg LysTyrLeuAlaAspSerThrLysLysAlaAspLeuArgLeuValTyrLeuAlaLeuAla HisMetIleLysTyrArgGlyHisPheLeuIleGluGlyGluPheAsnSerLysAsnAsn AspIleGlnLysAsnPheGlnAspPheLeuAspThrTyrAsnAlaIlePheGluSerAsp LeuSerLeuGluAsnSerLysGlnLeuGluGluIleValLysAspLysIleSerLysLeu GluLysLysAspArgIleLeuLysLeuPheProGlyGluLysAsnSerGlyIlePheSer GluPheLeuLysLeuIleValGlyAsnGlnAlaAspPheArgLysCysPheAsnLeuAsp GluLysAlaScrLeuHisPhcScrLysGluSerTyrAspGluAspLcuGluThrLcuLcu GlyTyrIleGlyAspAspTyrSerAspValPheLeuLysAlaLysLysLeuTyrAspAla IleLeuLeuSerGlyPheLeuThrValThrAspAsnGluThrGluAlaProLeuSerSer AlaMetIleLysArgTyrAsnGluHisLysGluAspLeuAlaLeuLeuLysGluTyrIle ArgAsnIleSerLeuLysThrTyrAsnGluValPheLysAspAspThrLysAsnGlyTyr AlaGlyTyrIlcAspGlyLysThrAsnGlnGluAspPhcTyrValTyrLcuLysAsnLcu LeuAlaGluPheGluGlyAlaAspTyrPheLeuGluLysIleAspArgGluAspPheLeu ArgLysGlnArgThrPheAspAsnGlySerIleProTyrGlnIleHisLeuGlnGluMet ArgAlaIleLeuAspLysGlnAlaLysPheTyrProPheLeuAlaLysAsnLysGluArg IleGluLysIleLeuThrPheArgIleProTyrTyrValGlyProLeuAlaArgGlyAsn SerAspPheAlaTrpSerIleArgLysArgAsnGluLysIleThrProTrpAsnPheGlu AspValIleAspLysGluSerSerAlaGluAlaPheIleAsnArgMetThrSerPheAsp LeuTyrLeuProGluGluLysValLeuProLysHisSerLeuLeuTyrGluThrPheAsn ValTyrAsnGluLeuThrLysValArgPheIleAlaGluSerMetArgAspTyrGlnPhe LeuAspSerLysGlnLysLysAspIleValArgLeuTyrPheLysAspLysArgLysVal ThrAspLysAspIlcIlcGluTyrLcuHisAlaIlcTyrGlyTyrAspGlyIlcGluLcu LysGlyIleGluLysGlnPheAsnSerSerLeuSerThrTyrHisAspLeuLeuAsnIle IleAsnAspLysGluPheLeuAspAspSerSerAsnGluAlaIleIleGluGluIleIle HisThrLeuThrIlePheGluAspArgGluMetIleLysGlnArgLeuSerLysPheGlu AsnIlePheAspLysSerValLeuLysLysLeuSerArgArgHisTyrThrGlyTrpGly LysLeuSerAlaLysLeuIleAsnGlyIleArgAspGluLysSerGlyAsnThrIleLeu AspTyrLeuIleAspAspGlyIleSerAsnArgAsnPheMetGinLeuIleHisAspAsp AlaLeuSerPheLysLysLysIleGlnLysAlaGlnIleIleGlyAspGluAspLysGly AsnIleLysGluValValLysSerLeuProGlySerProAlaIleLysLysGlyIleLeu GlnSerIleLysIleValAspGluLeuValLysValMetGlyGlyArgLysProGluSer IleValValGluMetAlaArgGluAsnGlnTyrThrAsnGlnGlyLysSerAsnSerGln GlnArgLeuLysArgLeuGluLysSerLeuLysGluLeuGlySerLysIleLeuLysGlu AsnIleProAlaLysLeuSerLysIleAspAsnAsnAlaLeuGlnAsnAspArgLeuTyr LeuTyrTyrLeuGlnAsnGlyLysAspMetTyrThrGlyAspAspLeuAspIleAspArg LeuSerAsnTyrAspIleAspHisIleIleProGlnAlaPheLeuLysAspAsnSerIle AspAsnLys ValLeuValSerSerAlaSerAsnArgGlyLysSerAspAspPheProSer LeuGluValValLysLysArgLysThrPheTrpTyrGlnLeuLeuLysSerLysLeuIle SerGlnArgLysPheAspAsnLeuThrLysAlaGluArgGlyGlyLeuLeuProGluAsp LysAlaGlyPheIleGlnArgGlnLeuValGluThrArgGlnIleThrLysHisValAla ArgLeuLeuAspGluLysPheAsnAsnLysLysAspGluAsnAsnArgAlaValArgThr ValLysIleIleThrLeuLysSerThrLeuValSerGlnPheArgLysAspPheGluLeu TyrLysValArgGlullcAsnAspPhcHisHisAlaHisAspAlaTyrLcuAsnAlaVal IleAlaSerAlaLeuLeuLysLysTyrProLysLeuGluProGluPheValTyrGlyAsp TyrProLysTyrAsnSerPheArgGluArgLysSerAlaThrGluLysValTyrPheTyr SerAsnIleMetAsnIlePheLysLysSerIleSerLeuAlaAspGlyArgValIleGlu ArgProLeuIleGluValAsnGluGluThrGlyGluSerValTrpAsnLysGluSerAsp LeuAlaThrValArgArgValLcuScrTyrProGlnValAsnValValLysLysValGlu GluGlnAsnHisGlyLeuAspArgGlyLysProLysGlyLeuPheAsnAlaAsnLeuSer SerLysProLysProAsnSerAsnGluAsnLeuValGlyAlaLysGluTyrLeuAspPro LysLysTyrGlyGlyTyrAlaGlyIleSerAsnSerPheAlaValLeuValLysGlyThr IleGluLysGlyAlaLysLysLysIleThrAsnValLeuGluPheGlnGlyIleSerIle LeuAspArgIleAsnTyrArgLysAspLysLeuAsnPheLeuLeuGluLysGlyTyrLys AspIleGluLeuIleIleGluLeuProLysTyrSerLeuPheGluLeuSerAspGlySer ArgArgMetLeuAlaSerIleLeuSerThrAsnAsnLysArgGlyGluIleHisLysGly AsnGlnIlePheLeuSerGlnLysPheValLysLeuLeuTyrHisAlaLysArgIleSer AsnThrIleAsnGluAsnHisArgLysTyrValGluAsnHisLysLysGluPheGluGlu LeuPhcTyrTyrIlcLeuGluPheAsnGluAsnTyrValGlyAlaLysLysAsnGlyLys LeuLeuAsnSerAlaPheGlnSerTrpGlnAsnHisSerIleAspGluLeuCysSerSer PheIleGlyProThrGlySerGluArgLysGlyLeuPheGluLeuThrSerArgGlySer AlaAlaAspPheGluPheLeuGlyValLysIleProArgTyrArgAspTyrThrProSer SerLeuLeuLysAspAlaThrLeuIleHisGlnSerValThrGlyLeuTyrGluThrArg IleAspLeuAlaLysLeuGlyGluGly SEQ ID NO: 3 N891A mutant One letter: mlfnkciiisinldfsnkekcmtkpysigldigtnsvgwavitdnykvpskkmkvlgntskkyikknllgvllfdsgitaegrrlkrtarrrytrrr nrilylqeifstematlddaffqrlddsflvpddkrdskypifgnlveekvyhdefptiyhlrkyladstkkadlrlvylalahmikyrghfliegef nsknndiqknfqdfldtynaifesdlslenskqleeivkdkisklekkdrilklfpgeknsgifseflklivgnqadfrkcfnldekaslhfskesy dedletllgyigddysdvflkakklydaillsgfltvtdneteaplssamikrynehkedlallkeyirnislktynevfkddtkngyagyidgktn qedfyvylknllaefegadyflekidredflrkqrtfdngsipyqihlqemraildkqakfypflaknkeriekiltfripyyvgplargnsdfaws irkrnekitpwnfedvidkessaeafinrmtsfdlylpeekvlpkhsllyetfnvyneltkvrfiaesmrdyqfldskqkkdivrlyfkdkrkvtd kdiieylhaiygydgielkgiekqfnsslstyhdllniindkeflddssneaiieeiihtltifedremikqrlskfenifdksvlkklsrrhytgwgkl saklingirdeksgntildyliddgisnrnfmqlihddalsfkkkiqkaqiigdedkgnikevvkslpgspaikkgilqsikivdelvkvmggrk pcsivvcmarenqytnqgksnsqqrlkrlekslkelgskilkenipaklskidnnalqndrlylyylqngkdmytgddldidrlsnydidhiip qaflkdnsidnkvlvssasargksddfpslevvkkrktfwyqllksklisqrkfdnltkaerggllpedkagfiqrqlvetrqitkhvarlldekfn nkkdennravrtvkiitlkstlvsqfrkdfelykvreindfhhahdaylnaviasallkkypklepefvygdypkynsfrerksatekvyfysni mnifkksisladgrvierplievneetgesvwnkesdlatvrrvlsypqvnvvkkveeqnhgldrgkpkglfnanlsskpkpnsnenlvgak eyldpkkyggyagisnsfavlvkgtiekgakkkitnvlefqgisildrinyrkdklnfIlekgykdieliielpkyslfelsdgsrrmlasilstnnkr kglfeltsrgsaadfeflgvkipryrdytpssllkdatlihqsvtglyetridlaklgeg Three letters: MetLeuPheAsnLysCysIleIleIleSerIleAsnLeuAspPheSerAsnLysGluLys CysMetThrLysProTyrSerIleGlyLeuAspIleGlyThrAsnSerValGlyTrpAla ValIleThrAspAsnTyrLysValProSerLysLysMetLysValLeuGlyAsnThrSer LysLysTyrIleLysLysAsnLeuLeuGlyValLeuLeuPheAspSerGlyIleThrAla GluGlyArgArgLeuLysArgThrAlaArgArgArgTyrThrArgArgArgAsnArgIle LeuTyrLeuGlnGluIlePheSerThrGluMetAlaThrLeuAspAspAlaPhePheGln ArgLcuAspAspScrPheLcuValProAspAspLysArgAspScrLysTyrProIlcPhe GlyAsnLeuValGluGluLysValTyrHisAspGluPheProThrIleTyrHisLeuArg LysTyrLeuAlaAspSerThrLysLysAlaAspLeuArgLeuValTyrLeuAlaLeuAla HisMetIleLysTyrArgGlyHisPheLeuIleGluGlyGluPheAsnSerLysAsnAsn AspIleGlnLysAsnPheGlnAspPheLeuAspThrTyrAsnAlaIlePheGluSerAsp LeuSerLeuGluAsnSerLysGlnLeuGluGluIleValLysAspLysIleSerLysLeu GluLysLysAspArgIleLeuLysLeuPheProGlyGluLysAsnSerGlyIlePheSer GluPheLeuLysLeuIleValGlyAsnGlnAlaAspPheArgLysCysPheAsnLeuAsp GluLysAlaSerLeuHisPheSerLysGluSerTyrAspGluAspLeuGluThrLeuLeu GlyTyrIleGlyAspAspTyrSerAspValPheLeuLysAlaLysLysLeuTyrAspAla IleLeuLeuSerGlyPheLeuThrValThrAspAsnGluThrGluAlaProLeuSerSer AlaMetIleLysArgTyrAsnGluHisLysGluAspLeuAlaLeuLeuLysGluTyrIle ArgAsnIleSerLeuLysThrTyrAsnGluValPheLysAspAspThrLysAsnGlyTyr AlaGlyTyrIleAspGlyLysThrAsnGlnGluAspPheTyrValTyrLeuLysAsnLeu LeuAlaGluPheGluGlyAlaAspTyrPheLeuGluLysIleAspArgGluAspPheLeu ArgLysGlnArgThrPheAspAsnGlySerIleProTyrGlnIleHisLeuGlnGluMet ArgAlaIleLeuAspLysGlnAlaLysPheTyrProPheLeuAlaLysAsnLysGluArg IleGluLysIleLeuThrPheArgIleProTyrTyrValGlyProLeuAlaArgGlyAsn SerAspPheAlaTrpSerIleArgLysArgAsnGluLysIleThrProTrpAsnPheGlu AspValIleAspLysGluSerSerAlaGluAlaPheIleAsnArgMetThrSerPheAsp LeuTyrLeuProGluGluLysValLeuProLysHisSerLeuLeuTyrGluThrPheAsn ValTyrAsnGluLcuThrLysValArgPhcIlcAlaGluScrMctArgAspTyrGlnPhc LeuAspSerLysGlnLysLysAspIleValArgLeuTyrPheLysAspLysArgLysVal ThrAspLysAspIleIleGluTyrLeuHisAlaIleTyrGlyTyrAspGlyIleGluLeu LysGlyIleGluLysGlnPheAsnSerSerLeuSerThrTyrHisAspLeuLeuAsnIle IleAsnAspLysGluPheLeuAspAspSerSerAsnGluAlaIleIleGluGluIleIle HisThrLcuThrIlcPhcGluAspArgGluMctIlcLysGlnArgLcuScrLysPhcGlu AsnIlePheAspLysSerValLeuLysLysLeuSerArgArgHisTyrThrGlyTrpGly LysLeuSerAlaLysLeuIleAsnGlyIleArgAspGluLysSerGlyAsnThrIleLeu AspTyrLeuIleAspAspGlyIleSerAsnArgAsnPheMetGlnLeuIleHisAspAsp AlaLeuSerPheLysLysLysIleGlnLysAlaGlnIleIleGlyAspGluAspLysGly AsnIleLysGluValValLysSerLeuProGlySerProAlaIleLysLysGlyIleLeu GlnSerIleLysIleValAspGluLeuValLysValMetGlyGlyArgLysProGluSer IleValValGluMetAlaArgGluAsnGlnTyrThrAsnGlnGlyLysSerAsnSerGln GlnArgLeuLysArgLeuGluLysSerLeuLysGluLeuGlySerLysIleLeuLysGlu AsnIleProAlaLysLeuSerLysIleAspAsnAsnAlaLeuGlnAsnAspArgLeuTyr LeuTyrTyrLeuGlnAsnGlyLysAspMctTyrThrGlyAspAspLeuAspIlcAspArg LeuSerAsnTyrAspIleAspHisIleIleProGInAlaPheLeuLysAspAsnSerIle AspAsnLysValLeuValSerSerAlaSerAlaArgGlyLysSerAspAspPheProSer LeuGhiValValLysLysArgLysThrPheTrpTyrGlnLeuLeuLysSerLysLeuIle SerGlnArgLysPheAspAsnLeuThrLysAlaGluArgGlyGlyLeuLeuProGluAsp LysAlaGlyPheIleGlnArgGlnLeuValGluThrArgGlnIleThrLysHisValAla ArgLeuLeuAspGluLysPheAsnAsnLysLysAspGluAsnAsnArgAlaValArgThr ValLysIleIleThrLeuLysSerThrLeuValSerGlnPheArgLysAspPheGluLeu TyrLysValArgGluIleAsnAspPheHisHisAlaHisAspAlaTyrLeuAsnAla Val IleAlaSerAlaLeuLeuLysLysTyrProLysLeuGluProGluPheValTyrGlyAsp TyrProLysTyrAsnSerPheArgGluArgLysSerAlaThrGluLysValTyrPheTyr SerAsnIleMetAsnIlePheLysLysSerIleSerLeuAlaAspGlyArgValIleGlu ArgProLeuIleGluValAsnGluGluThrGlyGluSerValTrpAsnLysGluSerAsp LeuAlaThrValArgArgValLeuSerTyrProGlnValAsnValValLysLysValGlu GluGlnAsnHisGlyLeuAspArgGlyLysProLysGlyLeuPheAsnAlaAsnLeuSer SerLysProLysProAsnSerAsnGluAsnLeuValGlyAlaLysGluTyrLeuAspPro LysLysTyrGlyGlyTyrAlaGlyIleSerAsnSerPheAlaValLeuValLysGlyThr IleGluLysGlyAlaLysLysLysIleThrAsnValLeuGluPheGlnGlyIleSerIle LeuAspArgIleAsnTyrArgLysAspLysLeuAsnPheLeuLeuGluLysGlyTyrLys AspIleGluLeuIleIleGluLeuProLysTyrSerLeuPheGluLeuSerAspGlySer ArgArgMetLeuAlaSerIleLeuSerThrAsnAsnLysArgGlyGluIleHisLysGly AsnGlnIlcPhcLeuSerGlnLysPhcValLysLeuLcuTyrHisAlaLysArgIleSer AsnThrIleAsnGluAsnHisArgLysTyrValGluAsnHisLysLysGluPheGluGlu LeuPheTyrTyrIleLeuGluPheAsnGluAsnTyrValGlyAlaLysLysAsnGlyLys LeuLeuAsnSerAlaPheGlnSerTrpGlnAsnHisSerIleAspGluLeuCysSerSer PheIleGlyProThrGlySerGluArgLysGlyLeuPheGluLeuThrSerArgGlySer AlaAlaAspPheGluPheLcuGlyValLysIlcProArgTyrArgAspTyrThrProSer SerLeuLeuLysAspAlaThrLeuIleHisGlnSerValThrGlyLeuTyrGluThrArg IleAspLeuAlaLysLeuGlyGluGly SEQ ID NO: 4 H868A mutant One letter mlfnkciiisinldfsnkckcmtkpysigldigtnsvgwavitdnykvpskkmkvlgntskkyikknllgvllfdsgitacgrrlkrtarrrytrrr nrilylqeifstematlddaffqrlddsflvpddkrdskypifgnlveekvyhdefptiyhlrkyladstkkadlrlvylalahmikyrghfliegef nsknndiqknfqdfldtynaifesdlslenskqleeivkdkisklekkdrilklfpgeknsgifseflklivgnqadfrkcfnldekaslhfskesy dedletllgyigddysdvflkakklydaillsgfltvtdneteaplssamikrynehkedlallkeyirnislktynevfkddtkngyagyidgktn qedfyvylknllaefegadyflekidredflrkqrtfdngsipyqihlqemraildkqakfypflaknkeriekiltfripyyvgplargnsdfaws irkrnekitpwnfedvidkessaeafinrmtsfdlylpeekvlpkhsllyetfnvyneltkvrfiaesmrdyqfldskqkkdivrlyfkdkrkvtd kdiieylhaiygydgielkgiekqfnsslstyhdllniindkeflddssneaiieeiihtltifedremikqrlskfenifdksvlkklsrrhytgwgkl saklingirdeksgntildyliddgisnrnfmqlihddalsfkkkiqkaqiigdedkgnikevvkslpgspaikkgilqsikivdelvkvmggrk pesivvemarenqytnqgksnsqqrlkrlekslkelgskilkenipaklskidnnalqndrlylyylqngkdmytgddldidrlsnydidaiipq aflkdnsidnkvlvssasnrgksddfpslevvkkrktfwyqllksklisqrkfdnltkaerggllpedkagfiqrqlvetrqitkhvarlldekfnn kkdennravrtvkiitlkstlvsqfrkdfelykvreindfhhahdaylnaviasallkkypklepefvygdypkynsfrerksatekvyfysnim nifkksisladgrvierplievneetgesvwnkesdlatvrrvlsypqvnvvkkveeqnhgldrgkpkglfnanlsskpkpnsnenlvgakey ldpkkyggyagisnsfavlvkgtiekgakkkitnvlefqgisildrinyrkdklnfIlekgykdieliielpkyslfelsdgsrrmlasilstnnkrge ihkgnqiflsqkfvkllyhakrisntinenhrkyvenhkkefeelfyyilefnenyvgakkngkllnsafqswqnhsidelcssfigptgserkgl feltsrgsaadfeflgvkipryrdytpssllkdatlihqsvtglyetridlaklgeg Three letters: MetLeuPheAsnLysCysIleIleIleSerIleAsnLeuAspPheSerAsnLysGluLys CysMetThrLysProTyrSerIleGlyLeuAspIleGlyThrAsnSerValGlyTrpAla ValIleThrAspAsnTyrLysValProSerLysLysMetLysValLeuGlyAsnThrSer LysLysTyrIleLysLysAsnLeuLeuGlyValLeuLeuPheAspSerGlyIleThrAla GluGlyArgArgLcuLysArgThrAlaArgArgArgTyrThrArgArgArgAsnArgIlc LeuTyrLeuGlnGluIlePheSerThrGluMetAlaThrLeuAspAspAlaPhePheGln ArgLeuAspAspSerPheLeuValProAspAspLysArgAspSerLysTyrProIlePhe GlyAsnLeuValGluGluLysValTyrHisAspGluPheProThrIleTyrHisLeuArg LysTyrLeuAlaAspSerThrLysLysAlaAspLeuArgLeuValTyrLeuAlaLeuAla HisMetIleLysTyrArgGlyHisPhcLcuIlcGluGlyGluPheAsnSerLysAsnAsn AspIleGlnLysAsnPheGlnAspPheLeuAspThrTyrAsnAlaIlePheGluSerAsp LeuSerLeuGluAsnSerLysGlnLeuGluGluIleValLysAspLysIleSerLysLeu GluLysLysAspArgIleLeuLysLeuPheProGlyGluLysAsnSerGlyIlePheSer GluPheLeuLysLeuIleValGlyAsnGlnAlaAspPheArgLysCysPheAsnLeuAsp GluLysAlaSerLeuHisPheSerLysGluSerTyrAspGluAspLeuGluThrLeuLeu GlyTyrIleGlyAspAspTyrSerAspValPheLeuLysAlaLysLysLeuTyrAspAla IleLeuLeuSerGlyPheLeuThrValThrAspAsnGluThrGluAlaProLeuSerSer AlaMetIleLysArgTyrAsnGluHisLysGluAspLeuAlaLeuLeuLysGluTyrIle ArgAsnIleSerLeuLysThrTyrAsnGluValPheLysAspAspThrLysAsnGlyTyr AlaGlyTyrIlcAspGlyLysThrAsnGlnGluAspPhcTyrValTyrLcuLysAsnLcu LeuAlaGluPheGluGlyAlaAspTyrPheLeuGluLysIleAspArgGluAspPheLeu ArgLysGlnArgThrPheAspAsnGlySerIleProTyrGlnIleHisLeuGlnGluMet ArgAlaIleLeuAspLysGlnAlaLysPheTyrProPheLeuAlaLysAsnLysGluArg IleGluLysIleLeuThrPheArgIleProTyrTyrValGlyProLeuAlaArgGlyAsn SerAspPheAlaTrpSerIleArgLysArgAsnGluLysIleThrProTrpAsnPheGlu AspValIleAspLysGluSerSerAlaGluAlaPheIleAsnArgMetThrSerPheAsp LeuTyrLeuProGluGluLysValLeuProLysHisSerLeuLeuTyrGluThrPheAsn ValTyrAsnGluLeuThrLysValArgPheIleAlaGluSerMetArgAspTyrGlnPhe LeuAspSerLysGlnLysLysAspIleValArgLeuTyrPheLysAspLysArgLysVal ThrAspLysAspIleIleGluTyrLeuHisAlaIleTyrGlyTyrAspGlyIleGluLeu LysGlyIleGluLysGlnPheAsnSerSerLeuSerThrTyrHisAspLeuLeuAsnIle IleAsnAspLysGluPheLeuAspAspSerSerAsnGluAlaIleIleGluGluIleIle HisThrLeuThrIlePheGluAspArgGluMetIleLysGlnArgLeuSerLysPheGlu AsnIlePheAspLysSerValLeuLysLysLeuSerArgArgHisTyrThrGlyTrpGly LysLeuSerAlaLysLeuIleAsnGlyIleArgAspGluLysSerGlyAsnThrIleLeu AspTyrLeuIleAspAspGlyIleSerAsnArgAsnPheMetGlnLeuIleHisAspAsp AlaLeuSerPheLysLysLysIleGlnLysAlaGlnIleIleGlyAspGluAspLysGly AsnIleLysGluValValLysSerLeuProGlySerProAlaIleLysLysGlyIleLeu GlnSerIleLysIleValAspGluLeuValLysValMetGlyGlyArgLysProGluSer IleValValGluMetAlaArgGluAsnGlnTyrThrAsnGlnGlyLysSerAsnSerGln GlnArgLcuLysArgLcuGluLysScrLcuLysGluLcuGlySerLysIlcLeuLysGlu AsnIleProAlaLysLeuSerLysIleAspAsnAsnAlaLeuGlnAsnAspArgLeuTyr LeuTyrTyrLeuGlnAsnGlyLysAspMetTyrThrGlyAspAspLeuAspIleAspArg LeuSerAsnTyrAspIleAspAlaIleIleProGlnAlaPheLeuLysAspAsnSerIle AspAsnLysValLeuValSerSerAlaSerAsnArgGlyLysSerAspAspPheProSer LeuGluValValLysLysArgLysThrPheTrpTyrGlnLcuLcuLysSerLysLeuIle SerGlnArgLysPheAspAsnLeuThrLysAlaGluArgGlyGlyLeuLeuProGluAsp LysAlaGlyPheIleGlnArgGlnLeuValGluThrArgGlnIleThrLysHisValAla ArgLeuLeuAspGluLysPheAsnAsnLysLysAspGluAsnAsnArgAlaValArgThr ValLysIleIleThrLeuLysSerThrLeuValSerGlnPheArgLysAspPheGluLeu TyrLysValArgGluIleAsnAspPheHisHisAlaHisAspAlaTyrLeuAsnAlaVal IleAlaSerAlaLeuLeuLysLysTyrProLysLeuGluProGluPheValTyrGlyAsp TyrProLysTyrAsnSerPheArgGluArgLysSerAlaThrGluLysValTyrPheTyr SerAsnIleMetAsnIlePheLysLysSerIleSerLeuAlaAspGlyArgValIleGlu ArgProLeuIleGluValAsnGluGluThrGlyGluSerValTrpAsnLysGluSerAsp LeuAlaThrValArgArgValLcuSerTyrProGlnValAsnValValLysLysValGlu GluGlnAsnHisGlyLeuAspArgGlyLysProLysGlyLeuPheAsnAlaAsnLeuSer SerLysProLysProAsnSerAsnGluAsnLeuValGlyAlaLysGluTyrLeuAspPro LysLysTyrGlyGlyTyrAlaGlyIleSerAsnSerPheAlaValLeuValLysGlyThr IleGluLysGlyAlaLysLysLysIleThrAsnValLeuGluPheGlnGlyIleSerIle LeuAspArgIleAsnTyrArgLysAspLysLeuAsnPheLeuLeuGluLysGlyTyrLys AspIleGluLeuIleIleGluLeuProLysTyrSerLeuPheGluLeuSerAspGlySer ArgArgMetLeuAlaSerIleLeuSerThrAsnAsnLysArgGlyGluIleHisLysGly AsnGlnIlePheLeuSerGlnLysPheValLysLeuLeuTyrHisAlaLysArgIleSer AsnThrIleAsnGluAsnHisArgLysTyrValGluAsnHisLysLysGluPheGluGlu LeuPheTyrTyrIleLeuGluPheAsnGluAsnTyrValGlyAlaLysLysAsnGlyLys LeuLeuAsnSerAlaPheGlnSerTrpGlnAsnHisSerIleAspGluLeuCysSerSer PheIleGlyProThrGlySerGluArgLysGlyLeuPheGluLeuThrSerArgGlySer AlaAlaAspPheGluPheLeuGlyValLysIleProArgTyrArgAspTyrThrProSer SerLeuLeuLysAspAlaThrLeuIleHisGlnSerValThrGlyLeuTyrGluThrArg IleAspLeuAlaLysLeuGlyGluGly SEQ ID NO: 5 Tra-crRNA, Unmature (102 nt): uaauaauaauugugguuugaaaccauucgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaacuugaaaagguggcac cgauucgguguuuuu SEQ ID NO: 6 Mature 78 nt tracrRNA: gggcgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaacuugaaaagguggcaccgauucgguguuuuu Shorter variants: gggcgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaacuugaaaagguggcaccgauucggug (SEQ ID NO: 44) gggcgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaacuugaaaagguggcaccgauu (SEQ ID NO: 45) gggcgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaacuugaaaagguggcac (SEQ ID NO: 46) gggcgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaacuugaaaaggu (SEQ ID NO: 47) gggcgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaacuugaa (SEQ ID NO: 48) gggcgaaacaacacagcgaguuaaaauaaggcuuaguccguacucaac (SEQ ID NO: 49) SEQ ID NO: 7 42 nt crRNA from spacer 1: 5′-CGCUAAAGAGGAAGAGGACAGUUUUAGAGCUGUGUUGUUUCG-3′ SEQ ID NO: 8 150 nt pre-crRNA 5′- ggguagaaaagauauccuacgagguuuuagagcuguguuguuucgaaugguuccaaaacaaauucuaa acgcuaaagaggaagaggacaguuuuagagcuguguuguuucgaaugguuccaaaacuacugcuguau uagcuugguuguug-3′ SEQ ID NO: 9 crRNA1 5′- ggguagaaaagauauccuacgagguuuuagagcuguguuguuucgaaugguuccaaaacTGTCATGA TAATAATGGTTTCTTAGACGTCguuuuagagcuguguuguuucgaaugguuccaaaacuacugcug uauuagcuugguuguug-3′ SEQ ID NO: 10 crRNA2 5′- ggguagaaaagauauccuacgagguuuuagagcuguguuguuucgaaugguuccaaaacacgagccg gaagcataaagtgtaaagcctgguuuuagagcuguguuguuucgaaugguuccaaaacuacugcug uauuagcuugguuguug-3′ SEQ ID NO: 11 Anti-A phage CRISPR RNA 5′- ggguagaaaagauauccuacgagguuuuagagcuguguuguuucgaaugguuccaaaactcaaggga gaatagaggctctogttgcattguuuuagagcuguguuguuucgaaugguuccaaaacuacugcug uauuagcuugguuguug-3′ SEQ ID NO: 12 Anti E. coli CRISPR RNA 5′- ggguagaaaagauauccuacgagguuuuagagcuguguuguuucgaaugguuccaaaaccgggaggg aagctgcatgatgcgatgttatguuuuagagcuguguuguuucgaaugguuccaaaacuacugcug uauuagcuugguuguug-3′ SEQ ID NO: 13 crRNA-HS1 5′-GCUCCCGGGGCUCGAUGAAGGUUUUAGAGCUGUGUUGUUUCG-3′ SEQ ID NO: 14 crRNA-HS2 UGAAUCGUGAAAUCUGCUCAGUUUUAGAGCUGUGUUGUUUCG

The application contains a Sequence Listing which has been submitted in ASCII format and is hereby incorporated by reference in its entirety. The ASCII copy, created on Mar. 20, 2013, is named 078981_6_SL.txt and is 64.4 kilobytes in size.

The embodiments shown and described in the specification are only specific embodiments of inventors who are skilled in the art and are not limiting in any way. Therefore, various changes, modifications, or alterations to those embodiments may be made without departing from the spirit of the invention in the scope of the following claims. The references cited are expressly incorporated by reference herein in their entirety.

Claims

1-49. (canceled)

50. A method for site-specific modification of a target DNA molecule, the method comprising:

expressing a polynucleotide encoding a tracrRNA and an engineered crRNA to produce the tracrRNA and the engineered crRNA, wherein the engineered crRNA has a spacer sequence complementary to a nucleotide sequence of the target DNA molecule;
expressing a polynucleotide encoding a Cas9 protein heterologous to the tracrRNA and the engineered crRNA to produce the Cas9 protein; and
combining the tracrRNA and the engineered crRNA with the Cas9 protein to form a Cas9-crRNA complex;
wherein the Cas9-crRNA complex is reprogrammed to cleave the target DNA molecule, the Cas9-crRNA complex being heterologous to the target DNA molecule.

51. The method of claim 50, further comprising preparing the polynucleotide encoding the tracrRNA and the engineered crRNA by chemical synthesis or in-vitro transcription.

52. The method of claim 50, wherein the spacer sequence of the engineered crRNA comprises at least 20 nucleotides.

53. The method of claim 50, wherein the Cas9 protein comprises a mutation in a RuvC active site motif or a HNH active site motif of the Cas9 protein.

54. The method of claim 50, wherein the polynucleotide encoding the tracrRNA and the engineered crRNA is a plasmid DNA.

55. The method of claim 50, further comprising contacting the Cas9-crRNA complex with the target DNA molecule to cleave the target DNA molecule.

56. The method of claim 55, wherein the target DNA molecule is double stranded.

57. A method for site-specific modification of a target DNA molecule, the method comprising:

expressing a polynucleotide encoding an engineered crRNA to produce the engineered crRNA, wherein the engineered crRNA has a spacer sequence complementary to a nucleotide sequence of the target DNA molecule;
expressing a polynucleotide encoding a Cas9 protein that is heterologous to the engineered crRNA to produce the Cas9 protein; and
combining the engineered crRNA with the Cas9 protein and a tracrRNA to form a Cas9-crRNA complex, wherein the Cas9-crRNA complex is reprogrammed to cleave the target DNA molecule.

58. The method of claim 57, further comprising contacting the Cas9-crRNA complex with the target DNA molecule.

59. The method of claim 58, wherein the target DNA molecule is double stranded, and wherein the Cas9-crRNA complex modifies the target DNA molecule by site-specific double stranded cleavage of the target DNA molecule.

60. The method of claim 58, wherein the target DNA molecule includes a protospacer-adjacent motif.

61. The method of claim 57, wherein the Cas9 protein is a nickase.

62. The method of claim 57, wherein the Cas9-crRNA complex is formed in the absence of RNaseIII.

63. A method for site-specific modification of a target DNA molecule, the method comprising:

re-programming a sequence of a polynucleotide to encode an engineered crRNA capable of site-specifically binding to a nucleotide sequence of the target DNA molecule;
expressing the polynucleotide to produce the engineered crRNA; and
combining the engineered crRNA with a tracrRNA, and a Cas9 protein heterologous to the engineered crRNA to form a Cas9-crRNA complex, wherein the Cas9-crRNA complex has engineered specificity towards the target DNA molecule.

64. The method of claim 63, further comprising contacting the Cas9-crRNA complex with the target DNA molecule to cleave the target DNA molecule, wherein the Cas9-crRNA complex modifies the target DNA molecule by site-specific cleavage of the target DNA molecule.

65. The method of claim 64, wherein the target DNA molecule is double stranded and includes a protospacer-adjacent motif.

66. The method of claim 63, wherein the spacer sequence of the engineered crRNA comprises at least 20 nucleotides.

67. The method of claim 63, wherein the polynucleotide encoding the engineered crRNA is prepared by chemical synthesis or in-vitro transcription.

68. The method of claim 63, wherein the Cas9-crRNA complex is formed in the absence of RNaseIII.

Patent History
Publication number: 20230272394
Type: Application
Filed: Dec 13, 2022
Publication Date: Aug 31, 2023
Applicant: VILNIUS UNIVERSITY (Vilnius)
Inventors: Virginijus SIKSNYS (Vilnius), Giedrius GASIUNAS (Birzu r.), Tautvydas KARVELIS (Vilniaus r.), Arvydas LUBYS (Vilnius), Lolita ZALIAUSKIENE (Vilnius), Monika GASIUNIENE (Vilnius), Anja SMITH (Lafyette, CO)
Application Number: 18/065,006
Classifications
International Classification: C12N 15/113 (20060101); C12N 15/10 (20060101); C12Q 1/6811 (20060101); C12P 19/34 (20060101); C12N 9/22 (20060101); C12N 15/90 (20060101);